Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer Ther ; 10(3): 427-36, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21282354

ABSTRACT

Many types of cancer cells require a supply of fatty acids (FA) for growth and survival, and interrupting de novo FA synthesis in model systems causes potent anticancer effects. We hypothesized that, in addition to synthesis, cancer cells may obtain preformed, diet-derived FA by uptake from the bloodstream. This would require hydrolytic release of FA from triglyceride in circulating lipoprotein particles by the secreted enzyme lipoprotein lipase (LPL), and the expression of CD36, the channel for cellular FA uptake. We find that selected breast cancer and sarcoma cells express and secrete active LPL, and all express CD36. We further show that LPL, in the presence of triglyceride-rich lipoproteins, accelerates the growth of these cells. Providing LPL to prostate cancer cells, which express low levels of the enzyme, did not augment growth, but did prevent the cytotoxic effect of FA synthesis inhibition. Moreover, LPL knockdown inhibited HeLa cell growth. In contrast to the cell lines, immunohistochemical analysis confirmed the presence of LPL and CD36 in the majority of breast, liposarcoma, and prostate tumor tissues examined (n = 181). These findings suggest that, in addition to de novo lipogenesis, cancer cells can use LPL and CD36 to acquire FA from the circulation by lipolysis, and this can fuel their growth. Interfering with dietary fat intake, lipolysis, and/or FA uptake will be necessary to target the requirement of cancer cells for FA.


Subject(s)
Cell Proliferation , Dietary Fats/metabolism , Fatty Acids/metabolism , Lipoprotein Lipase/metabolism , Neoplasms/metabolism , Animals , CD36 Antigens/genetics , Cell Line, Tumor , Fatty Acid Synthesis Inhibitors/pharmacology , Fatty Acids/pharmacology , Female , Humans , Lipolysis , Liposarcoma/genetics , Liposarcoma/metabolism , Male , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering/genetics
2.
Int J Oncol ; 36(5): 1309-14, 2010 May.
Article in English | MEDLINE | ID: mdl-20372807

ABSTRACT

Liposarcomas (LS) are mesenchymal tumors that can recur after surgical resection and often do not respond to presently available medical therapies. This study demonstrates the dependence of LS on de novo long-chain fatty acid synthesis for growth. Lipogenesis can be impaired by inhibiting the activities of lipogenic enzymes, including acetyl CoA-carboxylase (ACC) and fatty acid synthase (FASN), or by suppressing the expression of key genes involved in the pathway and its regulation. The FASN inhibitors cerulenin and orlistat reduced the growth of two LS cell lines (LiSa2, SW872), as did inhibition of ACC with soraphen A. CDDO-Me, a synthetic triterpenoid, suppressed expression of Spot 14 and FASN genes and likewise inhibited LS cell growth. Importantly, the anti-proliferative effect of each agent was prevented by the co-administration of palmitate, the major product of cellular long-chain fatty acid synthesis. In stark contrast to LS cells, these compounds had no effect on the growth of fibroblasts. Four biochemically distinct agents that target critical points in the fatty acid synthetic pathway exert anti-proliferative effects on LS cells, and rescue of cell growth by palmitic acid suggests that reduced tumor cell lipogenesis mediates the growth inhibition. These findings warrant further studies aimed at the clinical exploitation of the dependence of LS cell growth on fatty acids.


Subject(s)
Fatty Acids/chemistry , Gene Expression Regulation, Neoplastic , Liposarcoma/metabolism , Acetyl-CoA Carboxylase/metabolism , Cell Line, Tumor , Cell Proliferation , Dose-Response Relationship, Drug , Fatty Acid Synthases/metabolism , Fibroblasts/metabolism , Humans , Lactones/pharmacology , Lipids/chemistry , Orlistat , Reverse Transcriptase Polymerase Chain Reaction
3.
Cardiovasc Res ; 85(1): 224-31, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19720605

ABSTRACT

AIMS: Given the importance of IgG Fc receptors in immune regulation, we hypothesized that Fcg receptor type III (FcgRIII, CD16) plays an important role in atherogenesis. We therefore analysed the formation of arterial lesions in LDL receptor-deficient (LDLR(-/-)) and FcgRIII(-/-)xLDLR(-/-) double knockout mice at three different points up to 24 weeks of exposure to a high-fat diet. METHODS AND RESULTS: Analysis of Oil Red-O-stained sections revealed no difference in lesion formation between strains after 6 weeks of a high-fat diet, and a modest decrease after 14 weeks in double knockouts relative to LDLR(-/-) controls. After 24 weeks, lesion formation was decreased in the aortic root (30%) and innominate artery (50%) in FcgRIII double knockouts relative to LDLR(-/-) controls. Analysis of peripheral CD4+ T-cells by intracellular flow cytometry from double knockouts after 24 weeks of a high-fat diet revealed statistically significant increases in the percentages of cells producing interferon-gamma, interleukin (IL)-10, and IL-4 relative to controls, differences that were also observed by analyses of whole aortas for cytokine mRNA levels. As determined by flow cytometry, FcgRIII deficiency resulted in an expansion of CD4+ cells and an increase in the CD4 to CD8 ratio. Analysis of plasma anti-oxidized LDL (OxLDL) antibodies by chemiluminescent assay revealed that IgG1 and IgG2c titers to OxLDL were increased in FcgRIII (-/-)xLDLR(-/-) double knockouts relative to LDLR(-/-) controls, while total IgG levels were similar. CONCLUSION: These results reveal altered immunity in FcgRIII(-/-)xLDLR(-/-) mice and a reduction in lesion formation associated with increased production of IL-10 by an expansion of CD4+ T-cells. The reduction in lesion formation was manifest well after evidence of an immune response to OxLDL, suggesting that FcgRIII contributes to lesion progression in murine atherosclerosis.


Subject(s)
Arteries/pathology , Interleukin-10/physiology , Receptors, IgG/physiology , Animals , Atherosclerosis/pathology , Autoantibodies/blood , CD4-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Female , Immunity , Interferon-gamma/physiology , Lipids/blood , Lipoproteins, LDL/immunology , Male , Mice , Receptors, LDL/deficiency , Receptors, LDL/physiology
4.
Breast Cancer Res Treat ; 98(2): 231-40, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16552628

ABSTRACT

Most breast cancers are "lipogenic", defined by high fatty acid synthase (FAS) content and dependence on fatty acid synthesis for growth and survival. S14 (Spot 14; THRSP) is a nuclear protein that activates genes required for fatty acid synthesis. The S14 gene is amplified in approximately 15% of breast cancers, but clinical correlates of its expression were unknown. We analyzed 131 breast cancers by immunohistochemistry for S14 and FAS. Staining was graded 0, 1, or 2+, and scores were correlated with traditional tumor markers, histological features, and outcome. S14 and FAS staining were related to tumor size (p=0.05 for S14, p=0.038 for FAS), but not to stage. S14 but not FAS scores correlated with tumor grade in both DCIS (p=0.003) and invasive cases (p<0.001). Invasive cases (pooled node - and +) with weak S14 staining (n=21) showed no recurrence over 3000 d follow-up, including 10 cases with lymph node involvement, whereas 32% of 67 strongly-staining tumors recurred (log rank p<0.0001). S14 scores did not cosegregate with sex steroid receptors, Her2/neu, or cyclin D1. Low level S14 expression is associated with prolonged disease-free survival in invasive cases, including those with nodal metastasis. High-level expression of S14 identifies a subset of high-risk breast cancers that is not specified by analysis of sex steroid receptors, Her2/neu, or cyclin D1, and provides a molecular correlate to histologic features that predict recurrence.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Nuclear Proteins/analysis , Transcription Factors/analysis , Adult , Aged , Aged, 80 and over , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cyclin D1/analysis , Dietary Fats/administration & dosage , Female , Humans , Immunohistochemistry , Middle Aged
5.
Exp Cell Res ; 312(3): 278-88, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16300755

ABSTRACT

Most breast cancers exhibit brisk lipogenesis, and require it for growth. S14 is a lipogenesis-related nuclear protein that is overexpressed in most breast cancers. Sterol response element-binding protein-1c (SREBP-1c) is required for induction of lipogenesis-related genes, including S14 and fatty acid synthase (FAS), in hepatocytes, and correlation of SREBP-1c and FAS expression suggested that SREBP-1c drives lipogenesis in tumors as well. We directly tested the hypothesis that SREBP-1c drives S14 expression and mediates lipogenic effects of progestin in T47D breast cancer cells. Dominant-negative SREBP-1c inhibited induction of S14 and FAS mRNAs by progestin, while active SREBP-1c induced without hormone and superinduced in its presence. Changes in S14 mRNA were reflected in protein levels. A lag time and lack of progestin response elements indicated that S14 and FAS gene activation by progestin is indirect. Knockdown of S14 reduced, whereas overexpression stimulated, T47D cell growth, while nonlipogenic MCF10a mammary epithelial cells were not growth-inhibited. These data directly demonstrate that SREBP-1c drives S14 gene expression in breast cancer cells, and progestin magnifies that effect via an indirect mechanism. This supports the prediction, based on S14 gene amplification and overexpression in breast tumors, that S14 augments breast cancer cell growth and survival.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation/drug effects , Nuclear Proteins/genetics , Progestins/pharmacology , Sterol Regulatory Element Binding Protein 1/pharmacology , Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Breast/metabolism , Breast Neoplasms/metabolism , Cell Proliferation , Cells, Cultured , Epithelial Cells/metabolism , Fatty Acid Synthases , Gene Amplification , Genes, Dominant , Humans , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Response Elements , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Transcriptional Activation , fas Receptor/genetics , fas Receptor/metabolism
6.
Lab Invest ; 83(11): 1569-81, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14615411

ABSTRACT

To test the possibility that acyl-coenzyme A:cholesterol acyltransferase 2 (ACAT2) may be expressed in human macrophages under pathologic conditions, we employed specific anti-ACAT2 antibodies and found clear ACAT2 signals in lipid-laden as well as lipid-free macrophages under various disease conditions, including atherosclerosis. However, no ACAT2 signal was detectable in macrophages under normal physiologic conditions. Using cultured human macrophages derived from blood-borne monocytes, immunoblot and RT-PCR analyses demonstrated that immature macrophages expressed only ACAT1, but the fully differentiated macrophages expressed both ACAT1 and ACAT2. Furthermore, RT-PCR clearly revealed the presence of both ACAT1 and ACAT2 mRNAs in human atherosclerotic aorta. Double immunohistochemical staining indicated that in human atherosclerotic aorta, all macrophages expressed ACAT1, while approximately 70% to 80% of macrophages also expressed ACAT2. In congenital hyperlipidemic mice, immunohistochemistry and RT-PCR demonstrated that ACAT2 was also present in lipid-laden cells of the atheromatous plaques. Our results suggest that in atherosclerotic plaque, the ability of macrophage foam cell transformation may be augmented by the dual expressions of ACAT1 and ACAT2. Additional immunoblot and RT-PCR experiments showed that the ACAT2 signal was clearly detectable in thioglycollate-elicited exudate mouse macrophages but not in peritoneal resident macrophages. We conclude that under various pathologic conditions, fully differentiated macrophages express ACAT2 in addition to ACAT1.


Subject(s)
Foam Cells/enzymology , Leukocytes, Mononuclear/cytology , Sterol O-Acyltransferase/biosynthesis , Animals , Aorta/enzymology , Aorta/pathology , Arteriosclerosis/enzymology , Arteriosclerosis/pathology , Cells, Cultured , Fluorescent Antibody Technique, Indirect , Foam Cells/cytology , Humans , Immunoenzyme Techniques , Mice , Mice, Knockout , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase 2
7.
J Leukoc Biol ; 72(4): 711-7, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12377940

ABSTRACT

CD163, a monocyte and macrophage-specific surface glycoprotein, which is increased by interleukin-10 and glucocorticoids, is a scavenger receptor for hemoglobin/haptoglobin complexes. We report a rapid and highly reproducible rise in soluble CD163 in the plasma of human volunteers given intravenous lipopolysaccharide (LPS). We also show that LPS induces shedding of CD163 from the surface of isolated monocytes, identifying shedding from monocytes and macrophages as a likely mechanism for the endotoxemia-associated rise in plasma CD163 in vivo. Studies using the inhibitor TAPI-0 indicate that a metalloproteinase is responsible for LPS-mediated shedding of CD163. Finally, we demonstrate a marked increase in surface CD163 expression on circulating monocytes 24 h following experimental endotoxemia. These findings show that CD163 is rapidly mobilized in response to bacterial endotoxin. As hemoglobin can bind LPS and enhance its toxicity, it will be important to determine how cell surface and soluble CD163 influence inflammatory processes during sepsis.


Subject(s)
Endotoxemia/blood , Membrane Glycoproteins/blood , Metalloendopeptidases/antagonists & inhibitors , Monocytes/immunology , Receptors, Immunologic/blood , Up-Regulation , Cell Membrane/immunology , Dipeptides/pharmacology , Endotoxemia/immunology , Hydroxamic Acids/pharmacology , Injections, Intravenous , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/adverse effects , Lipopolysaccharides/pharmacology , Monocytes/drug effects , Receptors, Scavenger , Tetradecanoylphorbol Acetate/pharmacology , Time Factors
8.
J Morphol ; 191(2): 177-191, 1987 Feb.
Article in English | MEDLINE | ID: mdl-29921104

ABSTRACT

The cardiac ganglion in the lobster Homarus americanus was examined with a transmission electron microscope. Nerve terminals often existed in large aggregations surrounded by glial and connective tissue elements. Axo-axonic and axo-dendritic synapses were present. Six ultrastructurally different types of nerve terminal, each containing an abundance of vesicles, were distinguished: three formed discrete chemical synapses as indicated by typical release site morphology; three did not. The latter appear to be neurosecretory axon terminals of extrinsic neurons. More than one morphologically distinct type of synaptic vesicle occurred commonly in a given terminal, suggesting the presence of coexisting neurotransmitters and/or neuroregulatory factors. Symmetrical chemical synapses and electrotonic junctions between axons were present.

SELECTION OF CITATIONS
SEARCH DETAIL
...