Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Lipid Res ; 61(2): 205-218, 2020 02.
Article in English | MEDLINE | ID: mdl-31806729

ABSTRACT

We previously described the expression of CD36 and LPL by breast cancer (BC) cells and tissues and the growth-promoting effect of VLDL observed only in the presence of LPL. We now report a model in which LPL is bound to a heparan sulfate proteoglycan motif on the BC cell surface and acts in concert with the VLDL receptor to internalize VLDLs via receptor-mediated endocytosis. We also demonstrate that gene-expression programs for lipid synthesis versus uptake respond robustly to triglyceride-rich lipoprotein availability. The literature emphasizes de novo FA synthesis and exogenous free FA uptake using CD36 as paramount mechanisms for lipid acquisition by cancer cells. We find that the uptake of intact lipoproteins is also an important mechanism for lipid acquisition and that the relative reliance on lipid synthesis versus uptake varies among BC cell lines and in response to VLDL availability. This metabolic plasticity has important implications for the development of therapies aimed at the lipid dependence of many types of cancer, in that the inhibition of FA synthesis may elicit compensatory upregulation of lipid uptake. Moreover, the mechanism that we have elucidated provides a direct connection between dietary fat and tumor biology.-.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Endocytosis , Lipid Droplets/metabolism , Lipoproteins, VLDL/metabolism , Humans , Tumor Cells, Cultured
2.
J Cell Physiol ; 231(10): 2128-41, 2016 10.
Article in English | MEDLINE | ID: mdl-26844415

ABSTRACT

Brisk fatty acid (FA) production by cancer cells is accommodated by the Warburg effect. Most breast and other cancer cell types are addicted to fatty acids (FA), which they require for membrane phospholipid synthesis, signaling purposes, and energy production. Expression of the enzymes required for FA synthesis is closely linked to each of the major classes of signaling molecules that stimulate BC cell proliferation. This review focuses on the regulation of FA synthesis in BC cells, and the impact of FA, or the lack thereof, on the tumor cell phenotype. Given growing awareness of the impact of dietary fat and obesity on BC biology, we will also examine the less-frequently considered notion that, in addition to de novo FA synthesis, the lipolytic uptake of preformed FA may also be an important mechanism of lipid acquisition. Indeed, it appears that cancer cells may exist at different points along a "lipogenic-lipolytic axis," and FA uptake could thwart attempts to exploit the strict requirement for FA focused solely on inhibition of de novo FA synthesis. Strategies for clinically targeting FA metabolism will be discussed, and the current status of the medicinal chemistry in this area will be assessed. J. Cell. Physiol. 231: 2128-2141, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Breast Neoplasms/metabolism , Cell Proliferation/physiology , Fatty Acids/metabolism , Lipid Metabolism/physiology , Lipogenesis/physiology , Animals , Fatty Acid Synthases/metabolism , Female , Humans
3.
Carcinogenesis ; 36 Suppl 1: S203-31, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26106140

ABSTRACT

Environmental contributions to cancer development are widely accepted, but only a fraction of all pertinent exposures have probably been identified. Traditional toxicological approaches to the problem have largely focused on the effects of individual agents at singular endpoints. As such, they have incompletely addressed both the pro-carcinogenic contributions of environmentally relevant low-dose chemical mixtures and the fact that exposures can influence multiple cancer-associated endpoints over varying timescales. Of these endpoints, dysregulated metabolism is one of the most common and recognizable features of cancer, but its specific roles in exposure-associated cancer development remain poorly understood. Most studies have focused on discrete aspects of cancer metabolism and have incompletely considered both its dynamic integrated nature and the complex controlling influences of substrate availability, external trophic signals and environmental conditions. Emerging high throughput approaches to environmental risk assessment also do not directly address the metabolic causes or consequences of changes in gene expression. As such, there is a compelling need to establish common or complementary frameworks for further exploration that experimentally and conceptually consider the gestalt of cancer metabolism and its causal relationships to both carcinogenesis and the development of other cancer hallmarks. A literature review to identify environmentally relevant exposures unambiguously linked to both cancer development and dysregulated metabolism suggests major gaps in our understanding of exposure-associated carcinogenesis and metabolic reprogramming. Although limited evidence exists to support primary causal roles for metabolism in carcinogenesis, the universality of altered cancer metabolism underscores its fundamental biological importance, and multiple pleiomorphic, even dichotomous, roles for metabolism in promoting, antagonizing or otherwise enabling the development and selection of cancer are suggested.


Subject(s)
Carcinogenesis/chemically induced , Carcinogenesis/metabolism , Carcinogens, Environmental/adverse effects , Environmental Exposure/adverse effects , Neoplasms/chemically induced , Neoplasms/metabolism , Animals , Humans , Neoplasms/etiology
4.
Fed Pract ; 32(Suppl 4): 27S-30S, 2015 May.
Article in English | MEDLINE | ID: mdl-30766119

ABSTRACT

Therapeutic vaccines promise new and potentially more effective treatment options for solid tumors and hematologic malignancies.

5.
Prog Lipid Res ; 52(4): 585-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24001676

ABSTRACT

One of the most important metabolic hallmarks of cancer cells is enhanced lipogenesis. Depending on the tumor type, tumor cells synthesize up to 95% of saturated and mono-unsaturated fatty acids (FA) de novo in spite of sufficient dietary lipid supply. This lipogenic conversion starts early when cells become cancerous and further expands as the tumor cells become more malignant. It is suggested that activation of FA synthesis is required for carcinogenesis and for tumor cell survival. These observations suggest that the enzymes involved in FA synthesis would be rational therapeutic targets for cancer treatment. However, several recent reports have shown that the anti-tumor effects, following inhibition of endogenous FA synthesis in cancer cell lines may be obviated by adding exogenous FAs. Additionally, high intake of dietary fat is reported to be a potential risk factor for development and poor prognosis for certain cancers. Recently it was reported that breast and liposarcoma tumors are equipped for both de novo fatty acid synthesis pathway as well as LPL-mediated extracellular lipolysis. These observations indicate that lipolytically acquired FAs may provide an additional source of FAs for cancer. This review focuses on our current understanding of lipogenic and lipolytic pathways in cancer cell progression.


Subject(s)
Lipogenesis , Lipolysis , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Carcinogenesis , Fatty Acid Synthases/metabolism , Fatty Acids/metabolism , Humans , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/prevention & control
6.
Breast Cancer Res Treat ; 138(1): 175-83, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23417336

ABSTRACT

Conjugated linoleic acid (CLA) is widely used as a "nutraceutical" for weight loss. CLA has anticancer effects in preclinical models, and we demonstrated in vitro that this can be attributed to the suppression of fatty acid (FA) synthesis. We tested the hypothesis that administration of CLA to breast cancer patients would inhibit expression of markers related to FA synthesis in tumor tissue, and that this would suppress tumor proliferation. Women with Stage I-III breast cancer were enrolled into an open label study and treated with CLA (1:1 mix of 9c,11t- and 10t,12c-CLA isomers, 7.5 g/d) for ≥ 10 days before surgery. Fasting plasma CLA concentrations measured pre- and post-CLA administration, and pre/post CLA tumor samples were examined by immunohistochemistry for Spot 14 (S14), a regulator of FA synthesis, FA synthase (FASN), an enzyme of FA synthesis, and lipoprotein lipase (LPL), the enzyme that allows FA uptake. Tumors were also analyzed for expression of Ki-67 and cleaved caspase 3. 24 women completed study treatment, and 23 tumors were evaluable for the primary endpoint. The median duration of CLA therapy was 12 days, and no significant toxicity was observed. S14 expression scores decreased (p = 0.003) after CLA administration. No significant change in FASN or LPL expression was observed. Ki-67 scores declined (p = 0.029), while cleaved caspase 3 staining was unaffected. Decrements in S14 or Ki-67 did not correlate with fasting plasma CLA concentrations at surgery. Breast tumor tissue expression of S14, but not FASN or LPL, was decreased after a short course of treatment with 7.5 g/day CLA. This was accompanied by reductions in the proliferation index. CLA consumption was well-tolerated and safe at this dose for up to 20 days. Overall, CLA may be a prototype compound to target fatty acid synthesis in breast cancers with a "lipogenic phenotype".


Subject(s)
Biosynthetic Pathways/drug effects , Breast Neoplasms/metabolism , Linoleic Acid/pharmacology , Lipid Metabolism/drug effects , Adult , Aged , Aged, 80 and over , Apoptosis , Biomarkers, Tumor , Breast Neoplasms/pathology , Cell Proliferation , Female , Humans , Linoleic Acid/administration & dosage , Linoleic Acid/adverse effects , Middle Aged , Neoplasm Grading
7.
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
8.
Atlas Genet Cytogenet Oncol Haematol ; 15(6): 480-482, 2011 Jun.
Article in English | MEDLINE | ID: mdl-24174994
9.
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
10.
Pediatr Nephrol ; 17(8): 585-90, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12185462

ABSTRACT

In experimental animals, the decreased growth during mild uremia is not accompanied by a loss in the capacity of the pituitary gland to secrete growth hormone (GH). With the development of orally administered GH "secretagogues" (GHS), it might be possible to stimulate growth during uremia without injections. This study was designed to determine the effects of the GHS, L-163,255. Uremia was induced by 5/6 nephrectomy (NX). GHS was given orally, 3 mg/kg, twice a week. Four groups of animals included: (1) sham-operated, (2) sham-operated, pair-fed, (3) uremic (NX), and (4) uremic, GHS-treated (NX+GHS). Blood sampling was conducted via intra-atrial catheters, and GH was quantitated. Pituitary GH mRNA was measured by Northern blot, and liver GH receptor and insulin-like growth factor-I mRNAs by RNAase protection. Untreated NX animals had a specific decrease in the "mass" of the GH pulses. A burst of GH was induced by GHS, but the pulsatile pattern of GH secretion over 6 h was not affected. An increase or a return to non-uremic levels of GH-related mRNAs occurred after GHS. Thus, GHS stimulated an acute burst of GH secretion and increased specific mRNAs encoding GH-related proteins in uremic animals.


Subject(s)
Growth Hormone/blood , Piperidines/pharmacology , RNA, Messenger/biosynthesis , Spiro Compounds/pharmacology , Uremia/metabolism , Animals , Blotting, Northern , Creatinine/blood , Growth Hormone/biosynthesis , Half-Life , Insulin-Like Growth Factor I/biosynthesis , Liver/drug effects , Liver/metabolism , Male , Nephrectomy , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Somatotropin/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...