Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-37291785

ABSTRACT

This article has been withdrawn from the journal Anti-Cancer Agents in Medicinal Chemistry due to a Conflict of Interest between the authors.Bentham Science apologizes to the readers of the journal for any inconvenience this may have caused.The Bentham Editorial Policy on Article Withdrawal can be found at https://benthamscience.com/editorialpoliciesmain. php. BENTHAM SCIENCE DISCLAIMER: It is a condition of publication that manuscripts submitted to this journal have not been published and will not be simultaneously submitted or published elsewhere. Furthermore, any data, illustration, structure or table that has been published elsewhere must be reported, and copyright permission for reproduction must be obtained. Plagiarism is strictly forbidden, and by submitting the article for publication the authors agree that the publishers have the legal right to take appropriate action against the authors, if plagiarism or fabricated information is discovered. By submitting a manuscript, the authors agree that the copyright of their article is transferred to the publishers if and when the article is accepted for publication.

2.
Lipids ; 55(4): 403-412, 2020 07.
Article in English | MEDLINE | ID: mdl-32424826

ABSTRACT

Lipids such as cholesterol, triacylglycerols, and fatty acids play important roles in the regulation of cellular metabolism and cellular signaling pathways and, as a consequence, in the development of various diseases. It is therefore important to understand how their metabolism is regulated to better define the components involved in the development of various human diseases. In the present work, we describe the development and validation of a high-performance thin layer chromatography (HPTLC) method allowing the separation and quantification of free cholesterol, cholesteryl esters, nonesterified fatty acids, and triacylglycerols. This method will be of interest as the quantification of these lipids in one single assay is difficult to perform.


Subject(s)
Breast/chemistry , Lipids/analysis , Tissue Extracts/chemistry , Breast/pathology , Cell Line, Tumor , Cholesterol/analysis , Cholesterol Esters/analysis , Chromatography, Thin Layer , Fatty Acids, Nonesterified/analysis , Humans , MCF-7 Cells , Triglycerides/analysis
3.
Breast Cancer Res ; 22(1): 38, 2020 04 22.
Article in English | MEDLINE | ID: mdl-32321558

ABSTRACT

BACKGROUND: The highest incidence of breast cancer is in the Western world. Several aspects of the Western lifestyle are known risk factors for breast cancer. In particular, previous studies have shown that cholesterol levels can play an important role in the regulation of tumor progression. METHODS: In the present study, we modulated cholesterol metabolism in the human breast cancer cell lines MCF-7 and MDA-MB-231 using a genetic approach. Apolipoprotein A-I (apoA-I) and apolipoprotein E (apoE) were expressed in these cell lines to modulate cholesterol metabolism. The effects of these apolipoproteins on cancer cell properties were examined. RESULTS: Our results show that both apolipoproteins can regulate cholesterol metabolism and can control the epithelial-to-mesenchymal transition process. However, these effects were different depending on the cell type. We show that expressing apoA-I or apoE stimulates proliferation, migration, and tumor growth of MCF-7 cells. However, apoA-I or apoE reduces proliferation and migration of MDA-MB-231 cells. CONCLUSIONS: These data suggest that modulating sterol metabolism may be most effective at limiting tumor progression in models of triple-negative cancers.


Subject(s)
Apolipoprotein A-I/metabolism , Apolipoproteins E/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Cholesterol/metabolism , Lipid Metabolism , Animals , Breast Neoplasms/classification , Cell Line, Tumor , Cell Movement , Databases, Genetic , Epithelial-Mesenchymal Transition , Female , Humans , Mice , Mice, Nude , Survival Rate , Xenograft Model Antitumor Assays
4.
Biomolecules ; 10(1)2019 12 28.
Article in English | MEDLINE | ID: mdl-31905626

ABSTRACT

Despite recent advances in our understanding of the biological processes leading to the development and progression of cancer, there is still a need for new and effective agents to treat this disease. Phytoprostanes (PhytoPs) and phytofurans (PhytoFs) are non-enzymatically oxidized products of α-linolenic acid that are present in seeds and vegetable oils. They have been shown to possess anti-inflammatory and apoptosis-promoting activities in macrophages and leukemia cells, respectively. In this work, seven PhytoPs (PP1-PP7) and one PhytoFs (PF1) were evaluated for their cytotoxic, chemosensitization, and anti-migratory activities using the MCF-7 and MDA-MB-231 breast cancer cell lines. Among the tested compounds, only three PhytoPs had a significant effect on cell viability compared to the control group: Ent-9-L1-PhytoP (PP6) decreased cell viability in both cell lines, while 16-F1t-PhytoP (PP1) and 9-L1-PhytoP (PP5) decreased viability of MCF-7 and MDA-MB-231 cells, respectively. When combined with a sub-cytotoxic dose of doxorubicin, these three PhytoPs displayed significantly enhanced cytotoxic effects on MCF-7 cells while the chemotherapeutic drug alone had no effect. In cellular motility assays, Ent-9-(RS)-12-epi-ST-Δ10-13-PhytoF could significantly inhibit cellular migration of MDA-MB-231 cells. In addition, Ent-9-(RS)-12-epi-ST-Δ10-13-PhytoF also enhanced cellular adhesion of MDA-MB-231 cells.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Movement/drug effects , Furans/pharmacology , Prostanoic Acids/pharmacology , alpha-Linolenic Acid/chemistry , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Female , Furans/chemistry , Humans , MCF-7 Cells , Oxidation-Reduction , Prostanoic Acids/chemistry
5.
Nat Commun ; 7: 13648, 2016 12 05.
Article in English | MEDLINE | ID: mdl-27917859

ABSTRACT

The development of metastases largely relies on the capacity of cancer cells to invade extracellular matrices (ECM) using two invasion modes termed 'mesenchymal' and 'amoeboid', with possible transitions between these modes. Here we show that the SCN4B gene, encoding for the ß4 protein, initially characterized as an auxiliary subunit of voltage-gated sodium channels (NaV) in excitable tissues, is expressed in normal epithelial cells and that reduced ß4 protein levels in breast cancer biopsies correlate with high-grade primary and metastatic tumours. In cancer cells, reducing ß4 expression increases RhoA activity, potentiates cell migration and invasiveness, primary tumour growth and metastatic spreading, by promoting the acquisition of an amoeboid-mesenchymal hybrid phenotype. This hyperactivated migration is independent of NaV and is prevented by overexpression of the intracellular C-terminus of ß4. Conversely, SCN4B overexpression reduces cancer cell invasiveness and tumour progression, indicating that SCN4B/ß4 represents a metastasis-suppressor gene.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Movement , Genes, Tumor Suppressor , Voltage-Gated Sodium Channel beta-4 Subunit/genetics , Animals , Breast Neoplasms/ultrastructure , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Disease Progression , Down-Regulation/genetics , Epithelial Cells/metabolism , Extracellular Matrix/metabolism , Female , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ion Channel Gating , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Prognosis , Protein Subunits/metabolism , Sodium Channels/metabolism , Voltage-Gated Sodium Channel beta-4 Subunit/metabolism , Zebrafish , rhoA GTP-Binding Protein/metabolism
6.
Front Pharmacol ; 7: 338, 2016.
Article in English | MEDLINE | ID: mdl-27774064

ABSTRACT

Studies have demonstrated the significant role of cholesterol and lipoprotein metabolism in the progression of cancer. The SCARB1 gene encodes the scavenger receptor class B type I (SR-BI), which is an 82-kDa glycoprotein with two transmembrane domains separated by a large extracellular loop. SR-BI plays an important role in the regulation of cholesterol exchange between cells and high-density lipoproteins. Accordingly, hepatic SR-BI has been shown to play an essential role in the regulation of the reverse cholesterol transport pathway, which promotes the removal and excretion of excess body cholesterol. In the context of atherosclerosis, SR-BI has been implicated in the regulation of intracellular signaling, lipid accumulation, foam cell formation, and cellular apoptosis. Furthermore, since lipid metabolism is a relevant target for cancer treatment, recent studies have focused on examining the role of SR-BI in this pathology. While signaling pathways have initially been explored in non-tumoral cells, studies with cancer cells have now demonstrated SR-BI's function in tumor progression. In this review, we will discuss the role of SR-BI during tumor development and malignant progression. In addition, we will provide insights into the transcriptional and post-transcriptional regulation of the SCARB1 gene. Overall, studying the role of SR-BI in tumor development and progression should allow us to gain useful information for the development of new therapeutic strategies.

7.
Mod Pathol ; 29(2): 182-93, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26769141

ABSTRACT

Gastric cancers are the most frequent gastric malignancy and usually arise in the sequence of Helicobacter pylori-associated chronic gastritis. CpG methylation is a central mechanism of epigenetic gene regulation affecting cancer-related genes, and occurs early in gastric carcinogenesis. DNA samples from non-metaplastic gastric mucosa with variable levels of gastritis (non-metaplastic mucosa), intestinal metaplasia, or gastric cancer were screened with methylation arrays for CpG methylation of cancer-related genes and 30 gene targets were further characterized by high-definition bisulfite next-generation sequencing. In addition, data from The Cancer Genome Atlas were analyzed for correlation of methylation with gene expression. Overall, 13 genes had significantly increased CpG methylation in gastric cancer vs non-metaplastic mucosa (BRINP1, CDH11, CHFR, EPHA5, EPHA7, FGF2, FLI1, GALR1, HS3ST2, PDGFRA, SEZ6L, SGCE, and SNRPN). Further, most of these genes had corresponding reduced expression levels in gastric cancer compared with intestinal metaplasia, including novel hypermethylated genes in gastric cancer (FLI1, GALR1, SGCE, and SNRPN), suggesting that they may regulate neoplastic transformation from non-malignant intestinal metaplasia to cancer. Our data suggest a tumor-suppressor role for FLI1 in gastric cancer, consistent with recently reported data in breast cancer. For the genes with strongest methylation/expression correlation, namely FLI1, the expression was lowest in microsatellite-unstable tumors compared with other gastric cancer molecular subtypes. Importantly, reduced expression of hypermethylated BRINP1 and SGCE was significantly associated with favorable survival in gastric cancer. In summary, we report novel methylation gene targets that may have functional roles in discrete stages of gastric carcinogenesis and may serve as biomarkers for diagnosis and prognosis of gastric cancer.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Cell Transformation, Neoplastic/genetics , CpG Islands , DNA Methylation , Epigenesis, Genetic , Gastric Mucosa/chemistry , High-Throughput Nucleotide Sequencing , Sequence Analysis, DNA/methods , Stomach Neoplasms/genetics , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Cell Transformation, Neoplastic/pathology , Computational Biology , Databases, Genetic , Disease Progression , Gastrectomy , Gastric Mucosa/pathology , Gastric Mucosa/surgery , Gastritis/genetics , Gastritis/pathology , Genetic Predisposition to Disease , Humans , Metaplasia , Phenotype , Predictive Value of Tests , Prognosis , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery
8.
Front Cardiovasc Med ; 2: 27, 2015.
Article in English | MEDLINE | ID: mdl-26664898

ABSTRACT

Epidemiological studies have demonstrated the importance of cardiovascular diseases in Western countries. Among the cell types associated with a dysfunctional vasculature, smooth muscle (SM) cells are believed to play an essential role in the development of these illnesses. Vascular SM cells are key regulators of the vascular tone and also have an important function in the development of atherosclerosis and restenosis. While in the normal vasculature, contractile SM cells are predominant, in atherosclerotic vascular lesions, synthetic cells migrate toward the neointima, proliferate, and synthetize extracellular matrix proteins. In the present study, we have examined the role of caveolin-3 in the regulation of SM cell phenotype. Caveolin-3 is expressed in vivo in normal arterial SM cells, but its expression appears to be lost in cultured SM cells. Our data show that caveolin-3 expression in the A7r5 SM cell line is associated with increased expression of contractility markers such as SM α-actin, SM myosin heavy chain but decreased expression of the synthetic phenotype markers such as p-Elk and Klf4. Moreover, we also show that caveolin-3 expression can reduce proliferation upon treatment with LDL or PDGF. Finally, we show that caveolin-3-expressing SM cells are less sensitive to apoptosis than control cells upon treatment with oxidized LDL. Taken together, our data suggest that caveolin-3 can regulate the phenotypic switch between contractile and synthetic SM cells. A better understanding of the factors regulating caveolin-3 expression and function in this cell type will permit the development of a better comprehension of the factors regulating SM function in atherosclerosis and restenosis.

9.
Cell Tissue Res ; 358(3): 821-31, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25322709

ABSTRACT

Atherosclerosis is a complex disease initiated by the vascular accumulation of lipoproteins in the sub-endothelial space, followed by the infiltration of monocytes into the arterial intima. Caveolin-1 (Cav-1) plays an essential role in the regulation of cellular cholesterol metabolism and of various signaling pathways. In order to study specifically the role of macrophage Cav-1 in atherosclerosis, we used Cav-1 (-/-) Apoe (-/-) mice and transplanted them with bone marrow (BM) cells obtained from Cav-1 (+/+) Apoe (-/-) or Cav-1 (-/-) Apoe (-/-) mice and vice versa. We found that Cav-1 (+/+) mice harboring Cav-1 (-/-) BM-derived macrophages developed significantly larger lesions than Cav-1 (+/+) mice harboring Cav-1 (+/+) BM-derived macrophages. Cav-1 (-/-) macrophages were more susceptible to apoptosis and more prone to induce inflammation. The present study provides clear evidence that the absence of Cav-1 in macrophage is pro-atherogenic, whereas its absence in endothelial cells protects against atherosclerotic lesion formation. These findings demonstrate the cell-specific role of Cav-1 during the development of this disease.


Subject(s)
Atherosclerosis/pathology , Caveolin 1/metabolism , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Animals , Apoptosis/drug effects , Atherosclerosis/blood , Bone Marrow Transplantation , Caveolin 1/deficiency , Cytokines/metabolism , Inflammation/pathology , Lipopolysaccharides/pharmacology , Lipoproteins/blood , Macrophages, Peritoneal/drug effects , Mice, Inbred C57BL , Up-Regulation/drug effects
10.
Cell Tissue Res ; 356(1): 147-57, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24390341

ABSTRACT

Clinical studies have established the important impact of atherosclerotic disease in Western societies. This disease is characterized by the accumulation of lipids and the migration of various cell types in the sub-endothelial space of blood vessels. As demonstrated by many studies, endothelial cells play an essential role in the development of this disease. The endothelium acts as a gatekeeper of blood vessel integrity and cardiovascular health status. For instance, the transfer of lipids via the transport of lipoproteins in the arterial intima is believed to be mediated by endothelial cells through a process termed transcytosis. In addition, lipoproteins that accumulate in the sub-endothelial space may also be modified, in a process that can direct the activation of endothelial cells. These steps are essential for the initiation of an atherosclerotic plaque and may be mediated, at least in part, by caveolae and their associated protein caveolin-1. In the present study, we evaluate the role of caveolin-1/caveolae in the regulation of these two steps in endothelial cells. Our data clearly demonstrate that caveolin-1 is involved in the regulation of lipoprotein transcytosis across endothelial cells and in the regulation of vascular inflammation.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/pathology , Caveolin 1/metabolism , Endothelial Cells/metabolism , Albumins/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Caveolae/metabolism , Down-Regulation , Endocytosis , Endothelial Cells/pathology , Gene Silencing , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Inflammation/pathology , Lipoproteins, LDL/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , RNA, Small Interfering/metabolism , Signal Transduction
11.
Breast Cancer Res ; 15(5): R87, 2013.
Article in English | MEDLINE | ID: mdl-24060386

ABSTRACT

INTRODUCTION: Previous studies have identified cholesterol as an important regulator of breast cancer development. High-density lipoprotein (HDL) and its cellular receptor, the scavenger receptor class B type I (SR-BI) have both been implicated in the regulation of cellular cholesterol homeostasis, but their functions in cancer remain to be established. METHODS: In the present study, we have examined the role of HDL and SR-BI in the regulation of cellular signaling pathways in breast cancer cell lines and in the development of tumor in a mouse xenograft model. RESULTS: Our data show that HDL is capable of stimulating migration and can activate signal transduction pathways in the two human breast cancer cell lines, MDA-MB-231 and MCF7. Furthermore, we also show that knockdown of the HDL receptor, SR-BI, attenuates HDL-induced activation of the phosphatidylinositol 3-kinase (PI3K)/protein Kinase B (Akt) pathway in both cell lines. Additional investigations show that inhibition of the PI3K pathway, but not that of the mitogen-activated protein kinase (MAPK) pathway, could lead to a reduction in cellular proliferation in the absence of SR-BI. Importantly, whereas the knockdown of SR-BI led to decreased proliferation and migration in vitro, it also led to a significant reduction in tumor growth in vivo. Most important, we also show that pharmacological inhibition of SR-BI can attenuate signaling and lead to decreased cellular proliferation in vitro. Taken together, our data indicate that both cholesteryl ester entry via HDL-SR-BI and Akt signaling play an essential role in the regulation of cellular proliferation and migration, and, eventually, tumor growth. CONCLUSIONS: These results identify SR-BI as a potential target for the treatment of breast cancer.


Subject(s)
Breast Neoplasms/metabolism , CD36 Antigens/metabolism , Cell Transformation, Neoplastic , Cholesterol/metabolism , Signal Transduction , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , CD36 Antigens/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation , Cholesterol/pharmacology , Cholesterol, HDL/metabolism , Cholesterol, HDL/pharmacology , Disease Models, Animal , Enzyme Activation/drug effects , Female , Gene Knockdown Techniques , Heterografts , Humans , Lipoproteins, HDL/genetics , Lipoproteins, HDL/metabolism , MCF-7 Cells , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Lipoprotein/genetics , Receptors, Lipoprotein/metabolism , Tumor Burden/genetics
12.
Adv Exp Med Biol ; 729: 127-44, 2012.
Article in English | MEDLINE | ID: mdl-22411318

ABSTRACT

Atherosclerosis is a disease of the blood vessel characterized by the development of an arterial occlusion containing lipid and cellular deposits. Caveolae are 50-100 nm cell surface plasma membrane invaginations that are believed to play an important role in the regulation of cellular signaling and transport of molecules among others. These organelles are enriched in sphingolipids and cholesterol and are characterized by the presence of the protein caveolin-1. Caveolin-1 and caveolae are present in most of the cells involved in the development of atherosclerosis. The current literature suggests a rather complex role for caveolin-1 in this disease, with evidence of either pro- or anti-atherogenic functions depending on the cell type examined. In the present chapter, the various roles of caveolae and caveolin-1 in the development of atherosclerosis are examined.


Subject(s)
Atherosclerosis , Caveolae , Caveolin 1 , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Caveolae/metabolism , Caveolin 1/chemistry , Caveolin 1/deficiency , Caveolin 1/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Macrophages/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology
13.
J Cell Mol Med ; 12(4): 1130-50, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18400052

ABSTRACT

Caveolae are small plasma membrane invaginations that have been implicated in a variety of functions including transcytosis, potocytosis and cholesterol transport and signal transduction. The major protein component of this compartment is a family of proteins called caveolins. Experimental data obtained in knockout mice have provided unequivocal evidence for a requirement of caveolins to generate morphologically detectable caveolae structures. However, expression of caveolins is not sufficient per seto assure the presence of these structures. With respect to other roles attributed to caveolins in the regulation of cellular function, insights are even less clear. Here we will consider, more specifically, the data concerning the ambiguous roles ascribed to caveolin-1 in signal transduction and cancer. In particular, evidence indicating that caveolin-1 function is cell context dependent will be discussed.


Subject(s)
Caveolin 1/metabolism , Neoplasms/metabolism , Signal Transduction , Animals , Humans
14.
Reprod Toxicol ; 17(6): 667-72, 2003.
Article in English | MEDLINE | ID: mdl-14613818

ABSTRACT

This work was undertaken to examine possible embryotoxicity of Ruta graveolens (rue), a plant used by indigenous communities for the purposes of therapeutic and fertility regulation. Superovulated mice were mated and isolated after copulation. They were given aqueous extract of R. graveolens (5, 10, and 20% w/v) or plain water (control) orally for 4 days. Ninety-eight hours post-human chorionic gonadotrophin (hCG), embryos were flushed from oviducts and uterine horns to assess their state of development and extent of embryo transport. Ingestion of rue at 10 and 20% resulted in a high proportion of abnormal embryos (36.7 and 63.6%, respectively, P<0.05). Cell number was diminished (P<0.01) and embryo transport was slightly delayed in the highest dose group. These findings demonstrate that oral administration of R. graveolens extract can interfere with preimplantation development and embryo transport.


Subject(s)
Blastocyst/drug effects , Ruta/chemistry , Animals , Body Weight/drug effects , Cell Count , Dose-Response Relationship, Drug , Embryonic and Fetal Development/drug effects , Female , Injections, Intraperitoneal , Mice , Morula/drug effects , Plant Extracts/pharmacology , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...