Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Type of study
Publication year range
1.
PLoS One ; 7(10): e47480, 2012.
Article in English | MEDLINE | ID: mdl-23094054

ABSTRACT

Exosomes are of increasing interest as alternative mode of cell-to-cell communication. We previously reported that exosomes secreted by human SOJ-6 pancreatic tumor cells induce (glyco)protein ligand-independent cell death and inhibit Notch-1 pathway, this latter being particularly active during carcinogenesis and in cancer stem cells. Therefore, we asked whether exosomal lipids were key-elements for cell death and hypothesized that cholesterol-rich membrane microdomains were privileged sites of exosome interactions with tumor cells. To address these questions and based on the lipid composition of exosomes from SOJ-6 cells (Ristorcelli et al. (2008) FASEB J. 22; 3358-3369) enriched in cholesterol and sphingomyelin (lipids forming liquid-ordered phase, Lo) and depleted in phospholipids (lipids forming liquid-disordered phase, Ld), we designed Synthetic Exosome-Like Nanoparticles (SELN) with ratios Lo/Ld from 3.0 to 6.0 framing that of SOJ-6 cell exosomes. SELN decreased tumor cell survival, the higher the Lo/Ld ratio, the lower the cell survival. This decreased survival was due to activation of cell death with inhibition of Notch pathway. FRET analyses indicated fusions/exchanges of SELN with cell membranes. Fluorescent SELN co-localized with the ganglioside GM1 then with Rab5A, markers of lipid microdomains and of early endosomes, respectively. These interactions occurred at lipid microdomains of plasma and/or endosome membranes where the Notch-1 pathway matures. We thus demonstrated a major role for lipids in interactions between SELN and tumor cells, and in the ensued cell death. To our knowledge this is the first report on such effects of lipidic nanoparticles on tumor cell behavior. This may have implications in tumor progression.


Subject(s)
Biomimetic Materials/pharmacology , Exosomes/metabolism , Nanoparticles/chemistry , Pancreatic Neoplasms/metabolism , Receptor, Notch1/metabolism , Biological Transport , Biomimetic Materials/chemistry , Cell Communication , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cholesterol/chemistry , Cholesterol/pharmacology , Exosomes/chemistry , G(M1) Ganglioside/chemistry , Gene Expression , Humans , Membrane Microdomains/drug effects , Microscopy, Fluorescence , Nanoparticles/ultrastructure , Pancreatic Neoplasms/ultrastructure , Receptor, Notch1/genetics , Signal Transduction , Sphingomyelins/chemistry , Sphingomyelins/pharmacology , rab5 GTP-Binding Proteins/metabolism
2.
J Immunol ; 186(7): 4067-77, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21346236

ABSTRACT

Aberrant glycosylation or overexpression of cell-surface glycosylated tumor-associated Ags (TAA) distinguish neoplastic from normal cells. Interactions of TAA MUC1 and HER2/neu with dendritic cells (DC) preclude efficient processing, which impairs immune responses. It is thus important to define the mechanisms of interactions between DC and glycosylated TAA and their trafficking and processing for further T cell activation. In this work, we study interactions between DC and the oncofetal fucose-rich glycovariants of bile salt-dependent lipase (BSDL), expressed in pancreatic cancer tissues and referred to as pathological BSDL carrying the fucosylated J28 glycotope (pBSDL-J28) because it is characterized by the mAb J28. The expression of pBSDL-J28 was assessed by immunohistochemistry and quantified by confocal microscopy. Nontumoral pancreatic tissues and cells do not express pBSDL-J28. Using multidisciplinary approaches and functional studies, we provide the first evidence, to our knowledge, that this tumoral glycoprotein is rapidly internalized by human DC through macropinocytosis and endocytosis via mannose receptors and then transported to late endosomes for processing. Interestingly, pBSDL-J28 per se induced DC maturation with increased expression of costimulatory and CD83 molecules associated with cytokine secretion (IL-8 and IL-6). Surprisingly, DC retained their full ability to internalize Ags, making this maturation atypical. Finally, the allogeneic pBSDL-J28-treated DC stimulated lymphocyte proliferation. Besides, pulsing DC with pBSDL-J28 C-terminal glycopolypeptide and maturation with CD40L triggered CD4(+) and CD8(+) T cell proliferation. Therefore, interactions of pBSDL-J28, expressed on tumoral pancreatic tissue, with DC may lead to adequate Ag trafficking and processing and result in T cell activation.


Subject(s)
Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Cell Differentiation/immunology , Dendritic Cells/immunology , Endocytosis/immunology , Pancreatic Neoplasms/immunology , Sterol Esterase/metabolism , Antigen Presentation/immunology , Antigens, Neoplasm/physiology , Biomarkers, Tumor/physiology , Coculture Techniques , Dendritic Cells/metabolism , Dendritic Cells/pathology , HEK293 Cells , Humans , Lectins, C-Type/metabolism , Lymphocyte Activation/immunology , Mannose Receptor , Mannose-Binding Lectins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Transport/immunology , Receptors, Cell Surface/metabolism , Sterol Esterase/physiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
3.
J Biochem ; 149(2): 145-51, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21081507

ABSTRACT

Bile salt-dependent lipase was purified to homogeneity from lyophilized human milk and used to screen the influence of the acyl chain length (2-16 carbon atoms) on the kinetic constants k(cat) and K(m) of the hydrolysis of para-nitrophenyl (pnp) ester substrates in the presence or absence of sodium taurocholate (NaTC: 0.02-20 mM). The highest k(cat) value (∼3,500 s(-1)) was obtained with pnpC(8) as substrate, whereas the lowest K(m) (<10 µM) was that recorded with pnpC(10). In the absence of NaTC, the maximal catalytic efficiency (k(cat)/K(m)) was obtained with pnpC(8), while in the presence of NaTC k(cat)/K(m) was maximal with pnpC(8), pnpC(10) or pnpC(12). The bile salt activated the enzyme in two successive saturation phases occurring at a micromolar and a millimolar concentration range, respectively. The present data emphasize the suitability of this enzyme for the hydrolysis of medium-chain acyl-containing substrates and throw additional light on how BSDL is activated by NaTC.


Subject(s)
Sterol Esterase/metabolism , Taurocholic Acid/metabolism , Bile Acids and Salts/metabolism , Catalysis , Enzyme Activation , Esters/chemistry , Humans , Hydrogen-Ion Concentration , Hydrolysis , Kinetics , Micelles , Milk, Human/enzymology , Nitrophenols/chemistry , Substrate Specificity
4.
Int J Cancer ; 125(5): 1016-26, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19405120

ABSTRACT

We previously reported that exosomal nanoparticles secreted by human pancreatic tumoral cell lines decrease tumoral cell proliferation through the mitochondria-dependent apoptotic pathway, because of activation of pro-apoptotic phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and of glucose synthase kinase-3beta (GSK-3beta). Interactions between exosomal nanoparticles and cells are thought to involve membrane lipid rafts. However, the underlying mechanism is unknown. Here, we report that the interaction of exosomal nanoparticles with pancreatic cancer cells led to decreased expression of hairy and enhancer-of-split homolog-1 (Hes-1), the intranuclear target of Notch-1 signaling pathway, and to activation of the apoptotic pathway after a cell cycle arrest in G(0)G(1) phase. Strikingly, the expression level of Notch-1 pathway components was critical, because exosomal nanoparticles decreased the proliferation of cells in which these partners are either weakly represented, in differentiated adenocarcinoma cells, or inhibited, in poorly differentiated carcinoma cells, by blocking presenilin in the gamma-secretase complex that regulates the Notch-1 pathway. Overexpression of Notch-1 intracellular domain resulted in the reversion of the cell proliferation inhibition promoted by exosomal nanoparticles. Blocking presenilin unexpectedly resulted in activation of PTEN and GSK-3beta. Conversely, inhibiting either PTEN or GSK-3beta increased Hes-1 expression and partially counteracted the inhibition of proliferation promoted by exosomal nanoparticles, highlighting reciprocal regulations between Notch signaling and PTEN/GSK-3beta. We concluded that interactions of exosomal nanoparticles with target cells, at lipid rafts where Notch-1 pathway partners are localized, hampered the functioning of the Notch-1 survival pathway and activated the apoptotic pathway, which determines tumoral cell fate.


Subject(s)
Adenocarcinoma/pathology , Apoptosis , Exosomes/metabolism , Nanoparticles , Pancreatic Neoplasms/pathology , Receptors, Notch/physiology , Adenocarcinoma/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Carbamates/pharmacology , Caspase 3/metabolism , Cell Differentiation , Cell Proliferation , Dipeptides/pharmacology , Flow Cytometry , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mitochondria/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Pancreatic Neoplasms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor HES-1 , Tumor Cells, Cultured
5.
Cancer Res ; 69(3): 802-9, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19176384

ABSTRACT

Aggressive melanoma cells can engage in a process termed vasculogenic mimicry (VM) that reflects the ability of tumor cells to express a multipotent, stem cell-like phenotype. Melanoma cell plasticity contributes to the lack of efficient therapeutic strategies targeting metastatic tumors. This study reveals cyclic AMP as a mediator of VM in vitro. In uveal and cutaneous metastatic aggressive human melanoma cells, an increase in cyclic AMP by forskolin, dibutyryl cyclic AMP, or G protein-coupled receptor (GPCR) ligands such as adrenaline and vasoactive intestinal peptide inhibited VM to different extents. Although chemical modulators of protein kinase A (PKA) had no effect, a specific pharmacologic activator of Exchange protein directly activated by cyclic AMP (Epac) impaired VM. Ras-associated protein-1 (Rap1) activation assays revealed that cyclic AMP-elevating agents induce a PKA-independent activation of Epac/Rap1. Pharmacologic inhibition of extracellular signal-regulated kinase 1/2 (ERK1/2) activity abolished VM. Phosphorylation of ERK1/2 was PKA-independently inhibited by forskolin but not inhibited by Epac/Rap1 signaling, PKA modulation, or GPCR ligands. Furthermore, the forskolin also inhibited phosphatidyl inositol-3-kinase (PI3K)-mediated activation of protein kinase Akt, as monitored by Ser473 phosphorylation. The pharmacologic activation of Epac and GPCR ligands slightly stimulated Akt, a likely concomitant process of VM modulation. Collectively, these data show that forskolin strongly inhibits VM through PKA-independent activation of Epac/Rap1, PKA-, and Epac-independent inactivation of ERK1/2 and inhibition of PI3K/Akt. The data also show that VM inhibition by GPCR ligands involves mainly the Epac/Rap1-activated signal. Thus cyclic AMP inhibits VM through multiple signaling pathways.


Subject(s)
Cyclic AMP/metabolism , Melanoma/metabolism , Melanoma/pathology , 1-Methyl-3-isobutylxanthine/pharmacology , Cell Line, Tumor , Colforsin/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , Humans , Ligands , MAP Kinase Signaling System/drug effects , Melanoma/blood supply , Microvessels/drug effects , Microvessels/metabolism , Microvessels/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, G-Protein-Coupled/metabolism , Shelterin Complex , Signal Transduction/drug effects , Telomere-Binding Proteins/metabolism , Vasoactive Intestinal Peptide/pharmacology , alpha-MSH/pharmacology
6.
Gene ; 423(2): 160-71, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18691641

ABSTRACT

The syntaxins are proteins associated with various intracellular membrane compartments. They are major participants in a large variety of physiological processes where membrane fusion occurs, including exocytosis. We have identified a novel syntaxin isoform generated by alternative splicing of the human STX1B gene. In contrast with the canonical syntaxins, this isoform (STX1B-DeltaTMD) lacked the classical C-terminal transmembrane domain and localized to the nucleus of various tumoral and non-tumoral cell types including human brain cortical neurons in vivo. The reversible blockade of STX1B-DeltaTMD nuclear import demonstrated that nuclear import occurred via a Ran-dependent pathway. A specific and glycine-rich C-terminus of 15 amino acids served as an unconventional nuclear localization signal. STX1B-DeltaTMD colocalized with Lamin A/C and NuMA (NUclear Mitotic Apparatus protein) in interphasic nuclei, and with NuMA and gamma-tubulin in the pericentrosomal region of the mitotic spindle in dividing cells. In a series of 37 human primary brain tumors, the ratio of STX1B-DeltaTMD to Lamin A/C transcripts was a significant prognostic marker of survival, independent of tumor staging. The characterization of STX1B-DeltaTMD as the first nucleoplasmic syntaxin with no transmembrane domain, illustrates the importance of alternative splicing in the emergence of unsuspected properties of the syntaxins in human cells, in both physiological and pathological conditions.


Subject(s)
Cell Nucleus/metabolism , Syntaxin 1/metabolism , Alternative Splicing/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival , Centrosome/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lamin Type A/genetics , Mutant Proteins/metabolism , Nuclear Matrix/metabolism , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Structure, Tertiary , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Syntaxin 1/chemistry , ran GTP-Binding Protein/metabolism
7.
FASEB J ; 22(9): 3358-69, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18511551

ABSTRACT

Exosomes are vesicles secreted by most hematopoietic cells on fusion of multivesicular endosomes with the plasma membrane. Many studies have reported that exosomes may also be released by tumor cells. Exosomes are believed to play an antitumor role through immune cells. We asked whether tumor exosomes have biological activities on tumor cells. We report that human pancreatic tumor nanoparticles, exosome-like as characterized by proteomic analyses and rich in lipid rafts, decreased tumor cell proliferation. Nanoparticles increased Bax and decreased Bcl-2 expressions. Caspase-3 and -9 but not caspase-8 inhibitors impaired apoptosis, which implicates the mitochondria apoptotic pathway. The ceramide-sphingomyelin apoptotic pathway was inoperative. Moreover, nanoparticles induced phosphatase and tensin homolog (PTEN) and glycogen synthase kinase (GSK) -3beta activation and decreased pyruvate dehydrogenase activity. In nanoparticle-treated cells, PTEN formed complexes with actin, beta-catenin, and GSK-3beta. Thus, beta-catenin may no longer be available to activate the survival pathway. Nanoparticles triggered the down-regulation of cyclin D1 and poly(ADP-ribose) polymerase. Hence, nanoparticles counteracted the constitutively activated phosphatidylinositol 3-kinase/Akt survival pathway to drive tumor cells toward apoptosis. Our study provides the first evidence of an apoptotic function of tumor-derived nanoparticles on tumor cells. We propose a new role for nanoparticles, i.e., as signal carriers for interaction between cells, which may have implications in physiopathological situations.


Subject(s)
Apoptosis/drug effects , Membrane Microdomains , Nanoparticles , Pancreatic Neoplasms/pathology , Caspase Inhibitors , Cell Line, Tumor , Ceramides/physiology , Endosomes/physiology , Glycogen Synthase Kinase 3/physiology , Glycogen Synthase Kinase 3 beta , Humans , Lipids/analysis , Membrane Microdomains/physiology , Neoplasm Proteins/analysis , PTEN Phosphohydrolase/metabolism , Pancreatic Neoplasms/physiopathology , Phosphatidylinositol 3-Kinases/physiology , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-akt/physiology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Pyruvate Dehydrogenase Complex/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , bcl-2-Associated X Protein/biosynthesis
8.
Biochim Biophys Acta ; 1736(1): 67-76, 2005 Sep 05.
Article in English | MEDLINE | ID: mdl-16099206

ABSTRACT

The relationship between cholesterol and atherosclerosis has gained wide credence and red wine polyphenols have been shown to have an anti-atherogenic activity. In the present in vitro studies, we have evaluated and compared the effects of resveratrol, an active compound of red wine, and of a whole red wine polyphenolic extract (RWE) on the pancreatic bile salt-dependent lipase (BSDL). BSDL is involved in the duodenal hydrolysis of lipid esters and in part of cholesteryl esters thus favoring the bioavailability of free cholesterol. Resveratrol and RWE decrease the human and rat enzyme activities. Resveratrol and RWE also impaired the secretion of BSDL by the rat pancreatic AR4-2J cells used as secreting model. This effect is reversed by the removal of resveratrol or RWE from the cell culture medium. Further, resveratrol (but not RWE) affects the transcription of the gene encoding BSDL and dramatically diminishes the quantity of the enzyme that is expressed and secreted by AR4-2J cells. Results suggest that the hypolipemic effects of red wine polyphenols could partly originate from the inhibition of BSDL activity and secretion in the duodenum. In vivo, these effects could decrease the hydrolysis of dietary lipid esters and likely the absorption of free cholesterol.


Subject(s)
Flavonoids/physiology , Pancreas/enzymology , Sterol Esterase/metabolism , Animals , Cell Line, Tumor , Humans , Pancreas/drug effects , Pancreas/metabolism , Phenols , Polyphenols , RNA, Messenger/metabolism , Rats , Resveratrol , Sterol Esterase/biosynthesis , Sterol Esterase/genetics , Stilbenes/pharmacology , Wine , alpha-Amylases/metabolism
9.
Arterioscler Thromb Vasc Biol ; 25(2): 359-64, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15569825

ABSTRACT

OBJECTIVE: Bile salt-dependent lipase (BSDL), a lipolytic enzyme secreted in the duodenum by pancreatic acinar cells, has been detected in the serum of all patients and in atheromatous plaque, suggesting its potential implication in vascular pathophysiology. METHODS AND RESULTS: In vitro pancreatic BSDL evokes human umbilical vein endothelial cell (HUVEC) proliferation and chemotactic migration. BSDL at mitogen concentration is capable to heal wounded HUVEC monolayer and to promote capillary network formation. HUVEC proliferation depends on the displacement of basic fibroblast growth factor and vascular endothelial growth factor from the extracellular matrix and the activation of extracellular signal-regulated kinases (ERK1/2), p38 mitogen-activated protein kinase, and focal adhesion kinase signaling pathways. CONCLUSIONS: For the first time to our knowledge, it is suggested that circulating BSDL could be involved in pathophysiological angiogenesis. We delineate the in vitro effects of pancreatic BSDL on endothelial cells, and we show that BSDL promotes proliferation, migration, capillary network formation, and wound-healing of HUVECs via the displacement of bFGF and VEGF from the ECM, suggesting that BSDL could be involved in angiogenesis.


Subject(s)
Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Sterol Esterase/pharmacology , Animals , Cell Division/drug effects , Cells, Cultured/drug effects , Chemotaxis/drug effects , Collagen , DNA Replication/drug effects , Drug Combinations , Endothelium, Vascular/cytology , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblast Growth Factor 2/metabolism , Humans , Laminin , MAP Kinase Signaling System/drug effects , Macaca fascicularis , Neovascularization, Physiologic/physiology , Pancreas/enzymology , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Proteoglycans , Umbilical Veins , Vascular Endothelial Growth Factor A/metabolism , Wound Healing
10.
Mol Biol Cell ; 14(7): 2861-75, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12857870

ABSTRACT

We have recently shown that the pancreatic bile salt-dependent lipase (BSDL) can be taken up by intestinal cells and transported to the blood circulation. This mechanism likely involves (specific) receptor(s) able to bind BSDL and located at the apical intestinal cell membrane. In this study, using Int407 human intestinal cells cultured to form a tight epithelium, we attempted to characterize (the) BSDL receptor(s). We found that an apical 50-kDa protein was able to bind BSDL. Further, we have demonstrated that Int407 cells expressed the lectin-like oxidized-LDL receptor (LOX-1), the upregulation of which by oxidized-LDL potentiates the transcytosis of BSDL, whereas carrageenan and to a lesser extent polyinosinic acid and fucoidan decrease the enzyme transcytosis. The mAb JTX92, which blocks the LOX-1 receptor function, also impaired the BSDL transcytosis. To confirm these results, the cDNA encoding the human intestinal receptor LOX-1 has been cloned, inserted into vectors, and transfected into Int407 cells. Overexpression of LOX-1 by these cells leads to a substantial increase in the BSDL transcytosis. Globally, these data support the view that LOX-1 could be an intestinal receptor for BSDL, which is implicated in the transcytosis of this enzyme throughout Int407 cells.


Subject(s)
Endocytosis/physiology , Receptors, LDL/metabolism , Sterol Esterase/metabolism , Cell Line , Cloning, Molecular , Endocytosis/drug effects , Flow Cytometry , Humans , Intestinal Mucosa/metabolism , Intestines/cytology , Lipoproteins, LDL/metabolism , Poly I/pharmacology , Polysaccharides/pharmacology , Protein Binding , Receptors, Oxidized LDL , Scavenger Receptors, Class E , Transferrin/metabolism
11.
Circulation ; 108(1): 86-91, 2003 Jul 08.
Article in English | MEDLINE | ID: mdl-12821548

ABSTRACT

BACKGROUND: Because bile salt-dependent lipase (BSDL), an enzyme secreted by the pancreatic acinar cells and associated with LDL in circulating blood, also locates with smooth muscle cells (SMCs) in atherosclerotic lesions, we aimed to investigate its effects on SMCs. METHODS AND RESULTS: Immunohistochemical experiments allowed us to detect an expression of BSDL in atherosclerotic lesions from hypercholesterolemic monkeys and from human arteries. BSDL was found to be associated with SMCs but not with macrophages. BSDL was significantly mitogenic for cultured SMCs. This effect was inhibited by heparin and anti-BSDL antibodies, whereas heat-denaturated and diisopropylfluorophosphate-treated BSDL were inefficient. The mitogenic effect of BSDL was associated with an activation of the extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase pathway, which was inhibited by heparin, and involved several mechanisms, among them diacylglycerol and oleic acid production as well as a rapid basic fibroblast growth factor release. CONCLUSIONS: Circulating BSDL is associated with SMCs within the intimal arteria and may trigger SMC proliferation, which could contribute to the development of atherosclerotic lesions.


Subject(s)
Arteriosclerosis/enzymology , Muscle, Smooth, Vascular/drug effects , Pancreas/enzymology , Sterol Esterase/pharmacology , Animals , Antibodies/pharmacology , Aorta/enzymology , Aorta/pathology , Arteriosclerosis/complications , Arteriosclerosis/pathology , Cell Division/drug effects , Enzyme Inhibitors/pharmacology , Humans , Hypercholesterolemia/complications , Hypercholesterolemia/enzymology , Immunohistochemistry , Isoflurophate/pharmacology , Macaca fascicularis , Male , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , RNA, Messenger/metabolism , Rabbits , Second Messenger Systems/drug effects , Sterol Esterase/antagonists & inhibitors , Sterol Esterase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...