Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters










Publication year range
1.
Biochimie ; 125: 281-96, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26717905

ABSTRACT

Atherosclerosis is a multifactorial process that begins early in infancy and affects all the humans. Early steps of atherogenesis and the evolution towards complex atherosclerotic plaques are briefly described. After a brief history of the 'Lipid theory of atherosclerosis', we report the most prominent discoveries on lipoproteins, their receptors and metabolism, and their role in atherogenesis. The main focus is the 'oxidative theory of atherosclerosis', with emphasis on free radicals and reactive oxygen species, lipid peroxidation and LDL oxidation, biological properties of oxidized LDL and their potential role in atherogenesis. Then, we report the properties of antioxidants and antioxidant systems and their effects in vitro, on cultured cells, in animal models and in humans. The surprising discrepancy between the efficacy of antioxidants in vitro and in animal models of atherosclerosis and the lack of protective effect against cardiovascular events and death in epidemiological study and clinical trials are discussed. In contrast, epidemiological studies seem to indicate that the Mediterranean diet may protect (in part) against atherosclerosis complications (myocardial infarction and cardiovascular death).


Subject(s)
Antioxidants/metabolism , Atherosclerosis/metabolism , Models, Cardiovascular , Animals , Atherosclerosis/pathology , Disease Models, Animal , Humans , Oxidation-Reduction
2.
Gynecol Obstet Fertil ; 43(11): 751-6, 2015 Nov.
Article in French | MEDLINE | ID: mdl-26476891

ABSTRACT

Preeclampsia is a leading cause of pregnancy complications and affects 3-7% of pregnant women. Pathophysiology of preeclampsia is still unclear. According to the two-stage model of preeclampsia, the abnormal and hypoperfused placenta (stage 1) releases factors to the bloodstream, which are responsible for the maternal symptoms (stage 2), characterised by a systemic inflammation and endothelial dysfunction. Oxidative stress plays an important role in the pathophysiology of the preeclampsia and could be the common denominator between the two. This review summarizes the current knowledge of a new potential etiology of the disease, with a special focus on oxidative stress. We also review the different factors that have been proposed to cause endothelial cell dysfunction in preeclampsia, and trials investigating the role of antioxidant supplementation in preeclampsia.


Subject(s)
Oxidative Stress , Pre-Eclampsia/etiology , Antioxidants/administration & dosage , Dietary Supplements , Endothelium/physiopathology , Female , Humans , Inflammation , Pre-Eclampsia/physiopathology , Pregnancy
3.
Am J Transplant ; 15(8): 2050-61, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25930666

ABSTRACT

Transplant vasculopathy (TV) represents the main cause of late graft failure and limits the long-term success of organ transplantation. Cellular and humoral immune responses contribute to the pathogenesis of the concentric and diffuse intimal hyperplasia of arteries of the grafted organ. We recently reported that the mitogenic signaling, evoked in human vascular smooth muscle cells (hmSMC) by the anti-HLA class I monoclonal antibody W6/32, implicates neutral sphingomyelinase-2, suggesting a role for sphingolipids in intimal hyperplasia of TV. Here, we investigated whether the mitogenic sphingolipid, sphingosine-1-phosphate (S1P), is involved in intimal hyperplasia elicited by W6/32. Studies were done on cultured hmSMC and on an in vivo model of TV, consisting of human mesenteric arteries grafted into SCID/beige mice, injected weekly with W6/32. hmSMC migration and DNA synthesis elicited by W6/32 were inhibited by the sphingosine kinase-1 (SK1) inhibitor dimethylsphingosine, the anti-S1P antibody Sphingomab and the S1PR1/R3 inhibitor VPC23019. W6/32 stimulated SK1 activity, while siRNA silencing SK1, S1PR1 and S1PR3 inhibited hmSMC migration. In vivo, Sphingomab significantly reduced the intimal thickening induced by W6/32. These data emphasize the role of S1P in intimal hyperplasia elicited by the humoral immune response, and open perspectives for preventing TV with S1P inhibitors.


Subject(s)
Antibodies, Monoclonal/immunology , HLA Antigens/immunology , Lysophospholipids/physiology , Organ Transplantation/adverse effects , Sphingosine/analogs & derivatives , Vascular Diseases/etiology , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Endothelium, Vascular/pathology , Humans , Mice , Mice, SCID , Sphingosine/physiology
4.
Cell Death Dis ; 4: e520, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23449456

ABSTRACT

During atherogenesis, excess amounts of low-density lipoproteins (LDL) accumulate in the subendothelial space where they undergo oxidative modifications. Oxidized LDL (oxLDL) alter the fragile balance between survival and death of vascular smooth muscle cells (VSMC) thereby leading to plaque instability and finally to atherothrombotic events. As protein kinase C δ (PKCδ) is pro-apoptotic in many cell types, we investigated its potential role in the regulation of VSMC apoptosis induced by oxLDL. We found that human VSMC silenced for PKCδ exhibited a protection towards oxLDL-induced apoptosis. OxLDL triggered the activation of PKCδ as shown by its phosphorylation and nuclear translocation. PKCδ activation was dependent on the reactive oxygen species generated by oxLDL. Moreover, we demonstrated that PKCδ participates in oxLDL-induced endoplasmic reticulum (ER) stress-dependent apoptotic signaling mainly through the IRE1α/JNK pathway. Finally, the role of PKCδ in the development of atherosclerosis was supported by immunohistological analyses showing the colocalization of activated PKCδ with ER stress and lipid peroxidation markers in human atherosclerotic lesions. These findings highlight a role for PKCδ as a key regulator of oxLDL-induced ER stress-mediated apoptosis in VSMC, which may contribute to atherosclerotic plaque instability and rupture.


Subject(s)
Apoptosis , Endoplasmic Reticulum Stress , Lipoproteins, LDL/metabolism , Muscle, Smooth, Vascular/metabolism , Protein Kinase C-delta/metabolism , Animals , Cell Nucleus/metabolism , Cells, Cultured , Endoribonucleases/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Muscle, Smooth, Vascular/cytology , Phosphorylation , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
5.
Cell Death Differ ; 18(5): 817-28, 2011 May.
Article in English | MEDLINE | ID: mdl-21113143

ABSTRACT

The apoptotic effect of oxidized LDLs (oxLDLs) is mediated through a complex sequence of signaling events involving a deregulation of the cytosolic Ca(2+) homeostasis. OxLDLs also trigger ER stress that may lead to cellular dysfunction and apoptosis, through the activation of the IRE1α/c-Jun N-terminal kinase pathway. Moreover, ER stress and oxidized lipids have been shown to trigger autophagy. The antiatherogenic high-density lipoproteins (HDLs) display protective effects against oxLDLs toxicity. To more deeply investigate the mechanisms mediating the protective effects of HDLs, we examined whether ER stress and autophagy were implicated in oxLDLs-induced apoptosis and whether HDLs prevented these stress processes. We report that, in human endothelial cells, HDLs prevent the oxLDL-induced activation of the ER stress sensors IRE1α, eIF2α and ATF6 and subsequent activation of the proapoptotic mediators JNK and CHOP. OxLDLs also trigger the activation of autophagy, as assessed by LC3 processing and Beclin-1 expression. The autophagic process is independent of the proapoptotic arms of ER stress, but Beclin-1 contributes to PS exposure and subsequent phagocytosis of oxLDLs exposed cells. Induction of autophagy and PS exposure by oxLDLs is prevented by HDLs. Finally, the cytosolic Ca(2+) deregulation triggered by oxLDLs is a common signaling pathway that mediates ER stress-induced cell death and autophagy, all these events being blocked by HDLs.


Subject(s)
Autophagy , Endoplasmic Reticulum/physiology , Lipoproteins, HDL/physiology , Lipoproteins, LDL/physiology , Animals , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Biomarkers/metabolism , Calcium Signaling , Caspase 12/metabolism , Cell Line , Cell Survival , Endoribonucleases/metabolism , Endothelial Cells/physiology , Enzyme Activation , Eukaryotic Initiation Factor-2/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Lipoproteins, HDL/pharmacology , Lipoproteins, LDL/pharmacology , Membrane Proteins/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Phagocytosis , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Stress, Physiological , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Unfolded Protein Response
6.
Am J Transplant ; 9(11): 2607-14, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19843036

ABSTRACT

Antibodies toward HLA class I and/or MICA are commonly observed in transplanted patients suffering from allograft arteriosclerosis, also called chronic vascular rejection (CVR). The relative importance of cellular versus humoral alloreactivity for CVR is still disputed. We demonstrate that antibodies toward HLA class I provoke lesions typical for CVR in human arteries in vivo in the absence of cellular immunity. To show this, we grafted segments of human mesenteric arteries from 8 deceased organ donors into 36 immunodeficient SCID/beige mice in the infrarenal aortic position. Three mice died postoperatively. The remaining 33 mice received weekly i.v. injections of either a monoclonal antibody toward HLA class I, toward MICA or an irrelevant monoclonal antibody. At sacrifice after 6 weeks, mice receiving the HLA antibody showed a significant neointimal thickening in the grafted artery due to smooth muscle cell (SMC) proliferation while control mice receiving anti-MICA or irrelevant antibody showed little or no thickening. Whereas antibodies toward HLA class I were mitogenic to SMC in vitro, those directed toward MICA did not have any effect. Humoral alloreactivity toward HLA may thus play a causal role for the development of CVR and this opens new possibilities for the treatment of CVR.


Subject(s)
Antibodies, Heterophile/immunology , Arteriosclerosis/immunology , Graft Rejection/immunology , Histocompatibility Antigens Class I/immunology , Mesenteric Arteries/transplantation , Transplantation, Heterologous/immunology , Animals , Antibodies, Heterophile/blood , Arteriosclerosis/pathology , Cell Division/immunology , Graft Rejection/pathology , Humans , Mesenteric Arteries/immunology , Mesenteric Arteries/pathology , Mice , Mice, SCID , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/pathology , Tunica Intima/immunology , Tunica Intima/pathology
7.
Transplant Proc ; 41(2): 705-6, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19328962

ABSTRACT

We investigated the effects of the sphingolipid FTY720 on tumor necrosis factor-alpha (TNF-alpha)-induced proliferation and signal transduction in human smooth muscle cells (SMC). We showed that clinically relevant concentrations of FTY720 inhibited TNF-alpha-induced SMC proliferation and extracellular signal-regulated kinase (ERK) phosphorylation. We concluded that FTY720 may be a useful drug to inhibit chronic vascular rejection.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Graft Rejection/prevention & control , Immunosuppressive Agents/pharmacology , Muscle, Smooth/physiology , Propylene Glycols/pharmacology , Sphingosine/analogs & derivatives , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Cell Division/drug effects , Cell Survival/drug effects , Cells, Cultured , Fingolimod Hydrochloride , Humans , Immunosuppressive Agents/therapeutic use , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Muscle, Smooth/enzymology , Phosphorylation , Propylene Glycols/therapeutic use , Signal Transduction/drug effects , Signal Transduction/physiology , Sphingosine/pharmacology , Sphingosine/therapeutic use , Transplantation, Homologous/immunology , Tumor Necrosis Factor-alpha/physiology
8.
Cell Death Differ ; 15(8): 1255-65, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18404158

ABSTRACT

Oxidized LDLs (oxLDLs) induce apoptosis, which contributes to the pathogenesis of atherosclerosis. The 150 kDa oxygen-regulated protein (ORP150), an endoplasmic reticulum (ER)-resident chaperone, is upregulated by hypoxia and prevents ischemia-induced cell death. The aim of this work was to investigate whether and how ORP150 can prevent apoptosis induced by oxLDLs in vascular cells. OxLDLs induced ORP150 expression in the ER of human microvascular endothelial cell line (HMEC-1). ORP150 expression was blocked by antioxidants, by the permeant calcium chelator BAPTA-AM, and by inhibitors of the inositol-1,4,5 trisphosphate (IP3) receptors, 2-aminoethyl diphenylborinate (2-APB) and xestospongin C. ORP150 silencing by siRNA-enhanced oxLDL-induced apoptosis, while forced ORP150 expression increased the resistance of cells via an inhibition of the oxLDL-induced calcium rise, and of subsequent calpain activation, cytochrome c release, caspase 3 activation and apoptosis. A similar protective effect was achieved by BAPTA-AM, 2-APB and xestospongin C. Altogether, these data indicate that (i)ORP150 inhibits oxLDL-induced apoptosis by blocking calcium signaling and subsequent apoptosis, (ii)calcium released from ER stores through IP3 channels is involved in the oxLDL-induced calcium rise and apoptosis, and is inhibited by ORP150. Finally, ORP150 is expressed in advanced atherosclerotic lesions, where it may locally participate to reduce the apoptotic effect of oxLDLs and the subsequent risk of plaque rupture.


Subject(s)
Apoptosis , Atherosclerosis/metabolism , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Endothelial Cells/metabolism , Lipoproteins, LDL/metabolism , Proteins/metabolism , Antioxidants/pharmacology , Boron Compounds/pharmacology , Calcium Signaling , Carotid Artery Diseases/metabolism , Cell Line , Chelating Agents/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , HSP70 Heat-Shock Proteins , Homeostasis , Humans , Macrocyclic Compounds/pharmacology , Oxazoles/pharmacology , RNA Interference
9.
Br J Pharmacol ; 153(1): 6-20, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17643134

ABSTRACT

Reactive carbonyl compounds (RCCs) formed during lipid peroxidation and sugar glycoxidation, namely Advanced lipid peroxidation end products (ALEs) and Advanced Glycation end products (AGEs), accumulate with ageing and oxidative stress-related diseases, such as atherosclerosis, diabetes or neurodegenerative diseases. RCCs induce the 'carbonyl stress' characterized by the formation of adducts and cross-links on proteins, which progressively leads to impaired protein function and damages in all tissues, and pathological consequences including cell dysfunction, inflammatory response and apoptosis. The prevention of carbonyl stress involves the use of free radical scavengers and antioxidants that prevent the generation of lipid peroxidation products, but are inefficient on pre-formed RCCs. Conversely, carbonyl scavengers prevent carbonyl stress by inhibiting the formation of protein cross-links. While a large variety of AGE inhibitors has been developed, only few carbonyl scavengers have been tested on ALE-mediated effects. This review summarizes the signalling properties of ALEs and ALE-precursors, their role in the pathogenesis of oxidative stress-associated diseases, and the different agents efficient in neutralizing ALEs effects in vitro and in vivo. The generation of drugs sharing both antioxidant and carbonyl scavenger properties represents a new therapeutic challenge in the treatment of carbonyl stress-associated diseases.


Subject(s)
Aldehydes/toxicity , Lipid Peroxidation , Proteins/metabolism , Aging/metabolism , Animals , Antioxidants/pharmacology , Cardiovascular Diseases/etiology , Cell Cycle/drug effects , Humans , Inflammation/etiology , Lipoproteins, LDL/metabolism , NF-kappa B/metabolism , Neoplasms/etiology , Neurodegenerative Diseases/etiology , Oxidation-Reduction , Signal Transduction
10.
Transplant Proc ; 39(8): 2624-6, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17954194

ABSTRACT

BACKGROUND: Hyperproliferation of smooth muscle cells (SMCs) plays a key role in allograft arteriosclerosis. This prompted us to investigate the effect of the novel immune modulator and synthetic sphingolipid FTY720 on apoptosis of SMCs. METHODS: Rabbit SMC cultures were treated with FTY720 and apoptosis and necrosis were detected by fluorescence microscopy. RESULTS: We investigated dose- and time-dependent effects of FTY720 and found that clinically relevant low doses of FTY720 (<1 micromol/L) did not induce apoptosis, whereas 10 micromol/L FTY720 induced apoptosis after 48 hours incubation. CONCLUSION: At doses of FTY720 used in clinics for treatment of renal allografts and multiple sclerosis. FTY720 did not induce SMC apoptosis.


Subject(s)
Apoptosis/drug effects , Immunosuppressive Agents/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Propylene Glycols/pharmacology , Sphingosine/analogs & derivatives , Animals , Fingolimod Hydrochloride , Models, Animal , Muscle, Smooth, Vascular/drug effects , Rabbits , Sphingosine/pharmacology
11.
Arterioscler Thromb Vasc Biol ; 24(10): 1830-5, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15308555

ABSTRACT

OBJECTIVE: Cholesterol accumulation in macrophages is known to alter macrophage biology. In this article we studied the impact of macrophage cholesterol loading on gene expression and identified a novel gene implicated in cell death. METHODS AND RESULTS: The regulated in development and DNA damage response 2 (REDD2) gene was strongly upregulated as THP-1 macrophages are converted to foam cells. These results were confirmed by Northern blot of RNA from human monocyte-derived macrophages (HMDM) treated with oxidized LDL (oxLDL). Human REDD2 shares 86% amino acid sequence identity with murine RTP801-like protein, which is 33% identical to RTP801, a hypoxia-inducible factor 1-responsive gene involved in apoptosis. Treatment of HMDM with desferrioxamine, a molecule that mimics the effect of hypoxia, increased expression of REDD2 in a concentration-dependent fashion. Transfection of U-937 and HMEC cells with a REDD2 expression vector increased the sensitivity of the cells for oxLDL-induced cytotoxicity, by inducing a shift from apoptosis toward necrosis. In contrast, suppression of mRNA expression using siRNA approach resulted in increased resistance to oxLDL treatment. CONCLUSIONS: We showed that stimulation of REDD2 expression in macrophages increases oxLDL-induced cell death, suggesting that REDD2 gene might play an important role in arterial pathology.


Subject(s)
Cell Death/physiology , Hypoxia/pathology , Lipoproteins, LDL/pharmacology , Macrophages/drug effects , Macrophages/physiology , Proteins/physiology , Up-Regulation/drug effects , Up-Regulation/physiology , Adaptor Proteins, Signal Transducing , Arteriosclerosis/genetics , Cell Line , Cell Line, Tumor , Cells, Cultured , DNA/genetics , Deferoxamine/pharmacology , Dose-Response Relationship, Drug , Endothelium, Vascular/chemistry , Endothelium, Vascular/metabolism , Foam Cells/physiology , Humans , Monocytes/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Transfection/methods , U937 Cells/chemistry , U937 Cells/metabolism
12.
Circ Res ; 89(11): 957-68, 2001 Nov 23.
Article in English | MEDLINE | ID: mdl-11717151

ABSTRACT

Sphingolipids have emerged as a new class of lipid mediators. In response to various extracellular stimuli, sphingolipid turnover can be stimulated in vascular cells and cardiac myocytes. Subsequent generation of sphingolipid molecules such as ceramide, sphingosine, and sphingosine-1-phosphate, is followed by regulation of ion fluxes and activation of various signaling pathways leading to smooth muscle cell proliferation, endothelial cell differentiation or apoptotic cell death, cell contraction, retraction, or migration. The importance of sphingolipids in cardiovascular signaling is illustrated by recent observations implicating them in physiological processes such as vasculogenesis as well as in frequent pathological conditions, including atherosclerosis and its complications.


Subject(s)
Heart Diseases/etiology , Myocardium/metabolism , Sphingolipids/physiology , Animals , Apoptosis , Cell Division , Coronary Artery Disease/etiology , Humans , Ion Transport , Myocardial Contraction , Myocardial Reperfusion Injury/etiology , Myocardium/cytology , Neovascularization, Physiologic , Radiation Injuries/etiology , Signal Transduction , Sphingolipids/chemistry
13.
Circulation ; 104(15): 1814-21, 2001 Oct 09.
Article in English | MEDLINE | ID: mdl-11591620

ABSTRACT

BACKGROUND: Mildly oxidized LDL (moxLDL) is thought to play a role in atherogenesis. MoxLDL induces derivatization of cell proteins and triggers a variety of intracellular signaling. We aimed to investigate whether moxLDL-induced protein derivatization may influence the activity of platelet-derived growth factor receptor beta (PDGFRbeta), a tyrosine kinase receptor of major importance in vascular biology and atherogenesis. METHODS AND RESULTS: In cultured rabbit arterial smooth muscle cells, moxLDL induces activation of the PDGFRbeta signaling pathway, as shown by PDGFRbeta tyrosine phosphorylation on Western blot and coimmunoprecipitation of SH2-containing proteins. The cellular events involved in the moxLDL-induced PDGFRbeta activation can be summarized as follows. Oxidized lipids from moxLDL trigger two phases of PDGFRbeta activation involving two separate mechanisms, as shown by experiments on cultured cells (in situ) and on immunopurified PDGFRbeta (in vitro): (1) the first phase may be mediated by 4-hydroxynonenal, which induces PDGFRbeta adduct formation and subsequent PDGFRbeta activation (antioxidant-insensitive step); (2) the second phase involves ceramide-mediated generation of H(2)O(2) (these steps being inhibited by tosylphenylalanylchloromethylketone, an inhibitor of ceramide formation, and by antioxidant BHT, exogenous catalase, or overexpressed human catalase). Because 4-hydroxynonenal-PDGFRbeta adducts are also detected in atherosclerotic aortas, it is suggested that this novel mechanism of moxLDL-induced PDGFRbeta activation may occur during atherogenesis. CONCLUSIONS: MoxLDL acts as a local autoparacrine mediator in the vascular wall, and PDGFRbeta acts as a sensor for both oxidized lipids and oxidative stress. This constitutes a novel mechanism of PDGFRbeta activation in atherosclerotic areas.


Subject(s)
Lipoproteins, LDL/metabolism , Macrolides , Muscle, Smooth, Vascular/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction/physiology , Aldehydes/metabolism , Aldehydes/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Antioxidants/pharmacology , Aorta/metabolism , Aorta/pathology , Arteriosclerosis/chemically induced , Arteriosclerosis/metabolism , Arteriosclerosis/pathology , Autocrine Communication , Cells, Cultured , Ceramides/metabolism , Chloroquine/pharmacology , Diet, Atherogenic , Enzyme Inhibitors/pharmacology , Humans , Hydrogen Peroxide/metabolism , Lipoproteins, LDL/pharmacology , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Oxidative Stress/physiology , Phosphorylation/drug effects , Rabbits , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sphingomyelins/metabolism , Tyrosine/metabolism
15.
Br J Pharmacol ; 132(8): 1777-88, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11309250

ABSTRACT

Oxidized low density lipoproteins (oxLDL) are thought to play a major role in atherosclerosis. OxLDL act in part through alteration of intracellular signalling pathways in cells of the vascular wall. We recently reported that the EGF receptor (EGFR) signalling pathway is activated by lipid peroxidation products (among them 4-hydroxynonenal, 4-HNE) contained in oxLDL. The use of phenolic antioxidants, such as trolox, alpha-tocopherol, caffeic acid and tyrphostins A-25, A-46 or A-1478, showed that the oxLDL-induced EGFR activation is constituted by two separate components, the first (early) one being antioxidant-insensitive, the second (late) being antioxidant-sensitive. 4-HNE derivatization of EGFR and EGFR activation induced by exogenous 4-HNE, suggest that the early (0.5 - 3 h) component of oxLDL-induced EGFR activation is mediated (at least in part) by 4-HNE (and possibly by other oxidized lipids). This early component is antioxidant-insensitive. The second component (4 - 5 h) of the oxLDL-induced EGFR activation is antioxidant-sensitive, since it is blocked by antioxidants such as trolox, caffeic acid or PDTC, which act by blocking the cellular oxidative stress (H(2)O(2) generation) evoked by oxLDL. Conversely, exogenous H(2)O(2) induced EGFR autophosphorylation (thus mimicking the second component) and was also inhibited by antioxidants. This effect is mediated in part through inhibition by oxidative stress of protein tyrosine phosphatases involved in EGFR dephosphorylation.


Subject(s)
Antioxidants/pharmacology , Caffeic Acids/pharmacology , Chromans/pharmacology , ErbB Receptors/drug effects , Lipoproteins, LDL/antagonists & inhibitors , Aldehydes/pharmacology , Biotransformation/drug effects , Blotting, Western , Cell Line , Cross-Linking Reagents/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Lipoproteins, LDL/isolation & purification , Lipoproteins, LDL/pharmacology , Oxidation-Reduction , Phosphorylation , Precipitin Tests , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism
16.
FASEB J ; 15(2): 403-15, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11156956

ABSTRACT

The vitamin E analog alpha-tocopheryl succinate (alpha-TOS) can induce apoptosis. We show that the proapoptotic activity of alpha-TOS in hematopoietic and cancer cell lines involves inhibition of protein kinase C (PKC), since phorbol myristyl acetate prevented alpha-TOS-triggered apoptosis. More selective effectors indicated that alpha-TOS reduced PKCalpha isotype activity by increasing protein phosphatase 2A (PP2A) activity. The role of PKCalpha inhibition in alpha-TOS-induced apoptosis was confirmed using antisense oligonucleotides or PKCalpha overexpression. Gain- or loss-of-function bcl-2 mutants implied modulation of bcl-2 activity by PKC/PP2A as a mitochondrial target of alpha-TOS-induced proapoptotic signals. Structural analogs revealed that alpha-tocopheryl and succinyl moieties are both required for maximizing these effects. In mice with colon cancer xenografts, alpha-TOS suppressed tumor growth by 80%. This epitomizes cancer cell killing by a pharmacologically relevant compound without known side effects.


Subject(s)
Apoptosis/drug effects , Colonic Neoplasms/pathology , Colorectal Neoplasms/pathology , Vitamin E/analogs & derivatives , Vitamin E/toxicity , Animals , Antibodies, Monoclonal/pharmacology , Caspase 3 , Caspases/metabolism , Colonic Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Genes, bcl-2 , Humans , Isoenzymes/antagonists & inhibitors , Jurkat Cells , Kinetics , Mice , Mice, Nude , Oligodeoxyribonucleotides, Antisense/pharmacology , Phosphoprotein Phosphatases/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-alpha , Protein Phosphatase 2 , Tetradecanoylphorbol Acetate/pharmacology , Tocopherols , Transplantation, Heterologous , Tumor Cells, Cultured , Vitamin E/therapeutic use , fas Receptor/immunology , fas Receptor/physiology
17.
Prog Lipid Res ; 39(3): 207-29, 2000 May.
Article in English | MEDLINE | ID: mdl-10799716

ABSTRACT

The atherosclerotic lesion most probably develops through a number of cellular events which implicate all vascular cell types and include synthesis of extracellular proteins, cell proliferation, differentiation and death. Sphingolipids and sphingolipid metabolizing enzymes may play important roles in atherogenesis, not only because of lipoprotein alterations but also by mediating a number of cellular events which are believed to be crucial in the development of the vascular lesions such as proliferation or cell death. Exogenous sphingolipids may mediate various biological effects such as apoptosis, mitogenesis or differentiation depending on the cell type. Moreover, several molecules present in the atherogenic lesion, such as oxidized LDL, growth factors or cytokines, which activate intracellular signaling pathways leading to vascular cell modifications, can stimulate sphingomyelin hydrolysis and generation of ceramide (and other metabolites as sphingosine-1-phosphate). Here we review the potential implication of the sphingomyelin/ceramide cycle in vascular cell signaling related to atherosclerosis, and more generally the role of sphingolipids in the events observed during the atherosclerotic process as cell differentiation, migration, adhesion, retraction, proliferation and death.


Subject(s)
Arteriosclerosis/metabolism , Blood Vessels/metabolism , Signal Transduction/physiology , Sphingolipids/metabolism , Arteriosclerosis/physiopathology , Blood Vessels/cytology , Humans , Lipoproteins/metabolism
18.
FASEB J ; 14(3): 532-42, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10698969

ABSTRACT

Oxidized low-density lipoproteins (oxLDL) play a role in the genesis of atherosclerosis. OxLDL are able to induce apoptosis of vascular cells, which is potentially involved in the formation of the necrotic center of atherosclerotic lesions, plaque rupture, and subsequent thrombotic events. Because oxLDL may induce structural modifications of cell protein and altered proteins may impair cell viability, the present work aimed to evaluate the extent of protein alterations, the degradation of modified proteins through the ubiquitin-proteasome system (a major degradative pathway for altered and oxidatively modified proteins) and their role during apoptosis induced by oxLDL. This paper reports the following: 1) oxLDL induce derivatization of cell proteins by 4-hydroxynonenal (4-HNE) and ubiquitination. 2) Toxic concentrations of oxLDL elicit a biphasic effect on proteasome activity. An early and transient activation of endogenous proteolysis is followed rapidly by a subsequent decay (resulting probably from the 26S proteasome inhibition) and followed later by the inhibition of the 20S proteasome (as assessed by inhibition of sLLVY-MCA hydrolysis). 3) Specific inhibitors of proteasome (lactacystin and proteasome inhibitor I) potentiated considerably the toxicity of oxLDL (nontoxic doses of oxLDL became severely toxic). The defect of the ubiquitination pathway (in temperature-sensitive mutants) also potentiated the toxicity of oxLDL. This suggests that the ubiquitin-proteasome pathway plays a role in the cellular defenses against oxLDL-induced toxicity. 4) Dinitrophenylhydrazine (DNPH), an aldehyde reagent, prevented both the oxLDL-induced derivatization of cell proteins and subsequent cytotoxicity. Altogether, the reported data suggest that both derivatization of cell proteins (by 4-HNE and other oxidized lipids) and inhibition of the proteasome pathway are involved in the mechanism of oxLDL-induced apoptosis.


Subject(s)
Apoptosis/drug effects , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Lipoproteins, LDL/toxicity , Multienzyme Complexes/metabolism , Ubiquitins/metabolism , 3T3 Cells , Aldehydes/pharmacology , Animals , Apolipoproteins B/pharmacology , Apoptosis/physiology , Cell Line , Cell Survival/drug effects , Clone Cells , Endothelium, Vascular , Humans , Kinetics , Lipoproteins, LDL/physiology , Mice , Models, Biological , Proteasome Endopeptidase Complex
19.
Circ Res ; 85(10): 892-9, 1999 Nov 12.
Article in English | MEDLINE | ID: mdl-10559135

ABSTRACT

Nonesterified fatty acids (NEFAs) are acutely liberated during lipolysis and are chronically elevated in pathological conditions, such as insulin resistance, hypertension, and obesity, which are known risk factors for atherosclerosis. The purpose of this study was to investigate the effect and mechanism of action of NEFAs on the epithelial growth factor (EGF) receptor (EGFR). In the ECV-304 endothelial cell line, unsaturated fatty acids triggered a time- and dose-dependent tyrosine phosphorylation of EGFR (polyunsaturated fatty acids [PUFAs] were the most active), whereas saturated FAs were inactive. Although less potent than PUFAs, oleic acid (OA) was used because it is prominent in the South European diet and is only slightly oxidizable (thus excluding oxidation derivatives). EGFR is activated by OA independent of any autocrine secretion of EGF or other related mediators. OA-induced EGFR autophosphorylation triggered EGFR signaling pathway activation (as assessed through coimmunoprecipitation of SH2 proteins such as SHC, GRB2, and SHP-2) and subsequent p42/p44 mitogen-activated protein kinase (as shown by the use of EGFR- deficient B82L and EGFR- transduced B82LK(+) cell lines). OA induced in vitro both autophosphorylation and activation of intrinsic tyrosine kinase of immunopurified EGFR, thus suggesting that EGFR is a primary target of OA. EGFR was also activated by mild surfactants, Tween-20 and Triton X-100, both in vitro (on immunopurified EGFR) and in intact living cells, thus indicating that EGFR is sensitive to amphiphilic molecules. These data suggest that EGFR is activated by OA and PUFAs, acts as a sensor for unsaturated fatty acids (and amphiphilic molecules), and is a potential transducer by which diet composition may influence vascular wall biology.


Subject(s)
ErbB Receptors/metabolism , Fatty Acids, Unsaturated/physiology , Autocrine Communication , Cell Line , Dimerization , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Enzyme Activation , ErbB Receptors/chemistry , ErbB Receptors/drug effects , Fatty Acids, Unsaturated/pharmacology , Humans , Mitogen-Activated Protein Kinases/metabolism , Oleic Acid/pharmacology , Phosphorylation/drug effects , Protein-Tyrosine Kinases/metabolism , Proteins/chemistry , Proteins/metabolism , Surface-Active Agents/pharmacology , Tyrosine/metabolism , src Homology Domains
20.
J Biol Chem ; 274(31): 21533-8, 1999 Jul 30.
Article in English | MEDLINE | ID: mdl-10419457

ABSTRACT

Oxidized LDL (oxLDL) have been implicated in diverse biological events leading to the development of atherosclerotic lesions. We previously demonstrated that the proliferation of cultured vascular smooth muscle cells (SMC) induced by oxLDL is preceded by an increase in neutral sphingomyelinase activity, sphingomyelin turnover to ceramide, and stimulation of mitogen-activated protein kinases (Augé, N., Escargueil-Blanc, I., Lajoie-Mazenc, I., Suc, I., Andrieu-Abadie, N., Pieraggi, M. T., Chatelut, M., Thiers, J. C., Jaffrézou, J. P., Laurent, G., Levade, T., Nègre-Salvayre, A., and Salvayre, R. (1998) J. Biol. Chem. 273, 12893-12900). Since ceramide can be converted to other bioactive metabolites, such as the well established mitogen sphingosine 1-phosphate (S1P), we investigated whether additional ceramide metabolites are involved in the oxLDL-induced SMC proliferation. We report here that incubation of SMC with oxLDL increased the activities of both acidic and alkaline ceramidases as well as sphingosine kinase, and elevated cellular sphingosine and S1P. Furthermore, the mitogenic effect of oxLDL was inhibited by D-erythro-2-(N-myristoylamino)-1-phenyl-1-propanol and N,N-dimethylsphingosine which are inhibitors of ceramidase and sphingosine kinase, respectively. These findings suggest that S1P is a key mediator of the mitogenic effect of oxLDL. In agreement with this conclusion, exogenous addition of sphingosine stimulated the proliferation of cultured SMC, and this effect was abrogated by dimethylsphingosine but not by fumonisin B1, an inhibitor of the acylation of sphingosine to ceramide. Exogenous S1P also promoted SMC proliferation. Altogether, these results strongly suggest that the mitogenic effect of oxLDL in SMC involves the combined activation of sphingomyelinase(s), ceramidase(s), and sphingosine kinase, resulting in the turnover of sphingomyelin to a number of sphingolipid metabolites, of which at least S1P is critical for mitogenesis.


Subject(s)
Amidohydrolases/metabolism , Cell Division/physiology , Lipoproteins, LDL/pharmacology , Lysophospholipids , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Sphingosine/analogs & derivatives , Animals , Cell Division/drug effects , Cells, Cultured , Ceramidases , Enzyme Activation , Femoral Artery/cytology , Femoral Artery/drug effects , Femoral Artery/physiology , Humans , Kinetics , Muscle, Smooth, Vascular/drug effects , Oxidation-Reduction , Rabbits , Sphingosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...