Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 60(4): 810-9, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12785728

ABSTRACT

Fenofibrate, a peroxisome proliferator-activated receptor (PPAR)-alpha activator, used as a normolipidemic agent, is thought to offer additional beneficial effects in atherosclerosis. Since angiogenesis is involved in plaque progression, hemorrhage, and instability, the main causes of ischemic events, this study was designed to evaluate the action of fenofibrate on angiogenesis. Our results show that fenofibrate (i) inhibits endothelial cell proliferation induced by angiogenic factors, followed at high concentrations by an increase in apoptosis, (ii) inhibits endothelial cell migration in a healing wound model, (iii) inhibits capillary tube formation in vitro, and (iv) inhibits angiogenesis in vivo. Concerning the mechanism of action, the inhibition of endothelial cell migration by fenofibrate can be explained by a disorganization of the actin cytoskeleton. At the molecular level, fenofibrate markedly decreased basic fibroblast growth factor-induced Akt activation and cyclooxygenase 2 gene expression. This inhibition of angiogenesis could participate in the beneficial effect of fenofibrate in atherosclerosis.


Subject(s)
Fenofibrate/pharmacology , Hypolipidemic Agents/pharmacology , Neovascularization, Pathologic/drug therapy , Protein Serine-Threonine Kinases , Actins/metabolism , Apoptosis/drug effects , Capillaries/drug effects , Capillaries/growth & development , Cell Cycle/drug effects , Cell Line , Cyclooxygenase 2 , Dermis/drug effects , Dermis/growth & development , Endothelium/drug effects , Endothelium/growth & development , Humans , In Vitro Techniques , Isoenzymes/biosynthesis , Isoenzymes/genetics , Membrane Proteins , Prostaglandin-Endoperoxide Synthases/biosynthesis , Prostaglandin-Endoperoxide Synthases/genetics , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins c-akt
2.
Carcinogenesis ; 22(8): 1139-48, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11470741

ABSTRACT

Cerivastatin is used in the treatment of hypercholesterolemia to inhibit 3-hydroxy 3-methylglutaryl coenzyme A reductase and thus prevent the synthesis of cholesterol precursors, such as farnesyl pyrophosphate (FPP) and geranylgeranyl pyrophosphate (GGPP), responsible, respectively, for translocation of Ras and Rho to the cell membrane, a step required for their cell signaling, leading to cell proliferation and migration. Recently, it has been suggested that non lipid-related effects of statins could play a beneficial role in cancer therapy. In this study, we have investigated the mechanisms by which statins inhibit cancer and the types of cancers which could benefit from this therapy. In MDA-MB-231 cells, an aggressive breast cancer cell line with spontaneous activation of Ras and NFkappaB and overexpression of RhoA, cerivastatin induced inhibition of both cell proliferation and invasion through Matrigel. This anti-proliferative effect was related to G(1)/S arrest due to an increase in p21(Waf1/Cip1). The anti-invasive effect was observed from 18 h and could be explained by RhoA delocalization from the cell membrane, resulting in disorganization of the actin fibers and disappearance of focal adhesion sites. The importance of RhoA inactivation in both these inhibitory effects was proved by their reversion by GGPP but not by FPP. Moreover, cerivastatin was also shown to induce inactivation of NFkappaB, in a RhoA inhibition-dependent manner, resulting in a decrease in urokinase and metalloproteinase-9 expression, two proteases involved in cell migration. The participation of Ras inactivation is considered a subsidiary mechanism for the effects of cerivastatin, as they were not rescued by FPP. Prolonged treatment of MDA-MB-231 cells with high doses of cerivastatin induced a loss of cell attachment. Interestingly, the effect of cerivastatin was considerably lower on poorly invasive MCF-7 cells. In conclusion, our results suggest that cerivastatin inhibits cell signaling pathways involved in the invasiveness and metastatic properties of highly invasive cancers.


Subject(s)
Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neoplasm Invasiveness , Neoplasm Metastasis , Pyridines/pharmacology , Signal Transduction/drug effects , Apoptosis/drug effects , Base Sequence , Cell Cycle/drug effects , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA Primers , Humans , I-kappa B Proteins/metabolism , In Vitro Techniques , Matrix Metalloproteinase 9/metabolism , NF-kappa B/antagonists & inhibitors , RNA, Messenger/genetics , Thromboplastin/metabolism , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/metabolism
3.
Circulation ; 103(19): 2328-31, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11352878

ABSTRACT

BACKGROUND: Abciximab plus aspirin improves the TIMI 3 flow rate of the infarct-related artery in patients treated with either percutaneous coronary intervention or thrombolysis. The present study investigated whether the reperfusion efficacy of abciximab relates to modifications of clot architecture in patients admitted for acute myocardial infarction (AMI). METHODS AND RESULTS: A total of 23 AMI patients in the Abciximab before Direct angioplasty and stenting in Myocardial Infarction Regarding Acute and Long term follow-up (ADMIRAL) trial received, in a double-blind fashion, either abciximab (n=13) or placebo (n=10) before primary stenting. Viscoelastic (G' in dyne/cm(2)) and morphological (mean platelet aggregate surface area [SAG] in micrometer(2)) indexes of ex vivo platelet-rich clots (PRC) were assessed in a double-blind fashion before and after the bolus administration of abciximab or placebo. G' and SAG reflect the mechanical and morphological impact of activated platelets on the PRC fibrin network, respectively. Abciximab administration reduced G' by 63% (P=0.0001) and SAG by 65% (P=0.0007), and no effect was seen in the placebo group. These abciximab-related changes increased fibrin exposure as a consequence of the platelet-aggregate surface reduction and may have improved endogenous fibrinolysis. These effects were identified in all patients, independent of previous heparin administration. CONCLUSIONS: Abciximab dramatically reduces platelet aggregate size and increases the fibrin accessibility of ex vivo PRC in AMI patients. These modifications could participate in the better coronary artery patency observed with abciximab.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Blood Coagulation/drug effects , Blood Platelets/metabolism , Immunoglobulin Fab Fragments/therapeutic use , Myocardial Infarction/therapy , Platelet Aggregation Inhibitors/therapeutic use , Abciximab , Aged , Blood Platelets/chemistry , Blood Platelets/drug effects , Double-Blind Method , Female , Humans , Male , Myocardial Infarction/blood , Platelet Aggregation/drug effects , Treatment Outcome , Viscosity/drug effects
4.
FEBS Lett ; 495(3): 159-66, 2001 Apr 27.
Article in English | MEDLINE | ID: mdl-11334884

ABSTRACT

Recent studies have suggested that inhibitors of 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (statins) can play a role in protection against vascular risk, which is independent of cholesterol reduction. It could act by inhibiting the synthesis of isoprenoids (farnesylpyrophosphate (FPP) and geranylgeranylpyrophosphate (GGPP)), which are respectively essential for membrane attachment and biological activity of GTPases Ras and RhoA. This study demonstrates that a statin (cerivastatin) inhibits angiogenesis. This effect was due to a decrease in endothelial cell locomotion which was reversed by GGPP. It was mainly related to delocalization of RhoA from cell membrane to cytoplasm, responsible for the disorganization of actin stress fibers. Furthermore, a decrease in MMP-2 secretion, involved in cell invasion, was also observed. This effect is rather due to Ras inhibition as it was reversed by FPP. This anti-angiogenic activity could explain the beneficial effect of statins on atherosclerosis and on cancer prevention as shown by clinical studies.


Subject(s)
Cell Movement/drug effects , Endothelium, Vascular/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neovascularization, Physiologic/drug effects , Pyridines/pharmacology , Actins/metabolism , Cell Line , Cell Membrane/metabolism , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/physiology , Fibroblast Growth Factor 2/pharmacology , Humans , Lymphokines/pharmacology , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/genetics , Oncostatin M , Peptides/pharmacology , Polyisoprenyl Phosphates/pharmacology , Polymers/metabolism , RNA, Messenger/biosynthesis , Sesquiterpenes , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Wound Healing , rhoA GTP-Binding Protein/metabolism
5.
Blood ; 97(7): 1982-9, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11264162

ABSTRACT

It is shown that the tetraspanin CD9 has a complex pattern of distribution in hematopoietic cells and is heterogeneously expressed on human bone marrow CD34(+) cells. CD34(high)CD38(low)Thy1(+) primitive progenitors are contained in the population with intermediate CD9 expression, thus suggesting that CD9 expression may precede CD38 appearance. Cell sorting shows that colony-forming unit (CFU)-GEMM and CFU-GM are present in high proportions in this fraction and in the fraction with the lowest CD9 expression. Cells with the highest level of CD9 are committed to the B-lymphoid or megakaryocytic (MK) lineages, as shown by the co-expression of either CD19 or CD41/GPIIb and by their strong potential to give rise to CFU-MK. In liquid cultures, CD9(high)CD41(neg) cells give rise to cells with high CD41 expression as early as 2 days, and this was delayed by at least 3 to 4 days for the CD9(mid) cells; few CD41(high) cells could be detected in the CD9(low) cell culture, even after 6 days. Antibody ligation of cell surface CD9 increased the number of human CFU-MK progenitors and reduced the production of CD41(+) megakaryocytic cells in liquid culture. This was associated with a decreased expression of MK differentiation antigens and with an alteration of the membrane structure of MK cells. Altogether these data show a precise regulation of CD9 during hematopoiesis and suggest a role for this molecule in megakaryocytic differentiation, possibly by participation in membrane remodeling. (Blood. 2001;97:1982-1989)


Subject(s)
Antigens, CD/physiology , Megakaryocytes/cytology , Membrane Glycoproteins , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation/biosynthesis , B-Lymphocytes/cytology , Cell Differentiation , Cell Lineage , Cell Membrane/immunology , Cell Membrane/ultrastructure , Cells, Cultured , Colony-Forming Units Assay , Flow Cytometry , Gene Expression Regulation, Developmental , Humans , Immunophenotyping , Megakaryocytes/metabolism , Megakaryocytes/ultrastructure , Microscopy, Electron , NAD+ Nucleosidase/biosynthesis , Platelet Glycoprotein GPIIb-IIIa Complex/biosynthesis , Tetraspanin 29
6.
Blood ; 97(4): 1043-9, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11159535

ABSTRACT

Following stress signals, the p53 tumor suppressor protein plays a critical role in regulation of cell proliferation, mainly through induction of growth arrest or apoptosis. Therefore, this protein needs to be strictly regulated and numerous studies have shown that the MDM2 protein is an essential element for p53 regulation in normal cells and, most importantly, that overexpression of MDM2 is responsible for p53 inactivation in various types of tumors. A previous study showed that this is the case in some Burkitt lymphoma (BL) cell lines, where enhanced translation of mdm2 messenger RNA results in overexpression of the protein that complexes and inactivates wild-type p53. To further investigate the role of the p53/MDM2 complex in these BL cells, as well as in other lymphoid cells that do not overexpress MDM2, this study used antisense oligodeoxynucleotides directed either against mdm2 or against p53. Results show that the mdm2 antisense oligodeoxynucleotide induces apoptosis of cells that express a high or low level of MDM2 protein, only if they contain wild-type p53. Moreover, apoptosis is independent of the accumulation of p53 following mdm2 antisense treatment. Finally, the p53 antisense oligodeoxynucleotide, which inhibits the expression of wild-type p53, also induces a decrease of the MDM2 level in cells, whether or not they overexpress this protein, and causes apoptosis of these cells. These results indicate that decreasing the MDM2 protein level by directly or indirectly targeting its biosynthesis is a potent tool for the induction of apoptosis.


Subject(s)
Apoptosis/physiology , Burkitt Lymphoma/pathology , Genes, p53 , Lymphocytes/cytology , Neoplasm Proteins/physiology , Neoplastic Stem Cells/cytology , Nuclear Proteins , Oligodeoxyribonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins/physiology , Tumor Suppressor Protein p53/physiology , Burkitt Lymphoma/genetics , Depression, Chemical , Gene Expression Regulation/drug effects , Humans , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2 , Signal Transduction , Tumor Cells, Cultured/cytology , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
7.
Arterioscler Thromb Vasc Biol ; 21(1): 142-8, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11145946

ABSTRACT

The glycoprotein IIb/IIIa receptor inhibitor abciximab has been shown to facilitate the rate and the extent of pharmacological thrombolysis with recombinant tissue plasminogen activator (rtPA) in patients with acute myocardial infarction. However, the underlying mechanisms remain not fully determined. We sought to demonstrate that this facilitating effect of abciximab could be related to its potential to modify the clot architecture and the clot physical properties. Compared with fibrin-rich clots, platelets dramatically modified the in vitro properties of the fibrin network, leading to a significant increase of the permeability (K(s)) and the viscoelasticity (G') indexes but also leading to the appearance of platelet aggregates (surface area [S.ag]). These modifications resulted in a 2.6-fold decrease of the fibrinolysis rate when rtPA (1 nmol/L) was added before the initiation of clotting. Adding aspirin (100 microgram/mL) or abciximab (0.068 micromol/L) before the clotting of platelet-rich clots (PRCs) lowered K(s) by 50% and 70%, respectively (P<0.01), G' by 41% and 66%, respectively (P<0.01), and S.ag by 32% and 61%, respectively (P<0.01). As a consequence, the lysis speed was increased by 21% with aspirin (P<0.01) and 45% with abciximab (P<0.01). However, unlike aspirin, permeation of preformed PRCs with abciximab (0.068 micromol/L) decreased G' (37%, P<0.01), K(s) (35%, P<0.001) and S.ag (25%, P=NS) and resulted in a 27% (P<0.01) increase of the lysis speed when abciximab and rtPA (0.2 micromol/L) were simultaneously permeated. This effect was found to be time dependent and was observed only with early permeation, starting within the first 10 minutes of clotting. These changes in the physical properties of the PRC architecture suggest that fibrin is removed from the platelet-fibrin aggregates and reexposed into the surrounding fibrin network, increasing rtPA access to fibrin and therefore the fibrinolysis rate. The superiority of abciximab over aspirin in accelerating fibrinolysis of forming and preformed PRCs is related to its ability to modulate the interactions of fibrinogen and fibrin with platelets. These findings provide new mechanistic information on reperfusion therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Fibrin/metabolism , Fibrinolysis/drug effects , Immunoglobulin Fab Fragments/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Abciximab , Aspirin/metabolism , Blood Platelets/drug effects , Blood Platelets/physiology , Blood Platelets/ultrastructure , Blood Viscosity/drug effects , Cell Membrane Permeability/drug effects , Elasticity/drug effects , Humans , Platelet Aggregation/drug effects , Platelet Function Tests
8.
Ophthalmic Res ; 33(1): 7-19, 2001.
Article in English | MEDLINE | ID: mdl-11114599

ABSTRACT

We investigated the regulation of sodium absorption by steroid hormones in embryologically diverse cells from the human eye. A cell extract from human corneal fibroblasts was positive for both the epithelial sodium channel (ENaC) and the mineralocorticoid receptor (MCR) as 82- to 85-kD and 102-kD bands, respectively, by the Western blot technique. In fluorescent, confocal and electron microscopy, the MCR was revealed as a nucleocytoplasmic protein, whereas the ENaC was almost exclusively membrane bound; both appeared aligned along actin filaments of corneal keratocytes, and both were widely colocalized in various cell types of human cornea in situ. Following reverse transcription and amplification of total RNA isolated from corneal fibroblasts, the ENaC and MCR genes in the PCR product were evident as predicted bands of 520 and 843 bp, respectively, whose sequence exhibited 100% identity with those from known human sources. The multiplication of corneal fibroblasts was influenced by both the MCR-specific antagonist RU 26752 and the natural hormone aldosterone, and these steroids also stimulated protein phosphorylation. In quantitative PCR, both the basal and aldosterone-induced levels of ENaC were diminished by the MCR-specific antagonist ZK 91587. Consequently, the ocular sodium channel appears to be regulated by steroid signalling in cells of diverse embryological origins, contrary to the existing notions where (a) this process would be limited exclusively to the epithelial cells and (b) ocular sodium transport would be regulated via the Na(+)-K(+)-ATPase in the basolateral membrane.


Subject(s)
Aldosterone/pharmacology , Epithelium, Corneal/metabolism , Fibroblasts/metabolism , Sodium Channels/metabolism , Base Sequence , DNA Primers/chemistry , Epithelium, Corneal/drug effects , Epithelium, Corneal/ultrastructure , Fibroblasts/drug effects , Fibroblasts/ultrastructure , Humans , Ion Transport/drug effects , Ion Transport/physiology , Microscopy, Confocal , Molecular Sequence Data , RNA/analysis , Receptors, Mineralocorticoid/drug effects , Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Signal Transduction/drug effects , Sodium Channels/drug effects
9.
Oncogene ; 19(45): 5153-62, 2000 Oct 26.
Article in English | MEDLINE | ID: mdl-11064452

ABSTRACT

There are two IL-15 isoforms and eight isoforms for the IL-15Ralpha chain whose biological role is poorly understood. Here, we have analysed the intracellular trafficking of IL-15 and IL-15Ralpha and tried to shed some light on their function(s). In IL-15/GFP CHO transfectants both IL-15 isoforms show nuclear localization. Two melanoma cell lines (MELP and MELREO) spontaneously expressing the IL-15 isoforms, display different intracellular trafficking of the IL-15/IL-15Ralpha complex. In MELP cells only IL-15Ralpha is detected inside the nucleus, whereas IL-15 and IL-15Ralpha assemble at the cell surface and are internalized. Moreover, the transducing molecule TRAF2 co-immunoprecipitates with IL-15Ralpha and may be deflected to TNFRI using anti-IL-15 blocking mAbs and TNF-alpha. By contrast, MELREO cells display IL-15Ralpha and IL-15 nuclear localization but only a partial co-localization of these molecules on the cell surface. In these cells, TRAF2 is strongly associated with IL-15Ralpha and cannot be deflected by any treatment. Since TRAF2 activates the transcription factor NF-kappaB, IL-15 through IL-15Ralpha, could have a role in the control of this pathway. Indeed, anti-IL-15 MaB inhibit the constitutive nuclear localization of NFkappaB and the phosphorylation of its inhibitor Ikappa-Balpha. Thus, IL-15Ralpha controls NF-kappaB activation, however differences in the intracellular trafficking of the IL-15 and/or IL-15Ralpha suggest a different biological role for this complex in MELP versus MELREO cells.


Subject(s)
Interleukin-15/metabolism , Melanoma/metabolism , Receptors, Interleukin-2/metabolism , Animals , CHO Cells , Cell Compartmentation , Cell Nucleus , Cricetinae , Green Fluorescent Proteins , Humans , Interleukin-15/genetics , Interleukin-15/isolation & purification , Luminescent Proteins/genetics , Luminescent Proteins/isolation & purification , Microscopy, Confocal , NF-kappa B/metabolism , Protein Binding , Protein Sorting Signals , Protein Subunits , Protein Transport , Proteins/isolation & purification , Receptors, Interleukin-15 , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/isolation & purification , Recombinant Fusion Proteins/metabolism , Signal Transduction , TNF Receptor-Associated Factor 2
10.
Thromb Haemost ; 84(4): 680-8, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11057870

ABSTRACT

It is now recognised that acute myocardial infarction results from the rupture of atherosclerotic plaques. Lymphocytes and macrophages, which infiltrate rupture sites, contribute to plaque degradation by expressing urokinase (u-PA) bound to cell membrane by urokinase receptor (u-PAR) and by secreting metalloproteinase MMP-9. We have previously demonstrated that the uptake of oxidised LDL (ox-LDL) by monocytes induces an increase of u-PA and u-PAR expression. The present study shows that the expression of u-PA and u-PAR induced by ox-LDL on monocyte surface is suppressed by cerivastatin (a synthetic inhibitor of HMG-CoA reductase, Bayer) from 2 nM. This leads to reduced plasmin generation and monocyte adhesion to vitronectin. Furthermore, higher concentrations of cerivastatin (50-100 nM) reduce the expression of u-PA and u-PAR on unstimulated monocytes. It also inhibits MMP-9 secretion but has no effect on TIMP-1 secretion, suggesting that the decrease in MMP-9 has a real protective effect on plaque stabilisation. The inhibitory effect of cerivastatin on u-PA expression and MMP-9 secretion can be explained by the inhibition of NF-kappa B translocation into the nucleus, as shown by immunofluorescence. As farnesyl-pyrophosphate reverses the effect of cerivastatin, it is postulated that these effects could also be due to the inhibition of Ras prenylation. This was confirmed by confocal microscopy, which shows the Ras delocalisation from the monocyte membrane. The cerivastatin-induced effects on monocyte functions could explain, at least in part, the protective effect of this drug against atherothrombotic events.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Matrix Metalloproteinase 9/metabolism , Monocytes/drug effects , Monocytes/metabolism , Pyridines/pharmacology , Receptors, Cell Surface/biosynthesis , Urokinase-Type Plasminogen Activator/biosynthesis , Arteriosclerosis/drug therapy , Biological Transport/drug effects , Cells, Cultured , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Microscopy, Confocal , Monocytes/ultrastructure , Pyridines/therapeutic use , Receptors, Urokinase Plasminogen Activator , Thrombosis/drug therapy
11.
Viral Immunol ; 13(3): 313-27, 2000.
Article in English | MEDLINE | ID: mdl-11016596

ABSTRACT

Inbred athymic nude mice (BALB/c) were injected subcutaneously with the wild-type murine gammaherpesvirus 72 (MHV-72), which has been shown to induce the infectious mononucleosis (IM)-like syndrome in immunocompetent mice. The mice were also injected with UV-irradiated MHV-72. We studied the pattern of acute and chronic infection in the blood cells of the nude mice and detected viral DNA sequences in the infected leukocytes by polymerase chain reaction (PCR) technique up to when the animal died, close to 1 month postinfection. Using the UV-irradiated virus that induces an increase in mouse survival time, the viral sequences were present in the blood up to 3 months postinfection, then disappeared. We detected atypical lymphocytes in the blood of mice infected with both wt and UV-irradiated viruses. These atypical cells were similar in shape to those present in the blood of patients with IM induced by Epstein-Barr virus (EBV). Via Unscheduled DNA Synthesis (UDS), DNA synthesis was demonstrated in the atypical cells whose phenotype is identical to that of B cells, as shown with a panel of monoclonal antibodies. By double immunofluorescence staining, using an hyperimmune anti-MHV-72 serum and an anti-IgG + IgM + IgA monoclonal antibody, we demonstrated that these atypical B cells express some viral antigens. Contrary to the immunocompetent mice, the nude mice did not develop splenomegaly after infection with wt virus, probably due to the lack of T cell subsets. However, we observed an increase of nude mice B cells in the spleen. The nude mice died 1 month postinfection showing a high frequency (40%) of atypical lymphoblast-like B-cells in the blood; the increase in natural killer (NK) cell number was not detected after infection. Such findings suggest that NK cells probably did not play an important role in immune response to the MHV infection in nude mice. Finally, this mouse model could play an important role in antigammaherpesviral therapy of immunocompromised patients.


Subject(s)
B-Lymphocytes/immunology , Gammaherpesvirinae/immunology , Herpesviridae Infections/immunology , Immunophenotyping , Spleen/immunology , Animals , B-Lymphocytes/classification , Cell Line , DNA/biosynthesis , DNA, Viral/biosynthesis , DNA, Viral/blood , Female , Gammaherpesvirinae/genetics , Gammaherpesvirinae/isolation & purification , Gammaherpesvirinae/radiation effects , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Spleen/cytology , Ultraviolet Rays
12.
Leukemia ; 14(6): 1097-104, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10865975

ABSTRACT

We studied the expression of the mineralocorticoid receptor (MCR), and of the amiloride-sensitive sodium channel (ASSC) regulated by the MCR, in human leukemic cell lines. Cell extracts from TF1 (proerythroblastic), HEL (human erythroblastic leukemia) and U937 (myeloblastic) cell line were positive for the ASSC, as a 82 kDa band in Western blots developed with the aid of a polyclonal antibody raised against the peptide QGLGKGDKREEQGL, corresponding to the region 44-58 of the alpha subunit of the epithelial sodium channel (ENaC) cloned from rat colon, linked to KLH. The polyclonal antibody against the MCR revealed a single band of about 102 kDa in extracts from HEL and TF1 cells. The immunofluorescent labelling of the MCR in all cell lines showed a nucleocytoplasmic localization of the receptor but the ASSC was exclusively membrane-bound and these results were confirmed by confocal microscopy. The expression of the MCR in the HEL cells was evident as a predicted band of 843 bp (234 amino acids) in electrophoresis of the PCR product obtained after total RNA had been reverse transcribed and then amplified using the primers 5'-AGGCTACCACAGTCTCCCTG-3' and 5'-GCAGTGTAAAATCTCCAGTC-3' (sense and antisense, respectively). The ENaC was similarly evident with the aid of the primers 5'-CTGCCmATG GATGATGGT-3' (sense) and 5'-GTTCAGCTCGAAGAAGA-3' (antisense) as a predicted band of 520 bp. In both cases, 100% identity was observed between the sequences of the PCR products compared to those from known human sources. The multiplication of the HEL cells was influenced by antagonists (RU 26752, ZK 91587) targeted for specificity to the MCR and this was selectively reversed by the natural hormone aldosterone. These steroids also provoked chromatin condensation in the HEL population. These permit new and novel possibilities to understand the pathobiology of human leukemia and to delineate sodium-water homeostasis in nonepithelial cells.


Subject(s)
Leukemia/metabolism , Receptors, Mineralocorticoid/metabolism , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , DNA Primers , Humans , Immunohistochemistry , Leukemia/pathology , Microscopy, Confocal , Polymerase Chain Reaction , Rats , Tumor Cells, Cultured
13.
Oncogene ; 19(23): 2758-66, 2000 May 25.
Article in English | MEDLINE | ID: mdl-10851076

ABSTRACT

IL-15 and SCF fail to induce NK differentiation and proliferation of CD34+ hematopoietic progenitors from chronic myeloid leukemia patients in contrast to normal stem cells although, both normal and leukemic CD34+ cells display comparable expression of c-kit or IL-15 receptor subunits. Interestingly, confocal microscopy analysis revealed that leukemic and most normal CD34+ cells produce and secrete IL-15, as shown by its trafficking through the Golgi apparatus and early endosomes. However, only leukemic progenitors express the membrane bound IL-15. Colocalization and internalization of IL-15Rbeta/gammac and IL-15Ralpha/gammac complexes indicated that IL-15 was specifically uptaken by leukemic progenitors. We also demonstrated that in both normal and leukemic progenitors, the signaling kinase Jak3 is constitutively pre-associated with the gammac chain. Anti-IL-15 neutralizing mAb treatment resulted in down-regulation of gammac chain and disruption of gammac/Jak3 interaction in normal but had no effect in leukemic progenitors. Our results suggest the existence in both normal and leukemic CD34+ cells of a constitutive production of a bioactive IL-15 that does not lead to NK differentiation and further indicate that membrane bound IL-15 and constitutive activation of gammac are hallmarks of leukemic progenitors. Oncogene (2000).


Subject(s)
Antigens, CD34/metabolism , Killer Cells, Natural/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Lymphocyte Subsets/pathology , Cell Differentiation , Cell Division , Cell Line , Humans , Interleukin-15/metabolism , Interleukin-15/pharmacology , Killer Cells, Natural/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Lymphocyte Subsets/physiology , Microscopy, Confocal , Receptors, Interleukin-15 , Receptors, Interleukin-2/metabolism , Stem Cell Factor/metabolism , Stem Cell Factor/pharmacology , Tumor Cells, Cultured
14.
Biochemistry ; 39(18): 5332-40, 2000 May 09.
Article in English | MEDLINE | ID: mdl-10820003

ABSTRACT

gp17, a secretory CD4-binding factor isolated from the human seminal plasma, is identical to the gross cystic disease fluid protein-15, a specific marker for primary and metastatic breast tumors. We previously demonstrated that gp17 binds to CD4 with high affinity and strongly inhibits T lymphocyte apoptosis induced by sequential cross-linking of CD4 and T cell receptor (TCR). To further characterize the gp17/CD4 interaction and map the gp17 binding site, we produced a secreted form of recombinant gp17 fused to human IgG1 Fc, gp17-Ig. We showed that gp17-Ig exhibits a binding affinity for CD4 similar to that of native gp17. As no information about gp17 structure is presently available, 99 overlapping gp17 peptides were synthesized by the Spot method, which allowed the mapping of two CD4 binding regions. Alanine scanning of CD4-reactive peptides identified critical residues, selected for site-directed mutagenesis. Nine gp17-Ig mutants were generated and characterized. Three residues within the carboxy-terminal region were identified as the major binding domain to CD4. The Spot method combined with mutagenesis represents a refined approach to distinguish the contact residues from the ones contributing to the conformation of the CD4-binding domain.


Subject(s)
Apolipoproteins , Breast Neoplasms/metabolism , Carrier Proteins/chemistry , Glycoproteins/chemistry , Membrane Transport Proteins , Seminal Vesicles/metabolism , Amino Acid Sequence , Animals , Apolipoproteins D , CD4 Antigens/metabolism , COS Cells , Carrier Proteins/genetics , Fluorescent Antibody Technique , Glycoproteins/genetics , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Male , Molecular Sequence Data , Mutation , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Peptide Fragments/metabolism , Protein Binding/genetics , Recombinant Fusion Proteins , Transfection
15.
Thromb Haemost ; 82(5): 1482-9, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10595642

ABSTRACT

Porosity, viscoelasticity and morphological properties of plasma fibrin from 16 nephrotic patients and 16 healthy volunteers were compared. Nephrotic patients were characterized by formation of tight and rigid plasma fibrin gels which resulted in a slower rate of fibrin lysis studied either under pressure-driven permeation or diffusional transport of fibrinolytic agents. These latter findings indicated that both abnormal fibrin network conformation and abnormal fibrin fiber structure were involved in hypofibrinolysis. Albumin supplementation up to 40 mg/ml partially restored normal fibrin architecture and increased the rate of fibrinolysis in these patients. Multiparametric analysis showed that nephrotic patients were mainly characterized by a low plasma albumin level (R = -0.85), a low albumin to fibrinogen ratio (R = -0.89) and a high resistance to lysis (R = -0.82). High triglycerides level was the only plasma modification related to the slower fibrin lysis rate (R = -0.54). High fibrin rigidity (G') was the only fibrin parameter simultaneously related to the nephrotic state (R = 0.75) and the lysis resistance (R = -0.71). After eliminating the effects of age, albumin and fibrinogen levels, low fibrin porosity (Ks) and low fiber mass-length ratio (mu) were the main features of the nephrotic state. These findings are discussed in relation to both the pathophysiology of thrombotic complications in nephrotic syndrome and their pharmacological prevention.


Subject(s)
Fibrin/chemistry , Fibrinolysis , Nephrotic Syndrome/blood , Thrombophilia/etiology , Thrombosis , Adolescent , Adult , Aged , Elasticity , Female , Fibrinolytic Agents/pharmacology , Gels , Humans , Lipids/blood , Male , Microscopy, Confocal , Middle Aged , Nephrotic Syndrome/complications , Porosity , Serum Albumin/chemistry , Serum Albumin/deficiency , Serum Albumin/pharmacology , Viscosity
16.
Nucleosides Nucleotides ; 18(6-7): 1721-2, 1999.
Article in English | MEDLINE | ID: mdl-10474256

ABSTRACT

We studied the uptake and intracellular distribution of an FITC labelled phosphodiester oligodeoxynucleotide (ODN) vectorized by a dendrimeric structure in cell culture.


Subject(s)
Oligonucleotides/metabolism , Polyamines/chemistry , 3T3 Cells , Animals , Fluorescein-5-isothiocyanate , HeLa Cells , Humans , Mice , Microscopy, Confocal
17.
Ann N Y Acad Sci ; 876: 236-45, 1999 Jun 22.
Article in English | MEDLINE | ID: mdl-10415615

ABSTRACT

IL-15 is an immunostimulatory cytokine sharing with IL-2 the IL-2R beta gamma complex. In vivo, IL-15 detection in synovial fluids has been associated with the development of rheumatoid arthritis. A debate exists as to whether IL-15 has the potential to be secreted in meaningful amounts or to act as a pericellular cytokine. Our data show (1) the presence of two IL-15 isoforms displaying signal peptides of different length and the capacity to be secreted restricted to the isoform bearing the longer one; (2) in cells expressing the two isoforms, the existence of different nuclear localization and intracellular trafficking of IL-15 and IL-15R alpha; and (3) an intercellular microcirculation of IL-15, not detectable with ELISA kits, but displaying a role as an anti-apoptotic factor able to induce the deflection of the TNFR associated factor 2 (TRAF) to IL-15R alpha. Our data point to a juxtacrine mechanism of action of IL-15 and suggest a role for IL-15/IL-15R alpha in the regulation of apoptosis.


Subject(s)
Apoptosis/physiology , Interleukin-15/metabolism , Intracellular Membranes/metabolism , Receptors, Interleukin-2/metabolism , Animals , CHO Cells , Cricetinae , Humans , Microscopy, Confocal , Protein Isoforms/metabolism , Proteins/metabolism , Receptors, Interleukin-15 , TNF Receptor-Associated Factor 2 , Tissue Distribution/physiology , Tumor Cells, Cultured
18.
Biochem Pharmacol ; 58(1): 95-107, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10403523

ABSTRACT

Factors limiting the use of antisense phosphodiester oligodeoxynucleotides (ODNs) as therapeutic agents are inefficient cellular uptake and intracellular transport to RNA target. To overcome these obstacles, ODN carriers have been developed, but the intracellular fate of ODNs is controversial and strongly depends on the means of vectorization. Polyamidoamine dendrimers are non-linear polycationic cascade polymers that are able to bind ODNs electrostatically. These complexes have been demonstrated to protect phosphodiester ODNs from nuclease degradation and also to increase their cellular uptake and pharmacological effectiveness. We studied the intracellular distribution of a fluorescein isothiocyanate-labeled ODN vectorized by a dendrimer vector and found that intracellular ODN distribution was dependent on the phase of the cell cycle, with a nuclear localization predominantly in the G2/M phase. In addition, in order to evaluate the relevance of ODN vectors in enhancing the inhibition of the targeted genes' expression, we developed a rapid screening system which measures the transient expression of two reporter genes, one used as target, the other as control and vice versa. This system was validated through investigating the effect of the dendrimer vector on ODN biological activity. Antisense sequence-specific inhibition of more than 70% of one reporter gene was obtained with a chimeric ODN containing four phosphorothioate groups, two at each end.


Subject(s)
Cell Cycle/physiology , Oligonucleotides, Antisense/metabolism , Oligonucleotides, Antisense/pharmacology , 3T3 Cells , Animals , Cell Culture Techniques , Flow Cytometry/methods , Gene Expression/drug effects , Genes, Reporter , Genetic Vectors/metabolism , Genetic Vectors/pharmacology , Green Fluorescent Proteins , HeLa Cells , Humans , Kinetics , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Mice , Microscopy, Confocal , Oligonucleotides, Antisense/genetics , RNA, Messenger/biosynthesis
19.
Int J Cancer ; 81(4): 645-9, 1999 May 17.
Article in English | MEDLINE | ID: mdl-10225457

ABSTRACT

Latent membrane protein 1 (LMP1) is an Epstein-Barr virus (EBV) protein expressed in EBV-transformed B lymphocytes and in approximately 50% of nasopharyngeal carcinomas (NPCs). LMP1 signaling involves several cellular signaling intermediates, especially TNF receptor-associated factors (TRAFs). We have shown previously that LMP1 is highly concentrated in a cell fraction called glycosphingolipid-rich membrane complexes (GSL complexes). We report here that parallel accumulation of LMP1 and TRAF3, but not TRAF1 or TRADD, was observed in GSL complexes from lymphoblastoid and LMP1-positive NPC cells. In contrast, TRAF3 was not concentrated in GSL complexes from LMP1-negative cells. Binding of LMP1 and TRAF3 in GSL complexes was demonstrated in lymphoblastoid and NPC cells, by co-immunoprecipitation with both anti-LMP1 and anti-TRAF3 antibodies.


Subject(s)
Burkitt Lymphoma/metabolism , Glycosphingolipids/metabolism , Nasopharyngeal Neoplasms/metabolism , Proteins/metabolism , Viral Matrix Proteins/metabolism , Animals , Biopsy , Burkitt Lymphoma/chemistry , Burkitt Lymphoma/pathology , Centrifugation, Density Gradient , Glycosphingolipids/isolation & purification , Herpesvirus 4, Human/genetics , Humans , Mice , Mice, Nude , Nasopharyngeal Neoplasms/chemistry , Nasopharyngeal Neoplasms/pathology , Proteins/genetics , Proteins/isolation & purification , Receptors, Tumor Necrosis Factor/physiology , TNF Receptor-Associated Factor 3 , Transplantation, Heterologous , Tumor Cells, Cultured , Viral Matrix Proteins/genetics , Viral Matrix Proteins/isolation & purification
20.
Eur J Immunol ; 29(4): 1265-74, 1999 04.
Article in English | MEDLINE | ID: mdl-10229094

ABSTRACT

To analyze the intracellular trafficking of two IL-15 isoforms bearing 48- or 21-amino acid leader peptides (L), we have generated cDNA encoding the two proteins fused at the C terminus with green fluorescent protein (GFP). Confocal microscopy analyses showed that, when transfected in CHO cells, 48L IL-15/GFP was localized in the Golgi apparatus and in early endosomes, while 21L IL-15/GFP was detectable only in the cytosol. The presence of 48L IL-15/GFP in endosomes was confirmed by enzyme-linked immunosorbent assay on endosome-enriched subcellular fractions. Exogenous IL-15 was bound and taken up in endosomes by untransfected CHO cells, indicating that endosomal localization was, at least in part, related to a receptor-mediated uptake. The 48L IL-15/GFP fusion protein was efficiently secreted by COS-7 or CHO cell transfectants, while IL-15 secretion was less efficient in transfectants expressing 21L IL-15/GFP or untagged 48L or 21L IL-15. Treatment with brefeldin A or with inhibitors of N-linked glycosylation further indicated that the 48L IL-15/GFP is secreted through the endoplasmic reticulum/Golgi pathway. Our data suggest a different trafficking of the two IL-15 isoforms and multiple mechanisms controlling IL-15 secretion.


Subject(s)
Endosomes/metabolism , Interleukin-15/metabolism , Amino Acid Sequence , Animals , Biological Transport , CHO Cells , COS Cells , Cricetinae , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Green Fluorescent Proteins , Humans , Interleukin-15/analysis , Interleukin-15/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...