387 results on '"Caen JP"'
Search Results
2. Pharmacology of Platelet-Suppressive Agents
- Author
-
Wautier Jl and Caen Jp
- Subjects
Blood Platelets ,Platelet Aggregation ,Anti-Inflammatory Agents ,Arachidonic Acids ,Pharmacology ,Platelet Adhesiveness ,Text mining ,Prostaglandins, Synthetic ,Humans ,Medicine ,Platelet ,Clofibrate ,Aspirin ,Heparin ,business.industry ,Prostaglandins E ,Cell Membrane ,Imidazoles ,Dipyridamole ,Hematology ,Epoprostenol ,Adenosine Diphosphate ,Calcium ,Cardiology and Cardiovascular Medicine ,business ,Halofenate - Published
- 2008
3. Les glycoprotéines plaquettaires : vingt ans après
- Author
-
Nurden, AT, primary and Caen, JP, additional
- Published
- 1995
- Full Text
- View/download PDF
4. Ultrastructural demonstration of CD36 in the alpha-granule membrane of human platelets and megakaryocytes
- Author
-
Berger, G, primary, Caen, JP, additional, Berndt, MC, additional, and Cramer, EM, additional
- Published
- 1993
- Full Text
- View/download PDF
5. Defective adhesion of blood platelets to vascular microfibrils in the Bernard-Soulier syndrome
- Author
-
Fauvel-Lafeve, F, primary, Tabaka, V, additional, Caen, JP, additional, and Legrand, YJ, additional
- Published
- 1993
- Full Text
- View/download PDF
6. Differential redistribution of platelet glycoproteins Ib and IIb-IIIa after plasmin stimulation [published erratum appears in Blood 1991 Jul 15;78(2):545]
- Author
-
Cramer, EM, primary, Lu, H, additional, Caen, JP, additional, Soria, C, additional, Berndt, MC, additional, and Tenza, D, additional
- Published
- 1991
- Full Text
- View/download PDF
7. Des peptides à activité antithrombotique dans les protéines du lait : un lien entre thérapeutique et diététique
- Author
-
Caen, JP, primary, Sollier, CBD, additional, Mazoyer, E, additional, Drouet, L, additional, Jolles, P, additional, and Fiat, AM, additional
- Published
- 1991
- Full Text
- View/download PDF
8. Glanzmann's thrombasthenia: the spectrum of clinical disease
- Author
-
George, JN, primary, Caen, JP, additional, and Nurden, AT, additional
- Published
- 1990
- Full Text
- View/download PDF
9. Alpha-granule pool of glycoprotein IIb-IIIa in normal and pathologic platelets and megakaryocytes
- Author
-
Cramer, EM, primary, Savidge, GF, additional, Vainchenker, W, additional, Berndt, MC, additional, Pidard, D, additional, Caen, JP, additional, Masse, JM, additional, and Breton-Gorius, J, additional
- Published
- 1990
- Full Text
- View/download PDF
10. Role of Surface Glycoproteins in Human Platelet Function
- Author
-
Nurden At and Caen Jp
- Subjects
chemistry.chemical_classification ,biology ,Hematology ,Platelet membrane glycoprotein ,medicine.disease ,Bernard–Soulier syndrome ,Cell biology ,Sialic acid ,chemistry.chemical_compound ,Membrane ,chemistry ,Thrombasthenia ,Von Willebrand factor ,medicine ,biology.protein ,Platelet ,Glycoprotein - Abstract
SummaryGlycoproteins present at the external surface of cells probably play specific roles in cellular function. Increasing evidence suggests that the glycoproteins span the plasma membrane with the bulk of the bound carbohydrate asymmetrically distributed on the outer surface. Micellar association of glycoproteins in membranes leads to pore formation and functional roles in transport through the membrane, while surface glycoproteins have been shown to be enzymes, to determine cell specificity and contribute to the cell surface charge. The platelet plasma membrane contains 3 major glycoproteins, glycoproteins I, II and III as characterized in order of their decreasing molecular weight. Glycoprotein I appears to have the highest sialic acid content and to give rise to a platelet specific acidic macroglycopeptide on trypsin digestion. Specific glycoprotein abnormalities in the platelets of patients with Glanzmann’s thrombasthenia suggest that the glycoproteins play a role in the mechanism of platelet aggregation. A much reduced content of glycoprotein I in the platelets of 2 patients with the Bernard Soulier syndrome may be associated with their defective adhesion to subendothelium and indicates a possible relationship on the platelet surface with the von Willebrand factor protein. Preliminary evidence suggests that in common with other plasma membranes the platelet membrane has a fluid structure and that the organization of the glycoproteins on the platelet surface is extremely sensitive to stimuli and susceptible to change.
- Published
- 1976
11. Platelet Aggregation in Mammalians (Human, Rat, Rabbit, Guinea-Pig, Horse, Dog) A Comparative Study
- Author
-
Sinakos Z and Caen Jp
- Subjects
Andrology ,Guinea pig ,Platelet aggregation ,Chemistry ,Adenine nucleotide ,Horse ,Platelet ,Rabbit (nuclear engineering) ,Hematology - Abstract
SummaryA comparative study of mammalian (human, rat, rabbit, guinea-pig, horse, dog) platelet aggregation was done using phase contrast microscopy and photometric method and gave us the following results :1. The animal platelets do not spread spontaneously onto glass in contrast to human ones (in their own plasma).2. They do not aggregate in presence of adrenalin, noradrenalin, 5-HT but these amines increase the sensitivity of the platelets towards ADP.3. Although all the platelets aggregate in presence of ADP, bovine thrombin, collagen or thimerosal, there exists some quantitative differences from one species to another.4. Adenosine and AMP are inhibitors of ADP-induced aggregation for human, rabbit and dog platelets and not for guinea-pig and horse. As for the rat, if adenosine is not inhibitor, AMP can be inhibitor if used at strong doses.5. There exist great differences in the ability of the various platelet-poor plasmas studied to inactivate the aggregating property of ADP, as for instance the weakest being the human one and the strongest the rat one.6. The authors insist on the importance of ADP-inhibition by AMP and adenosine, and the ADP-inactivation by the plasmas for the mammalian platelet aggregation and almost disaggregation.
- Published
- 1967
12. Specific protein and glycoprotein deficiencies in platelets isolated from two patients with the gray platelet syndrome
- Author
-
Nurden, AT, Kunicki, TJ, Dupuis, D, Soria, C, and Caen, JP
- Abstract
The gray platelet syndrome is a rare inherited platelet disorder characterized by the absence of alp ha-granules as observed by electron microscopy. Analysis of the glycoprotein composition of the platelets of 2 such patients by SDS-polyacrylamide gel electrophoresis (SDS-PAGE) revealed decreased or absent staining for carbohydrate of several high molecular weight glycoproteins. The major periodic acid Schiff (PAS) staining membrane glycoproteins were normally detected and were normally labeled with 125I during lactoperoxidase-catalyzed iodination. Analysis of the protein composition of gray platelets by single or two- dimensional SDS-PAGE followed by Coomassie blue staining revealed an apparent absence of GP Ig (thrombospondin), markedly reduced platelet fibrinogen and albumin concentrations, and severely reduced levels of 2 low molecular weight polypeptides exhibiting identical rates of migration on SDS-PAGE as platelet factor 4 and beta-thromboglobulin. SDS-PAGE profiles similar to those of the gray platelets were observed with normal human platelets that had undergone the release reaction induced by thrombin. Analysis of gray platelet proteins by crossed immunoelectrophoresis using a rabbit anti-human platelet antibody preparation and rocket immunoelectrophoresis using monospecific antisera confirmed the above findings and showed additional severe deficiencies of factor VIIIR:Ag and cold-insoluble globulin. In contrast, factor XIII (subunit A), a cytoplasmic protein, was normally detected. Our studies provide further evidence that circulating gray platelets specifically lack, or have markedly decreased concentrations of, the alpha-granule proteins.
- Published
- 1982
- Full Text
- View/download PDF
13. Characterization of human platelet glycoprotein antigens giving rise to individual immunoprecipitates in crossed-immunoelectrophoresis
- Author
-
Kunicki, TJ, Nurden, AT, Pidard, D, Russell, NR, and Caen, JP
- Abstract
Washed human platelets were labeled with 125I by the lactoperoxidase- catalyzed method and solubilized in 1% Triton X-100. The soluble proteins were analyzed by crossed-immunoelectrophoresis in 1% agarose, employing a rabbit antibody raised against whole human platelets. Analysis of autoradiograms developed from dried agarose gels led to the establishment of a normal reference pattern that was consistent for platelets obtained from more than 50 normal individuals. Six platelet membrane glycoprotein antigens contained in four distinguishable precipitates were identified. Each identification was based on direct sodium dodecyl sulfate-polyacrylamide gel electrophoretic analysis of 125I-antigens contained in individually excised precipitates. These platelet antigens include major membrane glycoproteins previously designated la, lb, lla, llb, llla, and lllb. Glycoproteins llb and llla were shown to be contained in a single immunoprecipitate, while glycoproteins la and lla were routinely detected in a single different immunoprecipitate. Analysis of soluble proteins from platelets of five patients with Glanzmann's thrombasthenia demonstrated either a complete absence or a marked reduction of only one radiolabeled precipitate, that containing membrane glycoproteins llb and llla. Platelet samples from two patients with Bernard-Soulier syndrome were devoid of a different precipitate, that containing membrane glycoprotein lb.
- Published
- 1981
- Full Text
- View/download PDF
14. Evaluation of the inhibition by heparin and hirudin of coagulation activation during r-tPA-induced thrombolysis
- Author
-
Mirshahi, M, Soria, J, Soria, C, Faivre, R, Lu, H, Courtney, M, Roitsch, C, Tripier, D, and Caen, JP
- Abstract
Thrombin bound to a fibrin clot remains active and poorly accessible to heparin-AT III complex. During fibrinolysis, thrombin is released as thrombin-FDP complex and is inactivated by heparin-AT III. However, as successive fibrin layers are removed, inaccessible molecules of thrombin are exposed at the surface of the residual clot, possibly contributing to the occurrence during thrombolytic therapy of coagulation that is poorly controlled by heparin. We have investigated the accessibility of fibrin-bound thrombin to hirudin. The results clearly show that two recombinant hirudin variants neutralize thrombin both in solution and fibrin bound. Furthermore, we have found that in in vitro models, hirudin present in the surrounding medium of a clot under lysis is more efficient than heparin in preventing the activation of coagulation. This observation suggests that hirudin may be effective in the prevention of the rethrombotic process frequently encountered during thrombolytic therapy.
- Published
- 1989
- Full Text
- View/download PDF
15. KRDS--a tetrapeptide derived from lactotransferrin--inhibits binding of monoclonal antibody against glycoprotein IIb-IIIa on ADP-stimulated platelets and megakaryocytes
- Author
-
Raha, S, Dosquet, C, Abgrall, JF, Jolles, P, Fiat, AM, and Caen, JP
- Abstract
Short peptides isolated from fibrinogen and K-casein have been shown to inhibit platelet aggregation and fibrinogen binding to stimulated platelets. We studied the effects of synthetic peptides occurring in milk proteins (bovine K-casein, KNQDK, and human lactotransferrin, KRDS) and in fibrinogen (RGDS and L10) on subsequent binding of monoclonal antibodies (MoAb) against the glycoprotein (GP) IIb-IIIa complex (AP2 and P2) on adenosine diphosphate (ADP)-stimulated and unstimulated human platelets and megakaryocytes (MKs) by using an immunoperoxidase method to visualize antibody binding. Only KRDS (900 mumol/L) inhibited the binding of AP2 and P2 on ADP (5 mumol/L)- stimulated platelets, but not on unstimulated platelets. However, the binding of P2 was considerably more inhibited than that of AP2 as judged by immunoperoxidase intensity. Radiolabeled AP2 binding was inhibited by 30% with KRDS on ADP-stimulated platelets as compared with platelets incubated in the absence of ADP. KRDS did not inhibit the binding of MoAbs against GP IIIa (SZ 21), GP IIb (SZ 22), and GP Ib (SZ 2) on ADP-stimulated human platelets. Inhibition of P2 binding by KRDS was also observed in a section of MKs isolated from human bone marrow and stimulated by 15 or 20 micron ADP. A lower concentration of ADP (5 or 10 mumol/L) failed to produce any inhibition of binding. This indicates that MKs may not be equally responsive to agonists as platelets. Moreover, P2 binding inhibition was observed in a larger (P less than .001) percentage of mature MKs (29%) as compared with younger, maturing MKs (11%). The observations suggested that a functional ability possessed by platelets, namely, agonist-induced exposure of the site of interaction of KRDS, may occur at a late stage of MK development.
- Published
- 1988
- Full Text
- View/download PDF
16. Analysis of von Willebrand factor mRNA from the lung of pigs with severe von Willebrand disease by using a human cDNA probe
- Author
-
Wu, QY, Bahnak, BR, Coulombel, L, Kerbiriou-Nabias, D, Drouet, L, Pietu, G, Meulien, P, Pavirani, A, Caen, JP, and Meyer, D
- Abstract
To examine the control of porcine von Willebrand factor (vWF) biosynthesis we cloned human vWF complementary DNA (cDNA) and investigated the expression of the vWF gene in lungs from normal pigs and pigs with severe von Willebrand's disease (vWD). Recombinant clones spanning approximately 90% of human vWF cDNA were identified in a lambda gt10 human lung cDNA library by screening with oligonucleotides. One clone spanning nucleotides 960 to 3,240 of human vWF cDNA was used to investigate the steady-state levels of vWF mRNA in lungs from normal pigs and from pigs phenotypically determined to be homozygous for vWD. This clone hybridized with genomic DNA from pig leukocytes when Southern blots were processed under very stringent conditions; therefore, human cDNA clones were considered valid probes to detect porcine mRNA. Northern blot analysis of total RNA from normal pig lung and human umbilical vein endothelial cells identified the vWF mRNA as a molecular species of approximately 9.0 kilobases (kb). A very faint to undetectable band at 9.0 kb in total RNA from lungs of vWD pigs suggested a decreased rate of transcription of the vWF gene. Sucrose density gradient centrifugation of RNA from the vWD pigs confirmed by Northern analysis that the high-molecular weight fractions contained vWF mRNA and at the same size as normal pig mRNA. Dot blot hybridization analysis of vWF and actin mRNA processed under stringent conditions demonstrated that the relative ratio of vWF mRNA to actin mRNA in the vWD pigs varied from 21% to 41% of the ratio observed in normal pigs. Because the amount of vWF mRNA is not correlated to the amount of vWF activity or antigen in plasma of vWD pigs we conclude that posttranscriptional events are also probably involved in abnormal expression of vWF in these animals.
- Published
- 1988
- Full Text
- View/download PDF
17. Absence of tubular structures and immunolabeling for von Willebrand factor in the platelet alpha-granules from porcine von Willebrand disease [published erratum appears in Blood 1987 Feb;69(2):707]
- Author
-
Cramer, EM, Caen, JP, Drouet, L, and Breton-Gorius, J
- Abstract
The electron microscopic localization of von Willebrand factor (vWF) was studied in platelets from normal and von Willebrand disease (vWD) pigs. In normal pig platelets, immunolabeling for vWF was far more intense and extensive than in human platelets and was either localized at one pole of the alpha-granule or all along its periphery or long axis. As in human platelets, this immunolabeling coincided with the presence of tubules about 200 nm in diameter. These structures were more numerous than in human platelets, with up to 30 tubules per alpha- granule. They were easily identified either in transverse sections, usually grouped in a less electron-dense part of the matrix at one pole of the alpha-granule, or in longitudinal sections parallel to the long axis of the elongated granules, or coiled around the alpha-granule core. They closely resemble those structures found in Weibel-Palade bodies. In platelets from pigs with severe vWD, these structures were absent, as was the immunolabeling for vWF; however, cytoplasmic microtubules were normally present in these platelets. Thus, the granule-associated tubules can be distinguished from the microtubules, which are larger in diameter (250 nm), are present in both normal and vWD platelets, and do not stain for vWF. These results strongly suggest that the tubular structures present in the alpha-granules of normal porcine platelets correspond to the vWF molecule itself.
- Published
- 1986
- Full Text
- View/download PDF
18. Human platelet activation in the absence of aggregation: a calcium- dependent phenomenon independent of thromboxane formation
- Author
-
Levy-Toledano, S, Maclouf, J, Bryon, P, Savariau, E, Hardisty, RM, and Caen, JP
- Abstract
In response to ionophore A 23187, thrombasthenic and EDTA-treated control platelet-rich plasmas (PRP) undergo a change in light transmission (LT) accompanied by a normal 14C-serotonin (5HT) release and thromboxane (TX) synthesis in the absence of aggregation. Ultrastructural qualitative electron microscopy revealed central apposition of organelles and loosely packed platelets in both models, while a central gel mass appeared only in thrombasthenic patients. Quantitative analysis of this ultrastructural change showed an increase in the elongation and a decrease in the circularity coefficients of thrombasthenic platelets, indicating a shape change with pseudopod formation, while EDTA-treated platelets underwent a shape change in the absence of pseudopod formation. Morphometric analysis showed that the ionophore caused extensive degranulation in both types of platelets (decrease of the granule volume), which occurred in the presence of contraction of thrombasthenic PRP (decrease of the SCS plus granule volume) but in its absence in EDTA-treated platelets. The change in LT was not inhibited by aspirin, suggesting a dissociation between release of 14C-5HT and TX formation. Moreover, it was not inhibited by creatine phosphate plus creatine phosphokinase, prostaglandin E1, or cytochalasin and/or colchicine. It was not dependent on ADP, cAMP, or the integrity of microfilaments and microtubules. However, chlorpromazine, TMB 8, and dibucaine, which interfere with intracellular membrane transport of Ca2+, inhibited this platelet activation (change in LT, 14C-5HT release and TX synthesis.
- Published
- 1982
- Full Text
- View/download PDF
19. Acquired IgG antibody occurring in a thrombasthenic patient: its effect on human platelet function
- Author
-
Levy-Toledano, S, Tobelem, G, Legrand, C, Bredoux, R, Degos, L, Nurden, A, and Caen, JP
- Abstract
In subagglutinating amounts, an IgG antibody isolated from the plasma of a polytransfused thrombasthenic patient (L) inhibited ADP-, epinephrine-, collagen-, and thrombin-induced aggregation of normal human platelets. The inhibition of ADP-induced aggregation was strongly diminished following the prior incubation of the antibody with control human platelet stroma but not with the stroma prepared from the platelets of two different thrombasthenic patients. The IgG(L) did not affect the binding of 14C-ADP to control human platelet membranes and did not inhibit the ADP-induced shape change. Bovine factor VIIIVWF- induced agglutination and ristocetin-induced aggregation of control human platelets were not inhibited in the presence of the antibody. The IgG(L) strongly inhibited ADP-induced retraction of reptilase clot and thrombin-induced clot retraction. This antibody therefore induced a thrombasthenialike state in normal human platelets, suggesting that the antigenic site recognized by the antibody plays a central role in the later stages of the mechanism of platelet aggregation induced by physiologic aggregation-inducing agents.
- Published
- 1978
- Full Text
- View/download PDF
20. Platelet factor VIII-related antigen: studies in vivo after transfusion in patients with von Willebrand disease
- Author
-
Sultan, Y, Jeanneau, C, Lamaziere, J, Maisonneuve, P, and Caen, JP
- Abstract
Four unrelated patients with a severe form of von Willebrand disease showed no detectable factor VIII-related antigen (VIIIR:AG) in either their plasma or their platelets. They received cryoprecipitate infusions, three patients in a single injection each and one every day for 9 days before and after surgery. Platelet VIIIR:Ag was studied at different times during and after transfusion using electroimmunoassay of platelet extracts and electron microscopy of the platelets incubated with anti-VIIIR:Ag antibodies coupled to peroxidase. No VIIIR-Ag was detected in or around the patients' platelets, although this antigen was detected in the circulating blood. These results that there was no VIIIR:Ag uptake from the plasma by the platelets and that platelet VIIIR:Ag came from megakaryocytes.
- Published
- 1978
- Full Text
- View/download PDF
21. Treatment of Antithrombin III Deficiency with Danazol
- Author
-
Caen Jp and Wautier Jl
- Subjects
Danazol ,medicine.medical_specialty ,Antithrombin III Deficiency ,business.industry ,Antithrombin III deficiency ,General Medicine ,medicine.disease ,Endocrinology ,Pregnadienes ,Internal medicine ,medicine ,Humans ,business ,medicine.drug - Published
- 1984
22. A Glanzmann thrombasthenia family associated with a TUBB1-related macrothrombocytopenia.
- Author
-
Guillet B, Bayart S, Pillois X, Nurden P, Caen JP, and Nurden AT
- Subjects
- Female, Genetic Predisposition to Disease, Heterozygote, Humans, Integrin beta3 blood, Integrin beta3 chemistry, Male, Models, Molecular, Multifactorial Inheritance, Pedigree, Phenotype, Protein Conformation, Risk Factors, Structure-Activity Relationship, Thrombasthenia blood, Thrombasthenia diagnosis, Thrombocytopenia blood, Thrombocytopenia diagnosis, Tubulin blood, Tubulin chemistry, Blood Platelets pathology, Hemostasis genetics, Integrin beta3 genetics, Mutation, Thrombasthenia genetics, Thrombocytopenia genetics, Tubulin genetics
- Abstract
Background: Macrothrombocytopenia (MTP) is a rare but enigmatic complication of Glanzmann thrombasthenia (GT), an inherited bleeding disorder caused by the absence of platelet aggregation due to deficiencies of the αIIbβ3 integrin., Objectives: We report a family with type I GT and a prolonged bleeding time but unusually associated with congenital mild thrombocytopenia and platelet size heterogeneity with giant forms., Methods and Results: Sanger sequencing of DNA from the propositus identified 2 heterozygous ITGB3 gene mutations: p.P189S and p.C210S both of which prevent αIIbβ3 expression and are causative of GT but without explaining the presence of enlarged platelets. High-throughput screening led to the detection of a predicted disease-causing heterozygous mutation in the TUBB1 gene: p.G146R, encoding β1-tubulin, a component of the platelet cytoskeleton and a gene where mutations are a known cause of MTP., Conclusions: Family screening confirmed that this rare phenotype results from oligogenic inheritance while suggesting that the GT phenotype dominates clinically., (© 2019 International Society on Thrombosis and Haemostasis.)
- Published
- 2019
- Full Text
- View/download PDF
23. PML/RARalpha fusion protein transactivates the tissue factor promoter through a GAGC-containing element without direct DNA association.
- Author
-
Yan J, Wang K, Dong L, Liu H, Chen W, Xi W, Ding Q, Kieffer N, Caen JP, Chen S, Chen Z, and Xi X
- Subjects
- Base Sequence, Cell Line, Tumor, Coagulation Protein Disorders etiology, DNA metabolism, Humans, Promoter Regions, Genetic, Transcription Factor AP-1 metabolism, Coagulation Protein Disorders genetics, Gene Expression Regulation, Leukemic, Leukemia, Promyelocytic, Acute complications, Oncogene Proteins, Fusion pharmacology, Thromboplastin genetics, Transcriptional Activation
- Abstract
A severe coagulopathy is a life-threatening complication of acute promyelocytic leukemia (APL) and is ascribable mainly to the excessive levels of tissue factor (TF) in APL cells regulated in response to the promyelocytic leukemia/retinoic acid receptor alpha (PML/RARalpha) fusion protein. The underlying molecular mechanisms for this regulation remain ill-defined. With U937-PR9 cell lines stably expressing luciferase reporter gene under the control of different mutants of the TF promoter, both luciferase and ChIP data allowed the localization of the PML/RARalpha-responsive sequence in a previously undefined region of the TF promoter at position -230 to -242 devoid of known mammalian transcription factor binding sites. Within this sequence a GAGC motif (-235 to -238) was shown to be crucial because deletion or mutation of these nucleotides impaired both PML/RARalpha interaction and promoter transactivation. However, EMSA results showed that PML/RARalpha did not bind to DNA probes encompassing the -230 to -242 sequences, precluding a direct DNA association. Mutational experiments further suggest that the activator protein 1 (AP-1) sites of the TF promoter are dispensable for PML/RARalpha regulation. This study shows that PML/RARalpha transactivates the TF promoter through an indirect interaction with an element composed of a GAGC motif and the flanking nucleotides, independent of AP-1 binding.
- Published
- 2010
- Full Text
- View/download PDF
24. Suppression of tumor growth by viral vector-mediated gene transfer of N-terminal truncated platelet factor 4.
- Author
-
Li Y, Jin Y, Chen H, Jie G, Tobelem G, Caen JP, and Han ZC
- Subjects
- Animals, Disease Models, Animal, Disease Progression, Female, Humans, Mice, Mice, Inbred BALB C, Neoplasm Transplantation pathology, Neoplasms blood supply, Neoplasms genetics, Neoplasms therapy, Neovascularization, Pathologic drug therapy, Survival Rate, Genetic Therapy methods, Genetic Vectors genetics, Neoplasms pathology, Platelet Factor 4 genetics, Platelet Factor 4 therapeutic use, Retroviridae genetics
- Abstract
Platelet factor four (PF4), an inhibitor of endothelial cell proliferation in vitro, inhibits angiogenesis and tumor growth in vivo in experimental animals. The present study was designed to determine whether gene therapy-mediated expression of a form of PF4 lacking 16 amino acids of N-terminus from tumor cells could inhibit angiogenesis and tumor growth in vivo. Two replication-defective recombinant retroviral vectors were constructed. One encodes human PF4 (rRV-PF4) and the other encodes the N-truncated peptide (rRVp17-70). These vectors were then used to transduce KB cells, a human head and neck squamous carcinoma cell line. Expression of PF4 and p17-70 transgenes was confirmed by Western blot analysis. In vitro, both rRV-PF4 and rRVp17-70 were able to inhibit selectively the proliferation of human umbilical vascular endothelial cells (HUVEC) but not KB cells. In vivo activity was assessed by injecting 10(7) KB cells subcutaneously into nude mice and by monitoring subsequent tumor growth, xenograft vascular histochemistry, and animal survival. Viral vector-mediated cDNA transfer of PF4 and p17-70 resulted in inhibiting solid tumors through an anti-angiogenic action in vivo. Our data indicate that targeting tumor angiogenesis using viral-mediated gene transfer of full-length and N-terminal truncated PF4 represents a promising strategy for cancer gene therapy.
- Published
- 2003
- Full Text
- View/download PDF
25. Platelet aggregation induced by the C-terminal peptide of thrombospondin-1 requires the docking protein LAT but is largely independent of alphaIIb/beta3.
- Author
-
Trumel C, Plantavid M, Lévy-Tolédano S, Ragab A, Caen JP, Aguado E, Malissen B, and Payrastre B
- Subjects
- Amino Acid Sequence, Animals, Carrier Proteins genetics, Enzyme Precursors chemistry, Enzyme Precursors metabolism, Humans, In Vitro Techniques, Intracellular Signaling Peptides and Proteins, Mice, Mice, Knockout, Peptide Fragments chemistry, Peptide Fragments genetics, Peptide Fragments pharmacology, Phosphatidylinositol 3-Kinases metabolism, Phospholipase C gamma, Phosphoproteins deficiency, Phosphoproteins genetics, Phosphorylation, Platelet Glycoprotein GPIIb-IIIa Complex physiology, Protein-Tyrosine Kinases chemistry, Protein-Tyrosine Kinases metabolism, Syk Kinase, Thrombasthenia blood, Thrombospondin 1 chemistry, Thrombospondin 1 genetics, Type C Phospholipases metabolism, Tyrosine chemistry, Adaptor Proteins, Signal Transducing, Carrier Proteins physiology, Membrane Proteins, Phosphoproteins physiology, Platelet Aggregation drug effects, Platelet Aggregation physiology, Thrombospondin 1 pharmacology
- Abstract
Thrombospondin-1 (TSP1) is abundantly secreted during platelet activation and plays a role in irreversible platelet aggregation. A peptide derived from the C-terminal domain of TSP1, RFYVVMWK (RFY) can activate human platelets at least in part via its binding to integrin-associated protein. Although integrin-associated protein is known to physically interact with alphaIIb/beta3, we found that this major platelet integrin had only a partial implication in RFY-mediated platelet aggregation. Accordingly, RFY induced a significant Glanzmann type I thrombasthenic platelet aggregation. The alphaIIb/beta3-dependent part of platelet aggregation induced by RFY was mainly due to secreted ADP and thromboxane A2. In the absence of alphaIIb/beta3 and fibrinogen, RFY stimulated a rapid tyrosine phosphorylation of a set of proteins, including Syk, linker for activation of T cells (LAT) and phospholipase Cgamma2. This signaling pathway was critical for RFY-mediated platelet activation as revealed by the use of pharmacological inhibitors as well as LAT-deficient mouse platelets. Phosphoinositide 3-kinase activation was also required for RFY-mediated platelet aggregation. Our results unravel a new alphaIIb/beta3 and fibrinogen-independent mechanism for platelet aggregation in response to the active peptide from the C-terminal domain of TSP1.
- Published
- 2003
- Full Text
- View/download PDF
26. A Ser752-->Pro substitution in the cytoplasmic domain of beta3 in a Glanzmann thrombasthenia variant fails to prevent interactions between the alphaIIbbeta3 integrin and the platelet granule pool of fibrinogen.
- Author
-
Nurden P, Poujol C, Winckler J, Combrié R, Caen JP, and Nurden AT
- Subjects
- Adenosine Diphosphate pharmacology, Aged, Blood Platelets drug effects, Case-Control Studies, Humans, Integrin beta3 metabolism, Male, Microscopy, Immunoelectron, Mutation, Platelet Activation, Protein Binding, Stimulation, Chemical, Thrombasthenia blood, Blood Platelets metabolism, Fibrinogen metabolism, Integrin beta3 genetics, Platelet Membrane Glycoprotein IIb metabolism, Thrombasthenia genetics
- Abstract
A Glanzmann thrombasthenia variant with a beta3 Ser752-->Pro cytoplasmic domain substitution has platelets that fail to aggregate or bind soluble fibrinogen (Fg) after activation. Despite this, Fg is normally present in the alpha-granules. We have used immunoelectron microscopy to examine the reactivity of Fg with the different pools of alphaIIbbeta3 in the patient's platelets. Immunogold labelling was performed on cryosections using an anti-ligand-induced binding site (LIBS) monoclonal antibody (mAb), which binds to alphaIIbbeta3 only when Fg is bound, or a mixture of two anti-receptor-induced binding site (RIBS) mAbs that specifically recognize receptor-bound Fg. Labelling of the alpha-granule membrane and channels of the surface-connected canalicular system in unstimulated platelets confirmed that the mutated alphaIIbbeta3 retains the capacity to transport Fg. When the patient's platelets were stimulated with ADP in the presence of Fg, as expected there was a much-decreased activation of surface-exposed alphaIIbbeta3. However, thrombin-induced activation was associated with both secretion and a rapid increase in the labelling of internal membrane systems by anti-RIBS and anti-LIBS mAbs, with mobilization of the internal Fg pool. Yet labelling on the surface of the patient's platelets was transient. Our studies implied that alphaIIbbeta3 in platelets may bind fibrinogen in different activation states and that this patient specifically lacked high-affinity binding.
- Published
- 2002
- Full Text
- View/download PDF
27. Induction of acetylcholinesterase expression during apoptosis in various cell types.
- Author
-
Zhang XJ, Yang L, Zhao Q, Caen JP, He HY, Jin QH, Guo LH, Alemany M, Zhang LY, and Shi YF
- Subjects
- Acetylcholinesterase drug effects, Acetylcholinesterase genetics, Animals, Antisense Elements (Genetics) pharmacology, Apoptosis drug effects, Biomarkers, Cell Compartmentation drug effects, Cell Nucleus drug effects, Cell Nucleus enzymology, Cell Nucleus ultrastructure, Cholinesterase Inhibitors pharmacology, Cytoplasm drug effects, Cytoplasm enzymology, Cytoplasm ultrastructure, DNA Fragmentation drug effects, DNA Fragmentation physiology, Eukaryotic Cells drug effects, Eukaryotic Cells ultrastructure, Gene Expression Regulation, Enzymologic drug effects, Gene Expression Regulation, Enzymologic physiology, HeLa Cells, Humans, Immunohistochemistry, Microscopy, Electron, Protein Isoforms genetics, Protein Isoforms metabolism, RNA, Messenger metabolism, Rats, Time Factors, Acetylcholine metabolism, Acetylcholinesterase metabolism, Apoptosis physiology, Cell Compartmentation physiology, Eukaryotic Cells enzymology
- Abstract
Acetylcholinesterase (AChE) plays a key role in terminating neurotransmission at cholinergic synapses. AChE is also found in tissues devoid of cholinergic responses, indicating potential functions beyond neurotransmission. It has been suggested that AChE may participate in development, differentiation, and pathogenic processes such as Alzheimer's disease and tumorigenesis. We examined AChE expression in a number of cell lines upon induction of apoptosis by various stimuli. AChE is induced in all apoptotic cells examined as determined by cytochemical staining, immunological analysis, affinity chromatography purification, and molecular cloning. The AChE protein was found in the cytoplasm at the initiation of apoptosis and then in the nucleus or apoptotic bodies upon commitment to cell death. Sequence analysis revealed that AChE expressed in apoptotic cells is identical to the synapse type AChE. Pharmacological inhibitors of AChE prevented apoptosis. Furthermore, blocking the expression of AChE with antisense inhibited apoptosis. Therefore, our studies demonstrate that AChE is potentially a marker and a regulator of apoptosis.
- Published
- 2002
- Full Text
- View/download PDF
28. A Leu7Pro mutation in the signal peptide of platelet glycoprotein (GP)IX in a case of Bernard-Soulier syndrome abolishes surface expression of the GPIb-V-IX complex.
- Author
-
Lanza F, De La Salle C, Baas MJ, Schwartz A, Boval B, Cazenave JP, and Caen JP
- Subjects
- Animals, Bernard-Soulier Syndrome blood, Blood Platelets metabolism, CHO Cells metabolism, Child, Cricetinae, Flow Cytometry, Humans, Male, Microscopy, Confocal, Platelet Glycoprotein GPIb-IX Complex analysis, Platelet Glycoprotein GPIb-IX Complex metabolism, Transfection, Bernard-Soulier Syndrome genetics, Mutation, Missense, Platelet Glycoprotein GPIb-IX Complex genetics
- Abstract
This paper describes the molecular defect of the second case of Bernard-Soulier syndrome, initially reported in 1957. Analysis of the patient's platelets by flow cytometry and Western blotting failed to detect surface expression of any of the four subunits of the glycoprotein (GP)Ib-V-IX complex and revealed small amounts of intracellular GPIbalpha, GPIbbeta and GPV but no GPIX. DNA sequencing revealed a novel missense mutation in the GPIX gene which replaced Leu (CTG) by Pro (CCG) at position 7 of the signal peptide. This mutation is, to date, the only known example of a leader sequence defect in Bernard-Soulier syndrome. The change occurred in a prototypic alpha-helical hydrophobic core region, typically enriched in leucine and devoid of proline residues. Co-transfection of GPIXPro7 with normal GPIbalpha and GPIbbeta into Chinese hamster ovary cells reproduced the platelet phenotype, resulting in no detectable GPIX, low intracellular levels of GPIbalpha and GPIbbeta, and an absence of surface expression. This mutation presumably leads to an abnormal conformation and, hence, incorrect insertion of GPIX into the endoplasmic reticulum and/or to defective signal peptide cleavage, both of which are required for correct transport to the cell membrane. This provides further evidence for a critical role of GPIX in controlling biosynthesis of the GPIb-IX complex.
- Published
- 2002
- Full Text
- View/download PDF
29. The calcium inhibitor SR33805 reduces intimal formation following injury of the porcine carotid artery.
- Author
-
Hainaud P, Bonneau M, Pignaud G, Bal dit Sollier C, André P, Hadjiisky P, Fieffé JP, Caen JP, Herbert JM, Dol F, and Drouet LO
- Subjects
- Animals, Arterial Occlusive Diseases pathology, Arterial Occlusive Diseases prevention & control, Carotid Artery Injuries, Carotid Artery, Common drug effects, Cell Count, Cell Division drug effects, Male, Microscopy, Video, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular pathology, Swine, Tunica Intima drug effects, Calcium Channel Blockers pharmacology, Carotid Artery, Common pathology, Indoles pharmacology, Sulfones pharmacology, Tunica Intima pathology
- Abstract
We studied the effect of SR33805, a calcium channel blocker, in vitro on the proliferation of vascular smooth muscle cells (SMC) stimulated by foetal calf serum, basic fibroblast growth factor and platelet derived growth factor, and in vivo with regard to SMC migration and proliferation which occurred following injury of the porcine carotid artery. The intimal lesion was induced by a silasten collar surgically positioned around the carotid artery and by a stenosis reducing blood flow by 50% for 30 days. Animals received SR33805 (5 mg/kg/day) 8 days before the induction of the lesion and up to 30 days after. In vitro, SR33805 inhibited in a dose-dependent manner growth factor-induced proliferation of SMC (0.20
- Published
- 2001
- Full Text
- View/download PDF
30. Dual structural requirements for multilineage hematopoietic-suppressive activity of chemokine-derived peptides.
- Author
-
Lecomte-Raclet L, Rholam M, Alemany M, Lazar N, Simenel C, Delepierre M, Han ZC, Cohen P, and Caen JP
- Subjects
- Amino Acid Motifs physiology, Amino Acid Sequence, Animals, Cell Lineage physiology, Chemokines chemistry, Circular Dichroism, Mice, Mice, Inbred BALB C, Molecular Sequence Data, Nuclear Magnetic Resonance, Biomolecular, Peptide Fragments chemical synthesis, Platelet Factor 4 chemistry, Protein Conformation, Protein Structure, Secondary, Spectroscopy, Fourier Transform Infrared, Structure-Activity Relationship, Chemokines physiology, Growth Inhibitors chemistry, Growth Inhibitors physiology, Hematopoiesis physiology, Peptide Fragments chemistry, Peptide Fragments physiology, Platelet Factor 4 physiology
- Abstract
Many chemokines have direct suppressive activity in vitro and in vivo on primitive hematopoietic cells. However, few chemokine-derived peptides have shown a significant activity in inhibiting hematopoiesis. Interestingly, a peptide derived from the 34-58 sequence of the CXC chemokine platelet factor 4 (PF4) produced a 30-40% inhibition of proliferation of murine hematopoietic progenitors (CFU-MK, CFU-GM, and BFU-E) in vitro, at concentrations of 30-60-fold lower than PF4. The aim of the present work was to define the structural parameters and motifs involved in conferring biological activity to the peptide PF4(34-58). Both structural predictions and determinations revealed a new helical motif that was further localized between residues 38 and 46. This helix was necessary for binding of the peptide and for permitting the functional DLQ motif at position 54-56 to activate the putative receptor site. Peptides lacking either the helical or the DLQ motif were devoid of inhibitory activity on the hematopoietic progenitors in vitro. However, among inactive peptides, only those having the helical motif counteracted the inhibition induced by the active peptide PF4(34-58). This suggested that the helix might be required for peptide interactions with a putative receptor site, whereas the DLQ motif would be implicated in the activation of this receptor. These results identify for the first time the dual requirements for the design of chemokine-derived peptides with high suppressive activity on hematopoiesis, as well as for the design of molecules with antagonistic action.
- Published
- 2000
- Full Text
- View/download PDF
31. Ex vivo expansion of megakaryocytic cells.
- Author
-
Maurer AM, Liu Y, Caen JP, and Han ZC
- Subjects
- Cell Culture Techniques methods, Cell Division, Clinical Trials as Topic, Humans, Megakaryocytes transplantation, Review Literature as Topic, Tissue Transplantation methods, Tissue Transplantation standards, Megakaryocytes cytology
- Abstract
The use of platelet transfusion to ensure the recovery of thrombopoiesis in patients constitutes high-cost support. The identification and cloning of recombinant human thrombopoietin (TPO) and the development of efficient methods of purification of hematopoietic stem cells and progenitor cells have ameliorated the development of strategies of ex vivo expansion of megakaryocyte (MK) progenitor cells and mature MKs. Synergistic combinations of cytokines including TPO, interleukin (IL)-1, IL-3, IL-11, stem cell factor, and FLT-3 ligand induce the ex vivo expansion of colony-forming unit-MK progenitors and MKs from cytokine-mobilized peripheral blood cells, bone marrow, and cord blood CD34+ cells. Depending on the various culture conditions, i.e., combinations of growth factors, initial concentration of CD34+, serum or serum-free cultures, and/or oxygen tensions, the expansion-fold of MKs and their progenitor cells vary greatly. The clinical applications of the reinfusion of ex vivo-generated MK cells have been investigated successfully in cancer patients following high-dose chemotherapy. This review reports the latest information concerning ex vivo expansion of MKs and the current status of clinical trials.
- Published
- 2000
32. Regulation of megakaryocytopoiesis.
- Author
-
Caen JP, Han ZC, Bellucci S, and Alemany M
- Subjects
- Animals, Chemokines physiology, Clinical Trials as Topic, Cytokines physiology, Hematopoiesis drug effects, Hematopoietic Cell Growth Factors pharmacology, Humans, Interferon-alpha pharmacology, Macaca mulatta, Megakaryocytes drug effects, Neoplasms blood, Platelet Factor 4 physiology, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins physiology, Quinazolines pharmacology, Quinazolines therapeutic use, Radiation Injuries, Experimental drug therapy, Receptors, Thrombopoietin, Recombinant Proteins pharmacology, Thrombin physiology, Thrombocytopenia drug therapy, Thrombocytopenia etiology, Thrombocytopenia physiopathology, Thrombopoietin pharmacology, Thrombopoietin therapeutic use, Transforming Growth Factor beta physiology, Hematopoiesis physiology, Megakaryocytes cytology, Neoplasm Proteins, Receptors, Cytokine, Thrombopoietin physiology
- Abstract
After 35 years of research, a physiological regulator of platelet production has been identified and the recombinant protein is available. With the discovery of thrombopoietin (TPO), its potential use in a wide variety of clinical megakaryocytic and platelet disorders has been expected and clinical trials have been undertaken. To date, the reported encouraging pre-clinical studies indicate that, as with erythropoietin or G-CSF, minimal toxicity can be expected. A potential limiting side-effect of TPO could be the induction of thrombosis. Nevertheless, it is too early to know whether this cytokine will be of major therapeutic importance for patients with life-threatening thrombocytopenia, such as patients undergoing bone marrow transplantation or subjected to a high dose of chemotherapy. Several experimental and clinical studies are still needed to determine the efficacy of TPO in the prevention or the amelioration of bleeding, which is the ultimate goal for the appropriate use of cytokines with haemostatic benefit. Basic and clinical studies on regulators of megakaryocytopoiesis have rapidly progressed. Now, there is no doubt that some of these regulators are effective in correcting haematopoietic disorders of various aetiologies. Studies on negative regulators not only are important to understand the regulation of megakaryocytopoiesis in normal and pathological states but also have a potential clinical application. Some of these regulators have been shown to be effective in the treatment of essential thrombocythaemia and other myeloproliferative disorders. Platelet factor 4 (PF4) and some other chemokines are also capable of protecting progenitor cells from the cytotoxicity of chemotherapeutic drugs. However, detailed investigations are still required to determine the precise mechanism(s) of action of these regulators and to establish the optimal clinical protocols of negative regulators alone or in association with positive regulators for the treatment of various haematological diseases and cancer.
- Published
- 1999
- Full Text
- View/download PDF
33. Inhibition of in vitro angiogenesis by platelet factor-4-derived peptides and mechanism of action.
- Author
-
Jouan V, Canron X, Alemany M, Caen JP, Quentin G, Plouet J, and Bikfalvi A
- Subjects
- Animals, CHO Cells, Cricetinae, Endothelium, Vascular metabolism, Endothelium, Vascular pathology, Peptides metabolism, Peptides pharmacology, Peptides therapeutic use, Platelet Factor 4 pharmacology, Platelet Factor 4 therapeutic use, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Endothelial Growth Factors metabolism, Fibroblast Growth Factor 2 metabolism, Lymphokines metabolism, Neovascularization, Pathologic prevention & control, Neovascularization, Physiologic drug effects, Platelet Factor 4 metabolism
- Abstract
In this study, we examined in detail the interaction of platelet factor-4 (PF-4) with fibroblast growth factor-2 (FGF-2) and vascular endothelial growth factor (VEGF) and the effect of PF-4-derived synthetic peptides. We show that a peptide between amino acids 47 and 70 that contains the heparin-binding lysine-rich site inhibits FGF-2 or VEGF function. This is based on the following observations: PF-4 peptide 47-70 inhibited FGF-2 or VEGF binding to endothelial cells; it inhibited FGF-2 or VEGF binding to FGFRs or VEGFRs in heparan sulfate-deficient CHO cells transfected with FGFR1 (CHOFGFR1) or VEGFR2 (CHOmVEGFR2) cDNA; it blocked proliferation or tube formation in three-dimensional angiogenesis assays; and, finally, it competed with the direct association of (125)I-PF-4 with FGF-2 or VEGF, respectively, and inhibited heparin-induced FGF-2 dimerization. A shorter C-terminal peptide (peptide 58-70), which still contained the heparin-binding lysin-rich site, had no effect. Peptide 17-58, which is located in the central part of the molecule, although it does not inhibit FGF-2 or VEGF binding or biologic activity in endothelial cells, inhibited heparin-dependent binding of (125)I-FGF-2 or (125)I-VEGF to CHOmFGFR1 or CHOmVEGFR2 cells, respectively. Shorter peptides (peptides 34-58 and 47-58) did not show any of these effects.
- Published
- 1999
34. Effect of arsenic trioxide on viability, proliferation, and apoptosis in human megakaryocytic leukemia cell lines.
- Author
-
Lu M, Levin J, Sulpice E, Sequeira-Le Grand A, Alemany M, Caen JP, and Han ZC
- Subjects
- Arsenic Trioxide, Breast Neoplasms genetics, Breast Neoplasms pathology, Gene Expression Regulation, Neoplastic drug effects, Genes, bcl-2, HL-60 Cells, Humans, In Situ Nick-End Labeling, Leukemia, Megakaryoblastic, Acute genetics, Tumor Cells, Cultured, Apoptosis drug effects, Arsenicals pharmacology, Cell Division drug effects, Cell Survival drug effects, Leukemia, Megakaryoblastic, Acute pathology, Oxides pharmacology
- Abstract
Arsenic trioxide (As2O3) has been demonstrated to be effective for the treatment of acute promyelocytic leukemia (APL) and to inhibit proliferation and produce apoptosis in the APL cell line NB4. To determine if As2O3 might be useful for the treatment of other lineages, we investigated the effects of As2O3 on viability, proliferation, and induction of apoptosis in the megakaryocytic leukemia cell lines HEL, Meg-01, UT7, and M07e. Our results showed that As2O3, at concentrations of 0.1-2.0 microM, causes a dose- and time-dependent inhibition of survival and growth in all four megakaryocytic leukemia cell lines studied. In contrast, As2O3 at similar concentrations had no effects on either viability or growth of the nonmegakaryocytic leukemia cell line HL60 and two human breast cancer cell lines, ZR75 and MCF7. In situ end-labeling of DNA fragments (TUNEL assay) indicated that As2O3, at concentrations of 0.5-2 microM, could significantly induce apoptosis in the aforementioned four megakaryocytic leukemia cell lines, but not in the nonmegakaryocytic HL60, ZR75, and MCF7 cell lines. These results were confirmed using conventional morphologic assessment and the DNA ladder assay. Induction of apoptosis in arsenic-treated Meg-01 and UT7 cells was accompanied by a dose-response decrease of Bcl-2 protein, whereas As2O3 had no effect on this measurement in HL60, ZR75, and MCF7 cell lines. Pertinently, these concentrations of As2O3 produced identical changes in the characteristics of the APL cell line NB4. Collectively, these data demonstrate that As2O3 can selectively inhibit growth and induce apoptosis in megakaryocytic leukemia cell lines. The use of As2O3 for the treatment of malignant megakaryocytic disorders should be considered.
- Published
- 1999
- Full Text
- View/download PDF
35. 1-Deamino (8-D-arginine) vasopressin infusion partially corrects platelet deposition on subendothelium in Bernard-Soulier syndrome: the role of factor VIII.
- Author
-
Lozano M, Escolar G, Bellucci S, Monteagudo J, Pico M, Ordinas A, and Caen JP
- Abstract
The mechanism of the transient beneficial effect of 1-deamino(8-D-arginine) vasopressin (dDAVP) infusion in the hemostasis of some BSS patients is not fully understood. We have studied the effect of dDAVP infusion in a BSS patient using an ex vivo perfusion system. Additional coagulation and flow cytometry studies were also performed. Prolonged bleeding time (> 30 min) was not affected by dDAVP infusion. However, perfusion experiments performed with low molecular weight heparin anticoagulated blood (which permits the study of fibrin deposition on perfused subendothelium) showed a significant increase in platelet deposition (6.2% before dDAVP infusion; 20.3% after) and fibrin formation. dDAVP infusion also caused an increase in prothrombin consumption compared with base line values (33 vs 46%). Flow cytometry studies of the patients platelets showed no changes in binding of monoclonal antibodies against CD41, CD36, CD62P or CD63. The increase in thrombus formation observed in perfusions may be dependent on FVIII since it could be reproduced by adding purified free or von Willebrand factor (vWf)-associated FVIII to the patient's blood in vitro. The shortening effect of dDAVP on bleeding time observed in some Bernard-Soulier syndrome patients might be related to an increase in factor FVIII levels induced by dDAVP infusion.
- Published
- 1999
- Full Text
- View/download PDF
36. The tetrapeptide AcSDKP reduces the sensitivity of murine CFU-MK and CFU-GM progenitors to aracytine in vitro and in vivo.
- Author
-
Aidoudi S, Guigon M, Drouet V, Caen JP, and Han ZC
- Subjects
- Animals, Cell Count drug effects, Cell Division drug effects, Cell Survival drug effects, Hematopoietic Stem Cells cytology, Male, Mice, Mice, Inbred BALB C, Oligopeptides administration & dosage, Time Factors, Antimetabolites, Antineoplastic pharmacology, Cytarabine pharmacology, Growth Inhibitors pharmacology, Hematopoietic Stem Cells drug effects, Oligopeptides pharmacology
- Abstract
The tetrapeptide Acetyl-Ser-Asp-Lys-Pro (AcSDKP) has been described as an inhibitor of CFU-S entry into DNA synthesis; as a result, its administration can protect mice against lethal doses of cytosine arabinoside (Ara-C). In the present study, we tested the protective effect of AcSDKP on CFU-MK and CFU-GM progenitor cells in mice treated at lower doses of Ara-C more relevant to human clinical situations. Firstly, we report for the first time that in vitro pre-incubation of murine BM MNC with AcSDKP at concentrations of 10(-10) and 10(-9) M for 48 h decreased CFU-MK, in parallel to CFU-GM, progenitor growth. This resulted in an increase of recovery of these progenitors after exposure to Ara-C. Secondly, we tested the effect of AcSDKP on progenitor cells in vivo in different conditions in Ara-C treated mice. We show that the administration of AcSDKP before starting Ara-C treatment resulted in a significant increase in progenitor CFU-GM, CFU-MK and mature MK numbers, 6 and 8 days after the first Ara-C injection. Interestingly, no difference was observed whether AcSDKP was started 24 or 48 h before Ara-C. In a protocol in which AcSDKP was administered for 8 days starting 48 h before Ara-C treatment, the dose did not appear to be critical at least within the range tested (4 vs. 40 micrograms/injection). In addition, the administration of AcSDKP at 64 micrograms/kg per injection for 5 days and stopping it 3 days before the end of Ara-C treatment, i.e. five instead of eight applications, further increased its protective effect. Thus our results demonstrate protective effect of AcSDKP for progenitors during a fractionated protocol of Ara-C treatment and indicates an importance of the dose and the schedule of administration of AcSDKP in designing future clinical trials.
- Published
- 1998
- Full Text
- View/download PDF
37. Platelet factor 4 modulates fibroblast growth factor 2 (FGF-2) activity and inhibits FGF-2 dimerization.
- Author
-
Perollet C, Han ZC, Savona C, Caen JP, and Bikfalvi A
- Subjects
- Animals, CHO Cells, Cell Division, Cells, Cultured, Cricetinae, Dimerization, Endocytosis drug effects, Heparitin Sulfate physiology, Mice, Receptor, Fibroblast Growth Factor, Type 1, Endothelium, Vascular metabolism, Fibroblast Growth Factor 2 metabolism, Platelet Factor 4 physiology, Receptor Protein-Tyrosine Kinases, Receptors, Fibroblast Growth Factor metabolism
- Abstract
Platelet factor 4 (PF-4) inhibits angiogenesis in vitro and in vivo. The mechanism of inhibition is poorly understood. We have investigated the mechanism of inhibition by examining the interaction of PF-4 and the fibroblast growth factor-2 (FGF-2)/fibroblast growth factor receptor (FGFR) system. PF-4 inhibited the binding of FGF-2 to high-affinity and low-affinity binding sites in murine microvascular endothelial cells (LEII cells) and proliferation. Maximum inhibition of binding to endothelial FGF receptors was observed at PF-4 concentrations between 5 and 10 microg/mL (half maximum inhibition at 0.6 micro/mL), and proliferation was completely inhibited at 2 microg/mL. At this concentration, PF-4 reduced internalization of 125I-FGF-2 by threefold and delayed degradation. To gain insight into the mechanism of inhibition, we have analyzed the interaction of PF-4 with FGF-2/FGFR by using mutant heparan sulfate-deficient Chinese hamster ovary (CHO) cells transfected with the FGFR-1 cDNA (CHOm-FGFR-1) and by examining the direct interaction with FGF-2. In the absence of heparin, PF-4 inhibited binding of 125I-FGF-2 to CHOm-FGFR-1 cells in a concentration-dependent manner, although not completely. In the presence of heparin, PF-4 abolished totally the stimulatory effect of heparin. Furthermore, PF-4 complexed to FGF-2 and inhibited endogenous or heparin-induced FGF-2 dimerization. These results indicate that PF-4 interacts with FGF-2 by complex formation, inhibiting FGF-2 dimerization, binding to FGF receptors, and internalization. This mechanism most likely contributes to the antiangiogenic properties of PF-4.
- Published
- 1998
38. New insights into the negative regulation of hematopoiesis by chemokine platelet factor 4 and related peptides.
- Author
-
Lecomte-Raclet L, Alemany M, Sequira-Le Grand A, Amiral J, Quentin G, Vissac AM, Caen JP, and Han ZC
- Subjects
- Amino Acid Sequence, Animals, Cells, Cultured, Coagulants chemistry, Mice, Mice, Inbred BALB C, Molecular Sequence Data, Peptides chemistry, Peptides pharmacology, Platelet Factor 4 chemistry, Structure-Activity Relationship, Coagulants pharmacology, Hematopoiesis drug effects, Platelet Factor 4 pharmacology
- Abstract
Platelet factor 4 (PF4) has been recognized as an inhibitor of myeloid progenitors. However, the mechanism of action of this chemokine remains poorly understood. The present study was designed to determine its structure/function relationship. A series of peptides overlapping the C-terminal and central regions of PF4 were analyzed in vitro for their action on murine hematopoietic progenitor growth to assess the minimal sequence length required for activity. The peptides p17-58 and p34-58 possessed an increased hematopoietic inhibitory activity when compared with PF4, whereas the shorter peptides p47-58 and p47-70 were equivalent to the native molecule and the peptide p58-70 was inactive. The PF4 functional motif DLQ located in 54-56 was required for the activity of these peptides. The peptide p34-58 impaired to a similar extent the growth of colony-forming unit-megakaryocyte (CFU-MK) as well as burst-forming unit-erythroid (BFU-E) and colony-forming unit-granulocyte-macrophage (CFU-GM), whereas PF4 was more active on CFU-MK. In the experiments using purified murine CD34(+) marrow cells, statistically significant inhibition induced by p34-58 was shown at concentrations of 2.2 nmol/L or greater for progenitors of the three lineages, whereas that induced by PF4 was seen at 130 nmol/L for CFU-MK and 650 nmol/L for CFU-GM and BFU-E, indicating that the p34-58 acts directly on hematopoietic progenitors and its activity is approximately 60- to 300-fold higher than PF4. The p34-58, unlike PF4, lacked affinity for heparin and its inhibitory activity could not be abrogated by the addition of heparin. In addition, an antibody recognizing p34-58 neutralized the activity of p34-58 but not whole PF4 molecule. These results demonstrate that PF4 contains a functional domain in its central region, which is independent of the heparin binding properties, and provide evidence for a model of heparin-dependent and independent pathways of PF4 in inhibiting hematopoiesis.
- Published
- 1998
39. [Chemokines and the regulation of hematopoiesis].
- Author
-
Maurer AM, Caen JP, and Han ZC
- Subjects
- Animals, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells physiology, Homeostasis, Humans, Megakaryocytes cytology, Megakaryocytes physiology, Mice, Chemokines physiology, Hematopoiesis physiology
- Abstract
Chemokines are a large family of cytokines that act not only as immune and inflammatory regulators but also as regulators of hematopoiesis. Two major subfamilies of chemokines are distinguished on the basis of whether the first two cysteines are separated by a single residue (CXC) or three residues (CX3C) or they are adjacent (CC) or there is a single C. The Macrophage Inflammatory Protein 1 alpha (MIP-1 alpha), which belongs to CC family is a powerful inhibitor of hematopoisis in vitro and in vivo. The sub-family CXC comprises two main groups. The first sub-group includes the ELR chemokines, in which interleukin-8 (IL-8) is the most prototypic and possesses suppressive activities on hematopoiesis. Platelet Factor 4 (PF4) belongs to the sub-group of non-ELR CXC chemokines. PF4 acts as an inhibitor of hematopoiesis, particularly of the megakaryocytopoiesis. Recently, it has been shown that a peptide of PF4, 34-58 which does not contain the site of heparin binding, is able to inhibit the growth of hematopoietic progenitors in vitro, providing evidence for a model of heparin dependent and independent pathways of PF4 action on hematopoiesis. PF4 can reduce the chimiosensitivity of hematopoietic cells in mice treated by the cytotoxic drug 5-Fluorouracyl, suggesting a potential clinical application of PF4 in cancer therapy.
- Published
- 1998
40. Quantitation of megakaryocytopoiesis by computerized automatic in culture image analysis.
- Author
-
Lecomte-Raclet L, Aidoudi S, Lebeurier I, Bal dit Sollier C, Kishtoo G, Aubert JF, Caen JP, and Han ZC
- Abstract
A computer-assisted automatic image procedure was karyocytopoiesis in culture. This analysis system was based on acetylcholinesterase staining, a specific staining for murine bone marrow megakaryocytes, and an image capturing instrument with a computer program. Two kinds of routine megakaryocyte culture methods were used, the plasma clot and the serum-free agar systems. A comparison between manual counting and the instrument was made. The image analysis software was able to distinguish between megakaryocytes (MK) at different stages of maturation. The results show that this analysis system can simultaneously detect not only the number of megakaryocytes and their colonies in each dish, but also the surface area of individual megakaryocytes. In addition, this analysis system functions automatically 24 hours a day and the results obtained are reproducible. Using this system, we have confirmed previous observations that thrombopoietin (TPO) and heparin stimulate both proliferation and maturation of megakaryocytes. In addition, we found that platelet factor 4 (PF-4) significantly reduced the number of megakaryocytes but not their cell surface area, whereas TGFbeta1 decreased both number and surface area of megakaryocytes, suggesting that PF4 and TGFbeta1 negatively regulate megakaryocytopoiesis by different mechanisms. We noticed that megakaryocytes grown under agar culture conditions regularly had an increased size in comparison with those grown in a plasma clot system, which may be an indication that the plasma clot culture media contains an inhibitor(s) of megakaryocyte maturation. Our data indicate that this image analysis system, in addition to its automatic and reproducible features, is more efficient and allows detection of more parameters than routine manual microscopic detection.
- Published
- 1998
- Full Text
- View/download PDF
41. Vasculogenesis from embryonic bodies of murine embryonic stem cells transfected by Tgf-beta1 gene.
- Author
-
Zhang XJ, Tsung HC, Caen JP, Li XL, Yao Z, and Han ZC
- Subjects
- Animals, Blood Vessels cytology, Cell Line, Hematopoietic Stem Cells, Mice, Stem Cells cytology, Transfection, Transforming Growth Factor beta genetics, Blood Vessels embryology, Cell Differentiation, Stem Cells physiology, Transforming Growth Factor beta physiology
- Abstract
Mouse embryonic stem (ES) cells transfected with a 1.7 kb cDNA of porcine transforming growth factor type beta1 (TGFbeta1), known as ES-T cells, were found to be able to differentiate in vitro into cystic embryonic bodies (EBs) with outspread tubular structures. Morphological analysis using light, phase-contrast and electron microscopes revealed that in culture, the EBs of ES-T cells initially developed some flat endothelial-like cells which further proliferated and migrated to form thread structures. At 8-10 days after EB formation, these thread structures further developed into net-like and tubular structures connecting directly to EBs. Immunofluorescent assays using antibodies against Flk-1 and von Willebrand factor (vWF) indicated that these net-like and tubular structures of ES-T cells consisted of vascular endothelial cells. Further analysis by RT-PCR revealed that the EBs with tubular structures expressed the mRNA of other markers of vascular endothelial cells, including VE-cadherin and platelet-endothelial cell adhesion molecule (PECAM). Cells of hematopoietic origin were not detected on the outside of EBs by immunostaining using several antibodies specific for granulocytes, macrophages and lymphocytes as well as by benzidine staining for erythroid cells on the outside of EBs. Our data demonstrates that the transfer of TGFbeta1 into ES cells results in a significant vasculogenesis without concomitant hematopoiesis. ES-T cells could therefore provide an excellent model for studying blood vessel formation and vasculogenic and hematopoietic interactions.
- Published
- 1998
- Full Text
- View/download PDF
42. A 13-24 C-terminal peptide related to PF4 accelerates hematopoietic recovery of progenitor cells in vivo in mice treated with 5-fluorouracil.
- Author
-
Aïdoudi S, Lebeurier I, Amiral J, Quentin G, Caen JP, and Han ZC
- Subjects
- Amino Acid Sequence, Animals, Blood Proteins, Drug Evaluation, Preclinical, Humans, Male, Mice, Mice, Inbred BALB C, Molecular Sequence Data, Antimetabolites, Antineoplastic pharmacology, Fluorouracil pharmacology, Hematopoietic Stem Cells drug effects, Peptides pharmacology, Platelet Factor 4 pharmacology
- Abstract
We have recently reported that platelet factor 4 (PF4), a megakaryocyte-platelet protein, is a potent inhibitor of human and murine megakaryocytopoiesis. In addition, PF4 accelerated the recovery of the marrow precursor cells in 5-fluorouracil (5-FU) treated mice. We show in this study that a slight modification of the C-terminal peptide related to PF4 (C13-24DE), which was previously reported as the carboxy terminal region of PF4 implicated in PF4 inhibitory activity, is also able to significantly increase murine high proliferating-potential-colony forming cells (HPP-CFC), colony-forming-unit megakaryocyte (CFU-MK) and colony-forming unit granulocyte-macrophage (CFU-GM) progenitor number, eight days after 5-FU administration, when it was given intraperitoneally twice a day (200 micrograms/kg/inj) prior to 5-FU administration (150 mg/kg). Furthermore, the C13-24DE pretreatment enhanced both the number and the diameter of single megakaryocyte (MK) by day 8. These data indicate that the C13-24DE peptide related to PF4 accelerated the in vivo recovery of stem cells, progenitors (CFU-GM, CFU-MK) and single MK after 5-FU treatment and may have a hemoprotective effect against chemotherapeutic agents.
- Published
- 1997
- Full Text
- View/download PDF
43. Negative regulation of mitogen-activated protein kinase activation by integrin alphaIIbbeta3 in platelets.
- Author
-
Nadal F, Lévy-Toledano S, Grelac F, Caen JP, Rosa JP, and Bryckaert M
- Subjects
- Enzyme Activation, Humans, Mitogen-Activated Protein Kinase 1, Platelet Aggregation drug effects, Platelet Glycoprotein GPIIb-IIIa Complex antagonists & inhibitors, Signal Transduction, Thrombin metabolism, Thrombin pharmacology, Blood Platelets enzymology, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Platelet Glycoprotein GPIIb-IIIa Complex pharmacology, Protein-Tyrosine Kinases metabolism
- Abstract
Activation of the mitogen-activated protein (MAP) kinase pathway in nucleated cells is dependent on both growth factor receptors and integrins engaged in cell adhesion. Human platelets are an interesting model for studying cell adhesion and the involvement of integrin engagement on extracellular signal-regulated kinase (ERK) activation, independently from the nuclear-DNA signal pathway. Maximal phosphorylation and activity of ERK2 occurred late during thrombin-induced platelet aggregation (90 s and later), an alphaIIbbeta3 integrin-dependent event. Surprisingly, alphaIIbbeta3 inhibition by the RGDS ligand peptide, or (Fab')2 fragments of the AP-2 monoclonal antibody, resulted in a 2-fold enhancement in ERK2 phosphorylation and activity. A similar 2-fold enhancement of ERK2 activation was observed in thrombasthenic platelets which are defective in alphaIIbbeta3 and do not aggregate. This suggests that ERK2 activation in thrombin-induced platelet aggregation is dependent on thrombin rather than on alphaIIbbeta3 and is down-regulated by alphaIIbbeta3 engaged in ligand (fibrinogen) binding and/or aggregation. Finally, in the absence of stirring which allows fibrinogen binding to alphaIIbbeta3 but prevents aggregation, ERK2 was again overactivated. This overactivation appears to be consecutive to inhibition of aggregation itself and to alphaIIbbeta3 ligand binding. We conclude that in platelets, alphaIIbbeta3 engaged in aggregation down-regulates thrombin-induced ERK2 activation. To our knowledge, this is the first report of a down-regulation of the MAP kinase pathway by integrin engagement.
- Published
- 1997
- Full Text
- View/download PDF
44. Reassessment of protein tyrosine phosphorylation in thrombasthenic platelets: evidence that phosphorylation of cortactin and a 64-kD protein is dependent on thrombin activation and integrin alphaIIb beta3.
- Author
-
Rosa JP, Artçanuthurry V, Grelac F, Maclouf J, Caen JP, and Lévy-Toledano S
- Subjects
- Cortactin, Enzyme Activation, Fibrinogen metabolism, Humans, Kinetics, Oligopeptides pharmacology, Phosphorylation, Blood Platelets metabolism, Microfilament Proteins metabolism, Phosphoproteins metabolism, Platelet Aggregation physiology, Platelet Glycoprotein GPIIb-IIIa Complex physiology, Protein Processing, Post-Translational, Thrombasthenia metabolism, Thrombin physiology
- Abstract
Tyrosine phosphorylation of a number of platelet proteins is dependent on platelet integrin alphaIIb beta3 (also termed GPIIb-IIIa) and its engagement in aggregation. For instance, in type I thrombasthenic platelets, which lack alphaIIb beta3 and do not aggregate, several substrates are either poorly or not phosphorylated. We have compared thrombasthenic platelets of type I, type II (15% alphaIIb beta3, functional), and variant type (50% alphaIIb beta3, no fibrinogen binding). The platelets from the three patients exhibited the same low tyrosine phosphorylation profiles, confirming the key role of functional alphaIIb beta3 in initiating protein tyrosine phosphorylation. We noted that in addition to the characteristic absence of the 100 to 105 kD doublet, a 77 to 80 kD doublet and to a lesser extent a 64-kD band, exhibited low phosphorylation kinetics, but with normal initial phosphorylation rates (up to 60 seconds). Similar results were obtained by inhibition of thrombin aggregation of control platelets by alphaIIb beta3 antagonists (the RGDS peptide or the monoclonal antibody 10E5), or in the absence of stirring (fibrinogen binding, but no aggregation). These results suggest that tyrosine phosphorylation of the 77 to 80 kD doublet, identified by immunoprecipitation as the cytoskeletal protein cortactin, and the 64 kD band are dependent both on thrombin activation during early steps and on the late steps of alphaIIb beta3 engagement in aggregation. Implications as to involvement of step-specific kinase and/or phosphatase activities are discussed.
- Published
- 1997
45. Gastrointestinal angiodysplasia in constitutional thrombocytopathies.
- Author
-
Bellucci S and Caen JP
- Subjects
- Angiodysplasia drug therapy, Blood Platelet Disorders drug therapy, Gastrointestinal Hemorrhage drug therapy, Humans, Angiodysplasia complications, Blood Platelet Disorders complications, Gastrointestinal Hemorrhage complications
- Published
- 1997
46. Relative involvement of GPIb/IX-vWF axis and GPIIb/IIIa in thrombus growth at high shear rates in the guinea pig.
- Author
-
André P, Hainaud P, Bal dit Sollier C, Garfinkel LI, Caen JP, and Drouet LO
- Subjects
- Acetates pharmacology, Animals, Anticoagulants pharmacology, Aurintricarboxylic Acid pharmacology, Guinea Pigs, Male, Peptide Fragments pharmacology, Platelet Adhesiveness, Recombinant Proteins pharmacology, Tyrosine analogs & derivatives, Tyrosine pharmacology, Blood Coagulation, Blood Platelets physiology, Hemorheology, Platelet Glycoprotein GPIb-IX Complex physiology, von Willebrand Factor physiology
- Abstract
The relative involvement of the glycoprotein (GP) Ib/IX-von Willebrand factor (vWF) axis and GPIIb/IIIa in thrombus growth at high shear rates was assessed and compared by testing the pharmacological effects of VCL, a recombinant GPIb-binding fragment of vWF (residues 504-728), aurintricarboxylic acid (ATA), which binds to the 509-695 disulfide loop of vWF, and lamifiban, a specific synthetic GPIIb/IIIa antagonist. In vivo, their effects were evaluated in guinea pig mesenteric arteries, in a model of a laser-induced cyclic thrombotic process, and ex vivo, at a shear rate of 1800 s(-1), in a capillary perfusion chamber model, in which collagen-adherent platelets are exposed to nonanticoagulated guinea pig blood. In vivo, VCL, ATA, and lamifiban administered 2 minutes after intimal injuries stopped thrombus growth, prevented the cyclic thrombotic process, and induced gradual thrombus dissolution. Ex vivo, at 1800 s(-1), collagen exposure to untreated blood for 2 minutes, 4 minutes, or two consecutive periods of 2 minutes each resulted in similar platelet adhesion, 56%, 59%, and 61%, respectively, with an average thrombus volume of 6, 19, and 17.5 microm3/microm2, respectively, without any fibrin formation. This indicated that the two consecutive perfusions did not affect the dynamic process of thrombus growth. When collagen-adherent platelets deposited after the first 2-minute perfusion were perfused for 2 minutes with VCL-, ATA-, or lamifiban-treated blood, thrombus growth was prevented and platelet adhesion remained unchanged, but fibrin formation increased on and around the predeposited platelets. These results suggest that both the GPIb/IX-vWF axis and GPIIb/IIIa are involved in in vivo platelet-to-platelet interactions at high shear rates in the guinea pig.
- Published
- 1997
- Full Text
- View/download PDF
47. Platelet factor 4 and other CXC chemokines support the survival of normal hematopoietic cells and reduce the chemosensitivity of cells to cytotoxic agents.
- Author
-
Han ZC, Lu M, Li J, Defard M, Boval B, Schlegel N, and Caen JP
- Subjects
- Apoptosis drug effects, Cell Survival drug effects, Cells, Cultured, Colony-Forming Units Assay, Drug Resistance, Fetal Blood cytology, Humans, Leukemia pathology, Recombinant Proteins pharmacology, Transforming Growth Factor beta antagonists & inhibitors, Transforming Growth Factor beta pharmacology, Tumor Cells, Cultured drug effects, beta-Thromboglobulin, Hematopoietic Stem Cells drug effects, Interleukin-8 pharmacology, Peptides pharmacology, Platelet Factor 4 pharmacology
- Abstract
The effects of platelet factor 4 (PF4) on the viability and chemosensitivity of normal hematopoietic cells and cancer cell lines were studied to determine the mechanisms whereby PF4 functions as either an inhibitor or a protector and to evaluate its clinical significance. Two other chemokines, interleukin-8 (IL-8) and neutrophil-activating peptide-2 (NAP-2), were also studied in comparison to PF4. Using a tetrazolium salt assay for cell viability, we observed that PF4 at 1 to 50 microg/mL supported the viability of normal human bone marrow cells. Approximately 45% of cells cultured for 48 hours survived, whereas 80% or more survived in the presence of PF4 5 microg/mL. PF4 also supported the viability of CD34+ cord blood (CB) cells and protected them from apoptosis induced by transforming growth factor beta1 (TGFbeta1) and cytotoxic drugs. Pretreatment of CD34+ cells by PF4, but not by TGFbeta1, caused an increase in the number of megakaryocyte colonies after these cells were replated in secondary cultures. Flow cytometry analysis showed that when CD34+ cells were preincubated with PF4 or TGFbeta1 for 12 days in hematopoietic growth factor-rich medium, an increased number of remaining CD34+ cells was observed only for PF4-treated cells. Furthermore, PF4 significantly reduced the chemosensitivity of bone marrow cells, as shown by its ability to increase the 50% inhibition concentration (IC50) of several cytotoxic agents. Like PF4, IL-8 and NAP-2 at 0.1, 0.6, and 1 microg/mL supported the survival of myeloid progenitors, including colony-forming units granulocyte, erythroblast, monocyte, megakaryocyte (CFU-GEMM), CFU-megakaryocyte (CFU-MK), CFU-granulocyte/macrophage (CFU-GM), and burst-forming units-erythroblast (BFU-E), and reduced their sensitivity to the toxicity of etoposide (ETP). Protamine sulfate at 1 to 100 microg/mL showed no such activity of PF4. Interestingly, the three chemokines failed to affect significantly the viability and chemosensitivity of three leukemic and two other tumor cell lines. Based on these results, we conclude for the first time that PF4 and IL-8 and NAP-2 support the survival of normal hematopoietic precursors and protect them from the toxicity of chemotherapeutic agents. Because such activities are unique to normal hematopoietic cells but not to the cancer cell lines evaluated, a potential clinical application of these molecules in the treatment of cancer is suggested.
- Published
- 1997
48. Impaired prothrombin consumption in Bernard-Soulier syndrome is corrected in vitro by human factor VIII.
- Author
-
Bellucci S, Girma JP, Lozano M, Meyer D, and Caen JP
- Subjects
- Antibodies, Monoclonal pharmacology, Bernard-Soulier Syndrome pathology, Cells, Cultured, Factor IX pharmacology, Humans, Thrombin antagonists & inhibitors, Thrombin immunology, von Willebrand Factor immunology, von Willebrand Factor physiology, Bernard-Soulier Syndrome metabolism, Factor VIII pharmacology, Prothrombin metabolism
- Abstract
The Bernard-Soulier syndrome (BSS) is characterized by thrombocytopenia with giant platelets, a prolonged bleeding time with defective platelet adhesion to the subendothelium related to a defect in platelet membrane glycoprotein Ib (GPIb) and a decreased prothrombin consumption. The mechanism of the latter abnormality remains unknown. In this study, we showed that this defect was corrected by the addition of purified human factor VIII (FVIII) to blood from four patients with BSS. The correction of prothrombin consumption was almost complete at concentrations between 1.5 and 3 IU/ml of FVIII procoagulant activity (VIII:C) and partially abolished by a monoclonal antibody which neutralizes VIII:C. This correction was specific for FVIII and was not observed after addition of purified human FIX. It was obtained, in the same magnitude range, with FVIII complexed to von Willebrand factor (vWF) but not with free vWF. These data provide a new insight into the knowledge of the physiological interaction between the platelet membrane and the vWF-FVIII complex facilitating plasma coagulation activation and may lead to helpful therapeutic advances.
- Published
- 1997
49. Protein tyrosine phosphatase SHP-1 fails to associate with cytoskeleton but is normally phosphorylated upon thrombin stimulation of thrombasthenic platelets.
- Author
-
Li RY, Gaits F, Ragab-Thomas JM, Maclouf J, Caen JP, Lévy-Toledano S, and Chap H
- Subjects
- Blood Platelets pathology, Blood Platelets ultrastructure, Humans, Intracellular Signaling Peptides and Proteins, Male, Phosphorylation drug effects, Protein Binding drug effects, Protein Tyrosine Phosphatase, Non-Receptor Type 11, Protein Tyrosine Phosphatase, Non-Receptor Type 6, Blood Platelets metabolism, Cytoskeleton metabolism, Protein Tyrosine Phosphatases metabolism, Thrombasthenia blood, Thrombin pharmacology
- Abstract
SHP-1 is a cytoplasmic protein tyrosine phosphatase predominantly expressed in hematopoietic cells. Upon thrombin stimulation of human platelets, SHP-1 is rapidly phosphorylated on both serine and tyrosine residues, and becomes associated with the cytoskeleton, where it could participate in the formation of multiprotein signalling complexes. In order to discriminate between signalling events occurring downstream of G-protein-coupled thrombin receptor and those subsequent to integrin alpha IIb beta 3 engagement, SHP-1 behaviour was examined in platelets from two patients lacking integrin alpha IIb beta 3 (Glanzmann's thrombasthenia). Upon thrombin stimulation, phosphorylation of SHP-1 occurred normally in thrombasthenic platelets, whereas association with the cytoskeleton was abolished. Moreover, inhibition of normal platelet aggregation with the tetrapeptide arg-gly-asp-ser (RGDS) which impairs fibrinogen binding to integrin alpha IIb beta 3, did not alter significantly SHP-1 phosphorylation. It is concluded that SHP-1 phosphorylation is not a consequence of integrin signalling but might rather occur downstream of thrombin receptor and heterotrimeric G-proteins.
- Published
- 1997
50. Therapy of chronic autoimmune purpura (ITP)
- Author
-
Bellucci S, Han ZC, and Caen JP
- Subjects
- Autoantibodies, Blood Platelets, Chronic Disease, Dexamethasone therapeutic use, Humans, Purpura, Thrombocytopenic, Idiopathic therapy
- Published
- 1997
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.