Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 329
Filtrar
1.
J Thromb Haemost ; 1(2): 320-9, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12871507

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Transporte/fisiologia , Proteínas de Membrana , Fosfoproteínas/fisiologia , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/fisiologia , Trombospondina 1/farmacologia , Sequência de Aminoácidos , Animais , Proteínas de Transporte/genética , Precursores Enzimáticos/química , Precursores Enzimáticos/metabolismo , Humanos , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Knockout , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfolipase C gama , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Fosforilação , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/fisiologia , Proteínas Tirosina Quinases/química , Proteínas Tirosina Quinases/metabolismo , Quinase Syk , Trombastenia/sangue , Trombospondina 1/química , Trombospondina 1/genética , Fosfolipases Tipo C/metabolismo , Tirosina/química
2.
Cell Death Differ ; 9(8): 790-800, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12107822

RESUMO

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.


Assuntos
Acetilcolina/metabolismo , Acetilcolinesterase/metabolismo , Apoptose/fisiologia , Compartimento Celular/fisiologia , Células Eucarióticas/enzimologia , Acetilcolinesterase/efeitos dos fármacos , Acetilcolinesterase/genética , Animais , Elementos Antissenso (Genética)/farmacologia , Apoptose/efeitos dos fármacos , Biomarcadores , Compartimento Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/enzimologia , Núcleo Celular/ultraestrutura , Inibidores da Colinesterase/farmacologia , Citoplasma/efeitos dos fármacos , Citoplasma/enzimologia , Citoplasma/ultraestrutura , Fragmentação do DNA/efeitos dos fármacos , Fragmentação do DNA/fisiologia , Células Eucarióticas/efeitos dos fármacos , Células Eucarióticas/ultraestrutura , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/fisiologia , Células HeLa , Humanos , Imuno-Histoquímica , Microscopia Eletrônica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Ratos , Fatores de Tempo
3.
Atherosclerosis ; 154(2): 301-8, 2001 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11166762

RESUMO

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

Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Artéria Carótida Primitiva/patologia , Indóis/farmacologia , Sulfonas/farmacologia , Túnica Íntima/patologia , Animais , Arteriopatias Oclusivas/patologia , Arteriopatias Oclusivas/prevenção & controle , Lesões das Artérias Carótidas , Artéria Carótida Primitiva/efeitos dos fármacos , Contagem de Células , Divisão Celular/efeitos dos fármacos , Masculino , Microscopia de Vídeo , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Suínos , Túnica Íntima/efeitos dos fármacos
4.
Biochemistry ; 39(31): 9612-22, 2000 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-10924159

RESUMO

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.


Assuntos
Quimiocinas/fisiologia , Inibidores do Crescimento/química , Inibidores do Crescimento/fisiologia , Hematopoese/fisiologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/fisiologia , Fator Plaquetário 4/fisiologia , Motivos de Aminoácidos/fisiologia , Sequência de Aminoácidos , Animais , Linhagem da Célula/fisiologia , Quimiocinas/química , Dicroísmo Circular , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Fragmentos de Peptídeos/síntese química , Fator Plaquetário 4/química , Conformação Proteica , Estrutura Secundária de Proteína , Espectroscopia de Infravermelho com Transformada de Fourier , Relação Estrutura-Atividade
5.
Int J Hematol ; 71(3): 203-10, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10846824

RESUMO

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.


Assuntos
Megacariócitos/citologia , Técnicas de Cultura de Células/métodos , Divisão Celular , Ensaios Clínicos como Assunto , Humanos , Megacariócitos/transplante , Literatura de Revisão como Assunto , Transplante de Tecidos/métodos , Transplante de Tecidos/normas
6.
Haemostasis ; 29(1): 27-40, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10494032

RESUMO

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.


Assuntos
Hematopoese/fisiologia , Megacariócitos/citologia , Proteínas de Neoplasias , Receptores de Citocinas , Trombopoetina/fisiologia , Animais , Quimiocinas/fisiologia , Ensaios Clínicos como Assunto , Citocinas/fisiologia , Hematopoese/efeitos dos fármacos , Fatores de Crescimento de Células Hematopoéticas/farmacologia , Humanos , Interferon-alfa/farmacologia , Macaca mulatta , Megacariócitos/efeitos dos fármacos , Neoplasias/sangue , Fator Plaquetário 4/fisiologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/fisiologia , Quinazolinas/farmacologia , Quinazolinas/uso terapêutico , Lesões Experimentais por Radiação/tratamento farmacológico , Receptores de Trombopoetina , Proteínas Recombinantes/farmacologia , Trombina/fisiologia , Trombocitopenia/tratamento farmacológico , Trombocitopenia/etiologia , Trombocitopenia/fisiopatologia , Trombopoetina/farmacologia , Trombopoetina/uso terapêutico , Fator de Crescimento Transformador beta/fisiologia
7.
Blood ; 94(3): 984-93, 1999 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10419890

RESUMO

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.


Assuntos
Fatores de Crescimento Endotelial/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Linfocinas/metabolismo , Neovascularização Patológica , Neovascularização Fisiológica , Fator Plaquetário 4/metabolismo , Animais , Células CHO , Cricetinae , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Neovascularização Patológica/prevenção & controle , Neovascularização Fisiológica/efeitos dos fármacos , Peptídeos/metabolismo , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Fator Plaquetário 4/farmacologia , Fator Plaquetário 4/uso terapêutico , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
8.
Exp Hematol ; 27(5): 845-52, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10340400

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Arsenicais/farmacologia , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Leucemia Megacarioblástica Aguda/patologia , Óxidos/farmacologia , Trióxido de Arsênio , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes bcl-2 , Células HL-60 , Humanos , Marcação In Situ das Extremidades Cortadas , Leucemia Megacarioblástica Aguda/genética , Células Tumorais Cultivadas
9.
Platelets ; 10(2-3): 141-5, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-16801084

RESUMO

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.

10.
Int J Hematol ; 68(2): 145-55, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9803673

RESUMO

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.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Citarabina/farmacologia , Inibidores do Crescimento/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Oligopeptídeos/farmacologia , Animais , Contagem de Células/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Oligopeptídeos/administração & dosagem , Fatores de Tempo
11.
Blood ; 91(9): 3289-99, 1998 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-9558385

RESUMO

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.


Assuntos
Endotélio Vascular/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fator Plaquetário 4/fisiologia , Receptores Proteína Tirosina Quinases , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Animais , Células CHO , Divisão Celular , Células Cultivadas , Cricetinae , Dimerização , Endocitose/efeitos dos fármacos , Heparitina Sulfato/fisiologia , Camundongos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos
12.
Blood ; 91(8): 2772-80, 1998 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9531587

RESUMO

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.


Assuntos
Coagulantes/farmacologia , Hematopoese/efeitos dos fármacos , Fator Plaquetário 4/farmacologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Coagulantes/química , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/farmacologia , Fator Plaquetário 4/química , Relação Estrutura-Atividade
13.
Endothelium ; 6(2): 95-106, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9930643

RESUMO

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.


Assuntos
Vasos Sanguíneos/embriologia , Diferenciação Celular , Células-Tronco/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Vasos Sanguíneos/citologia , Linhagem Celular , Células-Tronco Hematopoéticas , Camundongos , Células-Tronco/citologia , Transfecção , Fator de Crescimento Transformador beta/genética
14.
C R Seances Soc Biol Fil ; 192(5): 917-23, 1998.
Artigo em Francês | MEDLINE | ID: mdl-9871804

RESUMO

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.


Assuntos
Quimiocinas/fisiologia , Hematopoese/fisiologia , Animais , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Homeostase , Humanos , Megacariócitos/citologia , Megacariócitos/fisiologia , Camundongos
15.
Platelets ; 9(2): 121-7, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-16793687

RESUMO

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.

16.
J Biol Chem ; 272(36): 22381-4, 1997 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-9278384

RESUMO

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.


Assuntos
Plaquetas/enzimologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/farmacologia , Proteínas Tirosina Quinases/metabolismo , Ativação Enzimática , Humanos , Proteína Quinase 1 Ativada por Mitógeno , Agregação Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Transdução de Sinais , Trombina/metabolismo , Trombina/farmacologia
17.
Blood ; 89(12): 4385-92, 1997 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-9192762

RESUMO

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.


Assuntos
Plaquetas/metabolismo , Proteínas dos Microfilamentos/metabolismo , Fosfoproteínas/metabolismo , Agregação Plaquetária/fisiologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/fisiologia , Processamento de Proteína Pós-Traducional , Trombastenia/metabolismo , Trombina/fisiologia , Cortactina , Ativação Enzimática , Fibrinogênio/metabolismo , Humanos , Cinética , Oligopeptídeos/farmacologia , Fosforilação
19.
Arterioscler Thromb Vasc Biol ; 17(5): 919-24, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9157956

RESUMO

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.


Assuntos
Coagulação Sanguínea , Plaquetas/fisiologia , Hemorreologia , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Fator de von Willebrand/fisiologia , Acetatos/farmacologia , Animais , Anticoagulantes/farmacologia , Ácido Aurintricarboxílico/farmacologia , Cobaias , Masculino , Fragmentos de Peptídeos/farmacologia , Adesividade Plaquetária , Proteínas Recombinantes/farmacologia , Tirosina/análogos & derivados , Tirosina/farmacologia
20.
Blood ; 89(7): 2328-35, 1997 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-9116276

RESUMO

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.


Assuntos
Células-Tronco Hematopoéticas/efeitos dos fármacos , Interleucina-8/farmacologia , Peptídeos/farmacologia , Fator Plaquetário 4/farmacologia , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Resistência a Medicamentos , Sangue Fetal/citologia , Humanos , Leucemia/patologia , Proteínas Recombinantes/farmacologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/farmacologia , Células Tumorais Cultivadas/efeitos dos fármacos , beta-Tromboglobulina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA