Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 522
Filtrar
1.
J Thromb Haemost ; 8(8): 1670-4, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20456750

RESUMO

Platelet activation and blood coagulation are essential for hemostasis and contribute to a variety of other biological processes such as inflammation, complement activation and tissue repair. Factor (F)XII, originally called Hageman factor, plays an important role in the kallikrein-kinin system by activating prekallikrein. In the 1960s, a platelet activity that promoted FXII activation was identified but its biochemical nature remained unknown. Inorganic polyphosphates (poly P) are polymers that consist of many phosphate residues linked by phosphoanhydride bonds. These polymers exist in all living organisms. In bacteria, poly P is important for growth and survival. Recently, poly P has been identified in human platelet dense granules. Studied have shown that upon platelet activation and secretion, poly P activates FXII, indicating that it is most likely the elusive platelet FXII activator. Poly P also regulates coagulation and fibrinolysis. In this review, we focus on early studies of FXII and the identification of platelet FXII activation activity, and discuss recent findings of poly P in FXII activation and coagulation.


Assuntos
Bioquímica/história , Plaquetas/citologia , Fator XII/metabolismo , Polifosfatos/metabolismo , Animais , Fenômenos Fisiológicos Bacterianos , Coagulação Sanguínea , Hemostasia , História do Século XX , Humanos , Inflamação , Fosfatos/química , Ativação Plaquetária
2.
J Thromb Haemost ; 2(1): 170-6, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14717981

RESUMO

Defective prothrombin consumption has been reported in the proband case of Bernard-Soulier syndrome (BSS). There is no consensus, however, on whether the formation of platelet procoagulant activity (PPA) is impaired in BSS and, if so, whether this is due to the lack of GPIb-V-IX-dependent binding of thrombin or of von Willebrand factor (VWF). We show thrombin generation (TG) in platelet-rich plasma of BSS (BSS-PRP) to be defective provided that fibrin remains present in the reaction mixture and that the giant platelets are not damaged by frequent subsampling. In BSS-PRP addition of (thrombin-free) fibrin did not increase TG as in normal PRP, supporting our previous hypothesis that the interaction of fibrin, VWF and GPIb triggers PPA development. Fibrin formed during the lag phase of TG by a snake venom enzyme which only removed fibrinopeptide A induced an immediate burst of TG, that was inhibited by a monoclonal antibody against GPIb (6D1) that abolishes ristocetin-induced binding of VWF to platelets. Inversely, inhibition of polymerization decreased TG and the residual activity was insensitive to 6D1. We conclude that polymerizing fibrin interacts with VWF so as to activate GPIb.


Assuntos
Síndrome de Bernard-Soulier/sangue , Fibrina/metabolismo , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Trombina/biossíntese , Fator de von Willebrand/metabolismo , Síndrome de Bernard-Soulier/genética , Biopolímeros/química , Biopolímeros/metabolismo , Plaquetas/metabolismo , Feminino , Fibrina/química , Humanos , Técnicas In Vitro , Pessoa de Meia-Idade , Modelos Biológicos , Complexo Glicoproteico GPIb-IX de Plaquetas/genética
3.
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
4.
Blood Rev ; 16(3): 193-202, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12163005

RESUMO

Glanzmann Thrombasthenia, an exceptional inherited platelet disorder is characterized by a complete lack of platelet aggregation due to a defect in the alpha(IIb)beta(3) complex or to a qualitative abnormality of this complex. Advances in molecular biology have permitted to precise the molecular abnormality on alpha(IIb) or beta(3) genes responsible for the disease and have also contributed to a better knowledge of normal platelet physiology. Hemorrhages are the main clinical problem. Current principles of therapeutic management are proposed, with special reference to the risk of platelet alloimmunisation.


Assuntos
Trombastenia/tratamento farmacológico , Trombastenia/etiologia , Gerenciamento Clínico , Feminino , Hemorragia/etiologia , Hemorragia/terapia , História do Século XX , História do Século XXI , Humanos , Masculino , Mutação , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Trombastenia/história
5.
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
6.
J Biol Chem ; 276(42): 38628-35, 2001 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-11507099

RESUMO

We have investigated receptor function and epitope expression of recombinant alpha(IIb)beta(3) mutated at Cys(177) or Cys(273) in the I-like domain as well as Cys(598), located in the fourth repeat of the membrane-proximal cysteine-rich region and mutated in a Glanzmann's thrombasthenia type II patient. The beta(3) mutants beta(3)C177A, beta(3)C273A, and beta(3)C598Y exhibited a decreased electrophoretic mobility in SDS-polyacrylamide gel electrophoresis under nonreducing conditions, confirming the disruption of the respective disulfide loops. Despite reduced surface expression, the alpha(IIb)beta(3)C177A, alpha(IIb)beta(3)C273A, and alpha(IIb)beta(3)C598Y receptors mediated cell adhesion to immobilized fibrinogen and translocated into focal adhesion plaques. The beta(3)C598Y mutation, but not the beta(3)C177A or beta(3)C273A mutations, induced spontaneous binding of the ligand mimetic monoclonal antibody PAC-1, while the beta(3)C177A and beta(3)C273A mutants exhibited reduced complex stability in the absence of Ca(2+). Epitope mapping of function-blocking monoclonal antibodies (mAbs) allowed the identification of two distinct subgroups; mAbs A2A9, pl2-46, 10E5, and P256 did not interact with alpha(IIb)beta(3)C273A and bound only weakly to alpha(IIb)beta(3)C177A, while mAbs AP2, LM609 and 7E3 bound normally to mutant alpha(IIb)beta(3)C273A, but interacted only weakly with mutant alpha(IIb)beta(3)C177A. Furthermore, a cryptic epitope recognized by mAb 4D10G3 and not exposed on wild type alpha(IIb)beta(3) became accessible only on mutant alpha(IIb)beta(3)C177A and was mapped to the 60-kDa chymotrypsin fragment of beta(3). Finally, the ligand-induced binding site (LIBS) epitopes AP5, D3, LIBS1, and LIBS2 were spontaneously expressed on all three mutants independent of RGDS or dithiothreitol treatment. Our results provide evidence that disruption of a single cysteine disulfide bond in the cysteine-rich repeat domain, but not in the I-like domain, activates integrin alpha(IIb)beta(3). In contrast, disruption of each of the disulfide bonds in the two long insertions of the I-like domain predicted to be in close contact with the alpha subunit beta-propeller domain affect the stability of the alpha(IIb)beta(3) heterodimer and inhibit complex-specific mAb binding without affecting the RGD binding capacity of the metal ion-dependent adhesion site-like domain.


Assuntos
Antígenos CD/química , Antígenos CD/genética , Cisteína/química , Mutação , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Glicoproteínas da Membrana de Plaquetas/química , Glicoproteínas da Membrana de Plaquetas/genética , Animais , Anticorpos Monoclonais/metabolismo , Western Blotting , Células CHO , Adesão Celular , Linhagem Celular , Cricetinae , Cisteína/genética , DNA Complementar/metabolismo , Dimerização , Dissulfetos , Ativação Enzimática , Epitopos/química , Fibrinogênio/metabolismo , Citometria de Fluxo , Humanos , Integrina beta3 , Ligantes , Microscopia de Fluorescência , Modelos Moleculares , Testes de Precipitina , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/metabolismo , Transfecção
7.
Blood ; 98(4): 1019-27, 2001 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-11493447

RESUMO

Administration of 5-fluorouracil (5-FU) to mice results in a marked increase in the level of circulating platelets in 10 days. Mice lacking Mpl, the receptor for thrombopoietin (TPO), are thrombocytopenic. To gain insight into the mechanism by which 5-FU produces such a substantial stimulation of platelet production, this study investigated whether 5-FU (150 mg/kg) produced thrombocytosis in c-mpl(-/-) mice, thus establishing whether TPO was required for this response. A 5- to 6-fold increase in platelet levels in c-mpl(-/-) mice (to approximately 1000 x 10(9)/L) was observed on days 20 and 25 after 5-FU injection. Thus, at the peak of the response, c-mpl(-/-) mice had platelet levels comparable to those in normal mice. Administration of 5-FU also produced thrombocytosis in previously splenectomized c-mpl(-/-) mice. Comparison of the platelet response to 5-FU in young (6-12 weeks) and old (33-46 weeks) c-mpl(-/-) mice found that older mice produced a much more marked response than younger mice, with a mean maximum platelet level of approximately 1700 x 10(9)/L. To determine whether this increase in circulating platelets was preceded by an increase in hematopoietic progenitors, serial cultures of bone marrow and spleen were evaluated. A considerable increase in all colony types studied was observed on days 15 and 20 in spleens of c-mpl(-/-) mice, but no similar elevations were detected in bone marrow. These results indicate that c-mpl(-/-) mice can achieve a normal level of platelets after 5-FU injection, by means of a TPO-independent mechanism, and that they respond to 5-FU myelosuppression by producing large numbers of megakaryocytic, myeloid, and erythroid progenitors. (Blood. 2001;98:1019-1027)


Assuntos
Plaquetas/efeitos dos fármacos , Fluoruracila/farmacologia , Proteínas de Neoplasias , Receptores de Citocinas , Trombocitopenia/tratamento farmacológico , Fatores Etários , Animais , Plaquetas/citologia , Células da Medula Óssea , Modelos Animais de Doenças , Fluoruracila/administração & dosagem , Megacariócitos/ultraestrutura , Camundongos , Camundongos Knockout , Contagem de Plaquetas , Ploidias , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Receptores de Trombopoetina , Baço/citologia , Esplenectomia , Células-Tronco/citologia , Trombocitopenia/sangue , Trombocitopenia/genética
8.
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
9.
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
10.
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
11.
Eur J Pharmacol ; 391(1-2): 1-9, 2000 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-10720629

RESUMO

The aim of the study was to evaluate the effect of the protein Seminal Vesicle Protein No. 4 (SV-IV), a potent inhibitor of antithrombin III (antithrombin), on the coagulation of blood obtained from patients affected by hemophilia A. In the coagulating blood of these patients, the antithrombin/thrombin ratio was found to be markedly higher (about 44) than in normal individuals (about 4. 4). This high ratio was related to the low efficiency of thrombin-generating reactions induced by the factor VIII deficiency and to the high levels of free (not bound to serine proteases) antithrombin present in the hemophilic serum (antithrombin concentration was the same in normal and hemophilic plasma). The elevated concentration of free antithrombin in hemophiliacs was primarily a consequence of a reduced consumption caused by the scarce availability in the hemophilic serum of factors Xa and IIa, which are serine proteases possessing strong binding affinity for antithrombin. Addition of SV-IV to coagulating hemophilic blood reduced markedly the serum antithrombin and thrombin-antithrombin complexes, normalizing, as a consequence, the clotting time and other coagulation parameters. Similar results were obtained by using appropriate concentration of factor VIII.


Assuntos
Antitrombina III/antagonistas & inibidores , Coagulação Sanguínea/efeitos dos fármacos , Hemofilia A/sangue , Proteínas/farmacologia , Proteínas Secretadas pela Vesícula Seminal , Inibidores de Serina Proteinase/metabolismo , Antitrombina III/análise , Antitrombina III/metabolismo , Cálcio/sangue , Fator VIII/metabolismo , Fator Xa/análise , Humanos , Tempo de Protrombina , Trombina/metabolismo , Tempo de Coagulação do Sangue Total
12.
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
13.
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
14.
Leukemia ; 13(7): 1062-70, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10400422

RESUMO

The aberrant expression of tissue factor (TF) in acute promyelocytic leukemia (APL) cells has been implicated in the pathogenesis of the APL coagulopathy. In this study, we found that in APL patients receiving ATRA or As2O3 treatment, the improvement in hypercoagulobility and hyperfibrinolysis paralleled the correction of plasma fibrinogen level and amelioration of bleeding symptoms. Notably, clinical improvement was also correlated to ATRA/As2O3-induced rapid decrease of membrane procoagulant activity (PCA) and TF contents of APL blasts. Consistent with the in vivo findings, the membrane PCA, TF antigen and its mRNA level within NB4 cells were rapidly down-regulated by 1 microM ATRA or As2O3, while 0.2 microg/ml DNR increased these TF parameters prior to its effect upon apoptosis induction. The down-regulation of TF mRNA by ATRA was partially de novo protein synthesis-dependent and at least partially attributed to a mechanism of destabilizing TF mRNA. On the other hand, in addition to its modulation on mRNA, As2O3 could also induce an accelerated TF protein turnover. These distinct effects were corroborated with the properties of these agents in causing the degradation of PML-RARalpha protein. All three therapeutic agents, however, enhanced the potential of NB4 cells to stimulate the expression of TF and PCA in endothelium. Taken together, our data suggest that the rapid and distinct regulation of TF on APL cells by these therapeutic agents might at least partially contribute to their effects on APL coagulopathy.


Assuntos
Antineoplásicos/uso terapêutico , Arsenicais/uso terapêutico , Endotélio Vascular/efeitos dos fármacos , Leucemia Promielocítica Aguda/tratamento farmacológico , Óxidos/uso terapêutico , Tromboplastina/biossíntese , Tretinoína/uso terapêutico , Adolescente , Adulto , Trióxido de Arsênio , Coagulação Sanguínea/efeitos dos fármacos , Daunorrubicina/uso terapêutico , Ensaios de Seleção de Medicamentos Antitumorais , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Feminino , Fibrinólise/efeitos dos fármacos , Humanos , Leucemia Promielocítica Aguda/metabolismo , Masculino , Pessoa de Meia-Idade
15.
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
17.
Br J Haematol ; 104(4): 886-92, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10192455

RESUMO

Anagrelide (imidazoquinazolin derivative) is a new compound proposed for the treatment of myeloproliferative disorders. In this study, Anagrelide was given to patients with essential thrombocythaemia (ET) in a compassionate-use protocol. The aim of this study was to test the effect of this drug not only on the platelet count but also on platelet volume, chemistry and function, which has not previously been reported. Thus, in ET, different functional or structural platelet abnormalities were reported: a shortening of the bleeding time, hypoaggregation to several agonists, and in particular a lack of response to adrenalin, an increase in the amount of total platelet glycoprotein IV (or CD36), and an abnormal migration of thrombospondin on electrophoresis. These different parameters were studied before and during therapy with Anagrelide. Although the platelet count was corrected, no functional or chemical abnormality was improved. Furthermore, platelet volume was shown to be constantly increased under Anagrelide. Thus, Anagrelide, in reducing the platelet count, may possibly decrease the risk of thrombosis and haemorrhage. Nevertheless, if the risk of thromboses and/or myelofibrosis is related not only to the platelet count but also to the platelet abnormalities, the persistence of a thrombocytopathy in patients treated with Anagrelide must be taken in consideration. Our data suggest that thromboses and myelofibrosis are clinical end-points which should be included in future large-scale use of Anagrelide.


Assuntos
Plaquetas , Inibidores da Agregação Plaquetária/uso terapêutico , Quinazolinas/uso terapêutico , Trombocitemia Essencial/sangue , Tempo de Sangramento , Plaquetas/química , Plaquetas/fisiologia , Criança , Humanos , Pessoa de Meia-Idade , Agregação Plaquetária , Contagem de Plaquetas
18.
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.

19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA