Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Mol Biol ; 431(5): 885-903, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30653992

RESUMO

Proprotein convertase subtilisin/kexin 9 (PCSK9) is a key regulator of lipid metabolism by degrading liver LDL receptors. Structural studies have provided molecular details of PCSK9 function. However, the N-terminal acidic stretch of the PCSK9 prodomain (Q31-T60) has eluded structural investigation, since it is in a disordered state. The interest in this region is intensified by the presence of human missense mutations associated with low and high LDL-c levels (E32K, D35Y, and R46L, respectively), as well as two posttranslationally modified sites, sulfated Y38 and phosphorylated S47. Herein we show that a segment within this region undergoes disorder-to-order transition. Experiments with acidic stretch-derived peptides demonstrated that the folding is centered at the segment Y38-L45, which adopts an α-helix as determined by NMR analysis of free peptides and by X-ray crystallography of peptides in complex with antibody 6E2 (Ab6E2). In the Fab6E2-peptide complexes, the structured region features a central 2 1/4-turn α-helix and encompasses up to 2/3 of the length of the acidic stretch, including the missense mutations and posttranslationally modified sites. Experiments with helix-breaking proline substitutions in peptides and in PCSK9 protein indicated that Ab6E2 specifically recognizes the helical conformation of the acidic stretch. Therefore, the observed quantitative binding of Ab6E2 to native PCSK9 from various cell lines suggests that the disorder-to-order transition is a true feature of PCSK9 and not limited to peptides. Because the helix provides a constrained spatial orientation of the missense mutations and the posttranslationally modified residues, it is probable that their biological functions take place in the context of an ordered conformational state.


Assuntos
Pró-Proteína Convertase 9/genética , Sequência de Aminoácidos , Linhagem Celular Tumoral , Cristalografia por Raios X/métodos , Células Hep G2 , Humanos , Mutação de Sentido Incorreto/genética , Peptídeos/genética , Processamento de Proteína Pós-Traducional/genética , Receptores de LDL/genética , Serina Endopeptidases/genética
2.
Cancer Res ; 50(5): 1550-8, 1990 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-1689212

RESUMO

High levels of expression of either the epidermal growth factor receptor or the receptor-like HER2/neu gene product p185HER2 have been observed in a variety of human malignancies. Because of the association of this high level expression with certain human tumors, we have generated a panel of monoclonal antibodies specific for either the epidermal growth factor receptor or p185HER2 to study their structure, function, and antigenic domains in the normal and neoplastic states. We used the epidermoid carcinoma line A431 to generate five monoclonal antibodies which immunoprecipitate the epidermal growth factor receptor. These monoclonal antibodies bind to the extracellular domain of the epidermal growth factor receptor and demonstrate variable effects on epidermal growth factor binding. We used a stably transfected NIH 3T3 cell line expressing the HER2/neu gene to produce and characterize 10 monoclonal antibodies which immunoprecipitate p185HER2. These monoclonal antibodies bind to the extracellular domain of p185HER2 and do not cross-react with the epidermal growth factor receptor. The characteristics and potential applications of these monoclonal antibodies will be discussed.


Assuntos
Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Receptores ErbB/imunologia , Proteínas Proto-Oncogênicas/imunologia , Anticorpos Monoclonais/metabolismo , Reações Antígeno-Anticorpo/imunologia , Reações Cruzadas/imunologia , Epitopos , Receptores ErbB/metabolismo , Humanos , Imunoglobulina G/imunologia , Testes de Precipitina , Proteínas Proto-Oncogênicas/metabolismo , Receptor ErbB-2 , Células Tumorais Cultivadas/imunologia
3.
Circulation ; 103(8): 1135-41, 2001 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-11222478

RESUMO

BACKGROUND: Thrombosis and neointima formation limit the efficacy of coronary angioplasty (PTCA). Clinical trials have implicated the adhesion molecules integrin alpha(IIb)beta(3) and integrin alpha(v)beta(3) in these processes. The roles of these molecules in vascular smooth muscle cell adhesion, platelet aggregation, and the thrombotic and neointimal response to oversize porcine PTCA was investigated by use of a selective alpha(IIb)beta(3) antagonist (lamifiban), a selective alpha(v)beta(3) antagonist (VO514), and a combined alpha(IIb)beta(3)/alpha(v)beta(3) antagonist (G3580). METHODS AND RESULTS: In vitro, both alpha(v)beta(3) inhibitors caused dose-dependent inhibition of porcine vascular smooth muscle cell adhesion to vitronectin but not to collagen type IV, fibronectin, or laminin, whereas selective alpha(IIb)beta(3) inhibition had no effect. Intravenous infusions of either alpha(IIb)beta(3) inhibitor in swine profoundly inhibited ex vivo platelet aggregation to ADP, whereas selective alpha(v)beta(3) inhibition had no effect. In a porcine PTCA model, intravenous infusions of the integrin antagonists were administered for 14 days after oversized balloon angioplasty injury. After PTCA, there was regional upregulation of integrin alpha(v)beta(3) in the developing neointima, as assessed by immunohistochemistry. Six hours after PTCA, obstruction of lumen by thrombus was reduced significantly by alpha(IIb)beta(3) inhibition compared with either control or alpha(v)beta(3) inhibition (mean control, 18.7%; VO514, 18.5%; lamifiban, 6.4%; G3580, 7.9%). Twenty-eight days after PTCA, there was a significant reduction of neointima with inhibitors of either integrin (mean intima/media ratio: control, 3.08; VO514, 1.33; lamifiban, 0.97; G3580, 1.32). CONCLUSIONS: We conclude that both integrin alpha(IIb)beta(3) and integrin alpha(v)beta(3) participate in neointima development after experimental angioplasty.


Assuntos
Acetatos/uso terapêutico , Trombose/prevenção & controle , Tirosina/análogos & derivados , Tirosina/uso terapêutico , Acetatos/farmacologia , Angioplastia Coronária com Balão/efeitos adversos , Animais , Adesão Celular/efeitos dos fármacos , Modelos Animais de Doenças , Imuno-Histoquímica , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiologia , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/biossíntese , Receptores de Vitronectina/antagonistas & inibidores , Receptores de Vitronectina/biossíntese , Suínos , Trombose/etiologia , Túnica Íntima/efeitos dos fármacos , Tirosina/farmacologia
4.
J Med Chem ; 35(11): 2040-8, 1992 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-1597855

RESUMO

Stimulation of platelets activates GPIIbIIIa, the heterodimeric integrin receptor, to bind fibrinogen (Fg), which results in platelet aggregation. GPIIbIIIa/Fg binding inhibitors are potentially suitable for acute use during and after thrombolytic therapy as antithrombotic agents. Incorporation of the tripeptide sequence Arg-Gly-Asp (RGD), a common structural element of many integrin ligands, into cyclic peptides produced a series of peptides of the general structure BrAc-(AA1)-RGD-Cys-OH, which were prepared by solid-phase peptide synthesis. Cyclization was accomplished by reaction of the N-terminal bromoacetyl group with the cysteine sulfhydryl at pH 8 at high dilution, resulting in thioether-bridged cyclic peptides [cyclo-S-Ac-(AA1)-RGD-Cys-OH]. Use of alpha-substituted bromoacetyl groups gave rise to an analogous series of acetyl-substituted thioether-bridged cyclic peptides. Oxidation of the thioethers produced separable diastereomeric sulfoxide-bridged cyclic peptides. After thorough evaluation in a GPIIbIIIa ELISA assay and a platelet aggregation assay, G-4120 (70A; AA1 = D-Tyr; sulfoxide bridge) was selected for further investigation as an antithrombotic agent. G-4120 was equipotent in the platelet aggregation assay to kistrin, a highly potent inhibitor of fibrinogen-mediated platelet aggregation isolated from snake venom (IC50 = 0.15 microM).


Assuntos
Fibrinolíticos/síntese química , Peptídeos Cíclicos/síntese química , Peptídeos/síntese química , Inibidores da Agregação Plaquetária/síntese química , Receptores Imunológicos/química , Receptores de Peptídeos , Sulfóxidos/síntese química , Sequência de Aminoácidos , Ciclização , Ensaio de Imunoadsorção Enzimática , Fibrinolíticos/química , Fibrinolíticos/farmacologia , Humanos , Conformação Molecular , Dados de Sequência Molecular , Estrutura Molecular , Peptídeos/química , Peptídeos/farmacologia , Peptídeos Cíclicos/química , Peptídeos Cíclicos/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/química , Inibidores da Agregação Plaquetária/farmacologia , Glicoproteínas da Membrana de Plaquetas/metabolismo , Receptores Imunológicos/metabolismo , Relação Estrutura-Atividade , Sulfóxidos/química , Sulfóxidos/farmacologia , Difração de Raios X
5.
J Med Chem ; 39(16): 3139-47, 1996 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-8759635

RESUMO

The potent and selective GP IIb-IIIa antagonist lamifiban (1, Ro 44-9883) is currently in clinical development as an injectable antithrombotic agent for treating and preventing acute coronary syndromes. However, for secondary prevention of thrombotic occlusions, orally active inhibitors are needed. By means of a prodrug strategy, the modest oral absorption of 1 in mice was improved by a factor of 9. In addition, these studies demonstrated that an amidoxime group can serve as a prodrug functionality for an amidino group. Application of this principle to the structurally related amidino carboxylate 13 led to the amidoxime ester 18 which was absorbed approximately 20 times better, after oral administration to mice, than 13. Due to the modification of the amidino group as well as of the carboxylate group, 18 completely lost its ability to interact with purified platelet GP IIb-IIIa. After oral administration of 18 to rats, dogs, and rhesus monkeys, the bioavailability of the active derivative 13 was 26 +/- 5, 25 +/- 6, and 33 +/- 6%, respectively, and the elimination half-life was 4.1 +/- 1.7, 11.4 +/- 1.1, and 5.1 +/- 1.4 h, respectively. On the basis of these properties, the orally active 18 (Ro 48-3657), a double prodrug of the potent and selective non-peptide GP IIb-IIIa antagonist 13 (Ro 44-3888), was selected as clinical candidate for evaluation as a prophylactic agent in patients at high risk for arterial thrombosis.


Assuntos
Amidinas/farmacologia , Oximas/farmacologia , Piperidinas/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Pró-Fármacos/farmacologia , Acetatos/farmacologia , Administração Oral , Amidinas/química , Amidinas/farmacocinética , Animais , Anticoagulantes/administração & dosagem , Anticoagulantes/síntese química , Anticoagulantes/farmacocinética , Anticoagulantes/farmacologia , Cães , Macaca mulatta , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Camundongos , Estrutura Molecular , Oximas/química , Oximas/farmacocinética , Piperidinas/química , Piperidinas/farmacocinética , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/química , Inibidores da Agregação Plaquetária/farmacocinética , Pró-Fármacos/administração & dosagem , Pró-Fármacos/síntese química , Pró-Fármacos/farmacocinética , Ratos , Tirosina/análogos & derivados , Tirosina/farmacologia
6.
Thromb Haemost ; 79(1): 169-76, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9459344

RESUMO

Ro 44-3888 is a potent and selective antagonist of GP IIb/IIIa. Following I.V. administration to rhesus monkeys, the (mean +/- SD.) clearance, volume of distribution and terminal half-life of Ro 44-3888 were 4.4 +/- 1.8 ml/min/kg, 0.8 +/- 0.4 l/kg and 2.5 +/- 0.8 h respectively. Oral administration of Ro 48-3657 (1 mg/kg), a doubly protected prodrug form, produced peak concentrations of Ro 44-3888 (152 +/- 51 ng/ml), 4.2 +/- 2.2 h after dosing. Terminal half-life and estimated bioavailability were 5.1 +/- 1.6 h and 33 +/- 6% respectively. No effect on blood pressure, heart rate or platelet counts were seen. Adenosine diphosohate (ADP) induced platelet aggregation (PA) and cutaneous bleeding times (CBT) were determined prior to and after the last of 8 daily oral administrations of Ro 48-3657 (0.25 or 0.5 mg/kg) to eight rhesus monkeys. Peak and trough plasma concentrations were proportional to dose and steady state was achieved after the second administration. Inhibition of PA and prolongation of CBT were concentration dependent. The ex vivo IC50 (82 nM) for ADP-mediated PA correlated with a value (58 nM) determined in vitro. The CBT response curve was displaced to the right of the PA curve. CBT was prolonged to > or = 25 min when levels of Ro 44-3888 exceeded 190 nM and PA was > 90% inhibited. Therefore, in rhesus monkeys, Ro 48-3657 is reproducibly absorbed and converted to its active form, is well tolerated, and has a concentration-dependent effect on PA and CBT. These properties make Ro 48-3657 an attractive candidate for evaluation in patients at high risk for arterial thrombosis.


Assuntos
Amidinas/farmacologia , Piperidinas/farmacologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Pró-Fármacos/farmacocinética , Administração Oral , Amidinas/efeitos adversos , Amidinas/farmacocinética , Animais , Esquema de Medicação , Testes Hematológicos , Macaca mulatta , Oximas/farmacologia , Piperidinas/efeitos adversos , Piperidinas/farmacocinética , Pró-Fármacos/efeitos adversos , Pró-Fármacos/farmacologia
7.
J Biol Chem ; 268(8): 5997-6003, 1993 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-8095500

RESUMO

The natriuretic peptide receptor type A (NPR-A) is a receptor-guanylyl cyclase whose cytoplasmic enzymatic activity is stimulated by atrial natriuretic peptide binding to the extracellular domain. NPR-A expressed in COS cells is heterogeneously glycosylated, and the more highly glycosylated protein is also phosphorylated. Upon hormone binding, dephosphorylation occurs from both serine and threonine residues, probably within the kinase homology domain of NPR-A, and may be involved with receptor desensitization. Using site-specific mutations in the kinase homology domain of NPR-A, we have identified several residues that are important for regulating the guanylyl cyclase activity of NPR-A. Some of these amino acids are probably essential for maintaining the proper tertiary structure of the intracellular domain, and others may form loops that allow for binding of ATP, which is required for proper enzymatic activity. The site-specific mutants which have greatly reduced enzymatic activity are not phosphorylated and are incompletely glycosylated. These results suggest a correlation between phosphorylation and complete glycosylation of NPR-A and that both are required for hormone-induced enzymatic activity.


Assuntos
Fator Natriurético Atrial/metabolismo , Guanilato Ciclase/metabolismo , Receptores do Fator Natriurético Atrial/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Células Cultivadas , Clonagem Molecular , DNA de Cadeia Simples , Ativação Enzimática , Glicosilação , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fosforilação , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Receptores do Fator Natriurético Atrial/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos
8.
J Biol Chem ; 258(15): 9254-61, 1983 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-6307999

RESUMO

Calcium-activated neutral protease has been purified partially from A431-V1 cells, a high passage variant of human epidermoid carcinoma A431 cells having extraordinary activity of this protease. This activity cleaves Mr = 160,000 epidermal growth factor receptors, converting them to a Mr = 145,000 form in detergent-treated membrane preparations, but not in intact cells or membrane vesicles. Properties of this activity, including molecular weight, resemble those of previously described Ca2+-activated neutral proteases. A431-V1 cell Ca2+-activated neutral protease action on epidermal growth factor receptors required 0.5 mM Ca2+ for half-maximal activity and the optimal pH was 6.5. Degradation was inhibited completely by Ca2+ chelators (EDTA and ethylene glycol bis(beta-aminoethyl ether)-N,N,N',N'-tetraacetic acid) and sulfhydryl group specific reagents (iodoacetate and 5,5'-dithiobis-(2-nitrobenzoic acid] and was inhibited partially by leupeptin. Degradation of epidermal growth factor receptors by Ca2+-activated neutral protease or trypsin was compared in intact cells, membrane vesicles, detergent-solubilized membranes, and membranes subjected to hypotonic shock in solutions containing protease. Results support the hypothesis that protease-susceptible domains of epidermal growth factor receptors, including the Ca2+-activated neutral protease-sensitive domain, are located on the cytosolic surface. The localization of Ca2+-activated neutral protease in A431-V1 cells is entirely cytosolic, making it available in cells to the protease-susceptible site on epidermal growth factor receptor substrates.


Assuntos
Cálcio/metabolismo , Receptores de Superfície Celular/metabolismo , Teratoma/metabolismo , Animais , Calpaína , Cátions Bivalentes/farmacologia , Linhagem Celular , Endopeptidases/metabolismo , Receptores ErbB , Fibroblastos/metabolismo , Humanos , Membranas/metabolismo , Peso Molecular
9.
Biochemistry ; 40(3): 675-82, 2001 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-11170384

RESUMO

Tissue factor is the cell membrane-anchored cofactor for factor VIIa and triggers the coagulation reactions. The initial step is the conversion of factor VII to factor VIIa which, in vitro, is efficiently catalyzed by low concentrations of factor Xa. To identify the tissue factor region that interacts with the activator factor Xa during this process, we evaluated a panel of soluble tissue factor (1-219) mutants for their ability to support factor Xa-mediated activation of factor VII. The tissue factor residues identified as most important for this interaction (Tyr157, Lys159, Ser163, Gly164, Lys165, Lys166, and Tyr185) were identical to those found to be important for the interaction of substrate factor X with the tissue factor.factor VIIa complex. The residues form a continuous surface-exposed patch with an area of about 500 A(2), which appears to be located outside the tissue factor-factor VII contact zone. In agreement, the two monoclonal antibodies 5G6 and D3H44-F(ab')(2), whose epitopes overlap with this identified region, inhibited the rates of factor VII activation by 86% and 95%, respectively. These antibodies also strongly inhibited the conversion of (125)I-labeled factor VII when cell membrane-expressed, full-length tissue factor (1-263) was employed. Together the results suggest the usage of a common surface region of tissue factor in its dual role-as a cofactor for factor Xa-mediated factor VII activation and as a cofactor for factor VIIa-mediated factor X activation. The finding that factor Xa and factor X may engage in similar, if not identical, molecular interactions with tissue factor further indicates that factor Xa and factor X are similarly oriented toward their respective interaction partners in the ternary catalytic complexes.


Assuntos
Fator VII/metabolismo , Fator Xa/metabolismo , Tromboplastina/metabolismo , Ácido 1-Carboxiglutâmico/metabolismo , Linhagem Celular , Membrana Celular/enzimologia , Membrana Celular/genética , Membrana Celular/metabolismo , Eletroforese em Gel de Poliacrilamida , Ativação Enzimática , Precursores Enzimáticos/metabolismo , Fator VIIa/metabolismo , Humanos , Radioisótopos do Iodo , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Mapeamento de Peptídeos , Ligação Proteica/genética , Estrutura Terciária de Proteína/genética , Solubilidade , Ressonância de Plasmônio de Superfície , Tromboplastina/genética
10.
Biochemistry ; 39(25): 7380-7, 2000 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-10858285

RESUMO

The enzymatic activity of coagulation factor VIIa is controlled by its cellular cofactor tissue factor (TF). TF binds factor VIIa with high affinity and, in addition, participates in substrate interaction through its C-terminal fibronectin type III domain. We analyzed surface-exposed residues in the C-terminal TF domain to more fully determine the area on TF important for substrate activation. Soluble TF (sTF) mutants were expressed in E. coli, and their ability to support factor VIIa-dependent substrate activation was measured in the presence of phospholipid vesicles or SW-13 cell membranes. The results showed that factor IX and factor X interacted with the same TF region located proximal to the putative phospholipid surface. According to the degree of activity loss of the sTF mutants, this TF region can be divided into a main region (residues Tyr157, Lys159, Ser163, Gly164, Lys165, Lys166, Tyr185) forming a solvent-exposed patch of 488 A(2) and an extended region which comprises an additional 7-8 residues, including the distally positioned Asn199, Arg200, and Asp204. Some of the identified TF residues, such as Trp158 and those within the loop Lys159-Lys165, are near the factor VIIa gamma-carboxyglutamic acid (Gla) domain, suggesting that the factor VIIa Gla-domain may also participate in substrate interaction. Moreover, the surface identified as important for substrate interaction carries a net positive charge, suggesting that charge interactions may significantly contribute to TF-substrate binding. The calculated surface-exposed area of this substrate interaction region is about 1100 A(2), which is approximately half the size of the TF area that is in contact with factor VIIa. Therefore, a substantial portion of the TF surface (3000 A(2)) is engaged in protein-protein interactions during substrate catalysis.


Assuntos
Fator IX/metabolismo , Fator X/metabolismo , Tromboplastina/metabolismo , Amidas/metabolismo , Anticorpos/imunologia , Ativação Enzimática , Cinética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Fosfolipídeos/metabolismo , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Tromboplastina/química , Tromboplastina/genética , Tromboplastina/imunologia
11.
Arch Biochem Biophys ; 259(2): 296-304, 1987 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-3122660

RESUMO

The specific tyrosine phosphorylation of glucose-6-phosphate dehydrogenase (G6PDH) by the epidermal growth factor (EGF) receptor in vitro is demonstrated. The Km values of the substrate G6PDH and of ATP for the receptor tyrosine kinase were ca. 1 and 10 microM, respectively. The rate of phosphorylation was EGF dependent, with a four-fold increase in Vmax in the presence of EGF. The phosphorylation was stimulated maximally by 0.2 microM or greater EGF, with an ED50 of ca. 20 nM which is consistent with the affinity of the solubilized receptor for EGF. Using conditions of 5 microM G6PDH, 100 microM ATP, 5 mM Mg2+, and 1 mM Mn2+, up to 0.3 mol phosphate was incorporated into 1 mol of the 55-kDa subunit of Baker's yeast G6PDH. Tryptic peptide mapping revealed several unique phosphopeptides for both Baker's yeast and bovine adrenal G6PDH. The patterns of phosphopeptides for a given enzyme were identical for basal and EGF-stimulated phosphorylation.


Assuntos
Receptores ErbB/metabolismo , Glucosefosfato Desidrogenase/metabolismo , Proteínas Tirosina Quinases/metabolismo , Animais , Bovinos , Cryptococcus/enzimologia , Geobacillus stearothermophilus/enzimologia , Humanos , Cinética , Leuconostoc/enzimologia , Magnésio/metabolismo , Manganês/metabolismo , Proteína Oncogênica pp60(v-src) , Mapeamento de Peptídeos , Fosfoproteínas/análise , Fosfopiruvato Hidratase/metabolismo , Fosforilação , Coelhos , Proteínas dos Retroviridae/metabolismo , Saccharomyces cerevisiae/enzimologia
12.
Proc Natl Acad Sci U S A ; 87(7): 2471-5, 1990 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-2320569

RESUMO

The purification, complete amino acid sequence, and biological activity are described for several homologous snake venom proteins that are platelet glycoprotein (GP) IIb-IIIa antagonists and potent inhibitors of platelet aggregation. The primary structures of kistrin (from Agkistrodon rhodostoma), bitan (from Bitis arietans), three isoforms of trigramin (from Trimeresusus gramineus), and an isoform of echistatin (from Echis carinatus) were determined by automated sequence analysis and fast atom bombardment mass spectrometry analysis. Each of the protein in this family, which range from 47 to 83 residues, contains an Arg-Gly-Asp amino acid sequence found in protein ligands that binds to GPIIb-IIIa, a high (17 +/- 1%) cysteine content conserved in the primary sequence, and a homologous N-terminal region absent only in the echistatin isoforms. Each protein directly inhibits the interaction of purified platelet GPIIb-IIIa to immobilized fibrinogen about 100 times more effectively than does the pentapeptide Gly-Arg-Gly-Asp-Ser; IC50 values range from 1.1 to 3.0 nM. The IC50 value for the inhibition of platelet aggregation, using human platelet-rich plasma stimulated with ADP, ranges from 110 to 550 nM for the various proteins, about 1000-fold more potent than Gly-Arg-Gly-Asp-Ser. Kistrin binds reversibly to both resting and ADP-activated human platelets with high affinity (Kd = 10.8 nM and 1.7 nM, respectively) and to purified GPIIb-IIIa with a lower affinity (Kd = approximately 100 nM). Finally, kistrin injected at 1.0 mg/kg into rabbits reversibly inhibits platelet aggregation ex vivo over 30 min without induction of thrombocytopenia. We propose that these proteins are members of a general class of proteins found in the venom of pit vipers that inhibit platelet aggregation by antagonism of the GPIIb-IIIa-fibrinogen interaction and as such serve as potential antithrombotic agents.


Assuntos
Inibidores da Agregação Plaquetária , Glicoproteínas da Membrana de Plaquetas/antagonistas & inibidores , Venenos de Serpentes/farmacologia , Sequência de Aminoácidos , Animais , Ensaio de Imunoadsorção Enzimática , Humanos , Masculino , Espectrometria de Massas , Dados de Sequência Molecular , Coelhos , Homologia de Sequência do Ácido Nucleico , Venenos de Serpentes/isolamento & purificação
13.
Growth Regul ; 1(2): 72-82, 1991 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-1688187

RESUMO

The HER2 protooncogene encodes a growth factor receptor-like transmembrane protein tyrosine kinase (p185HER2) whose ligand remains to be fully characterized. The overexpression of p185HER2 is implicated in aggressive forms of breast and ovarian cancers. The role of p185HER2 in aggressive malignancy, as well as its cell surface localization, makes it an attractive target for therapeutic monoclonal antibodies. In this report we have studied the modulation of p185HER2 function with 2 monoclonal antibodies, termed 4D5 and 6E9, which bind the extracellular domain of p185HER2. 4D5 inhibited proliferation of p185HER2 overexpressing SK-BR-3 human breast carcinoma cells (ED50 of approximately 1 nM) but did not inhibit proliferation of cultured human breast carcinoma MCF7 cells, low expressors of p185HER2. Monoclonal antibody 6E9 does not inhibit the growth of either cell line. Antibody binding studies revealed 2 populations of p185HER2 molecules on SK-BR-3 cells: one of high abundance (approximately 2 x 10(6) sites/cell) recognized by 4D5 (Kd approximately 6 nM) and the other of low abundance (2 x 10(4) sites/cell) recognized by 6E9 (Kd approximately 0.1 nM). 4D5, in an agonistic manner, downregulated SK-BR-3 cell surface p185HER2, was internalized, and stimulated p185HER2 phosphorylation in intact cells. Phosphoamino acid analysis of p185HER2 derived from SK-BR-3 cells incubated with the 4D5 monoclonal antibody demonstrated increased tyrosine, serine and threonine phosphorylation. 4D5, on short term (5 min) exposure to SK-BR-3 cells, stimulated inositol lipid hydrolysis as evidenced by increased intracellular levels of inositol polyphosphates (InsP) and sn-1,2-diacylglycerol (sn-1,2-DAG). On longer (24 h) exposure to the cells, the antibody appeared to downregulate this signalling pathway since the intracellular levels of InsP and sn-1,2-DAG decreased by 30 to 40%. 6E9 did not inhibit SK-BR-3 cell proliferation, downregulate surface p185HER2, stimulate receptor phosphorylation, or stimulate the second messenger pathway. Despite these agonistic properties, 4D5 was an inhibitor of SK-BR-3 cell proliferation at all concentrations tested (0.7 to 70 pM). The data suggest that 4D5 is a partial or weak agonist and thus may inhibit cell proliferation by mimicking ligand-like receptor downregulation.


Assuntos
Anticorpos Monoclonais/química , Proteínas Oncogênicas Virais/imunologia , Receptores de Superfície Celular/efeitos dos fármacos , Células Tumorais Cultivadas/efeitos dos fármacos , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Sítios de Ligação de Anticorpos , Neoplasias da Mama/tratamento farmacológico , Diglicerídeos/biossíntese , Feminino , Humanos , Proteínas Oncogênicas Virais/biossíntese , Neoplasias Ovarianas/tratamento farmacológico , Fosfatidilinositóis/biossíntese , Proteínas Tirosina Quinases/química , Receptor ErbB-2 , Transdução de Sinais , Células Tumorais Cultivadas/metabolismo
14.
J Biol Chem ; 267(29): 21172-8, 1992 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-1328224

RESUMO

An expression plasmid encoding the human 75-kDa tumor necrosis factor (TNF) type 2 receptor (TNF-R2) was constructed and used to generate a stable human cell line (293/TNF-R2) overexpressing TNF-R2. Ligand binding analysis revealed high affinity binding (Kd = 0.2 nM) with approximately 94,000 +/- 7,500 sites/cell for 125I-TNF-alpha and approximately 5-fold lower affinity for TNF-beta (Kd = 1.1 nM) with 264,000 +/- 2,000 sites/cell. Cross-linking of 125I-TNF-alpha and 125I-TNF-beta to 293/TNF-R2 cells yielded predominant complexes with apparent molecular weights of 211,000 for TNF-alpha and 205,000 and 244,000 for TNF-beta, suggesting these complexes contain two or three TNF-R2 molecules. Immunoprecipitation of TNF-R2 from 32P-labeled 293/TNF-R2 cells demonstrated that the receptor is phosphorylated. The majority (97%) of 32Pi incorporation was found in serine residues with a very low level of incorporation (3%) in threonine residues. TNF-alpha treatment of 293/TNF-R2 cells did not significantly affect the degree or pattern of phosphorylation. Cell surface-bound 125I-TNF-alpha was slowly internalized by the 293/TNF-R2 cell line with a t1/2 = 25 min. Shedding of the extracellular domain of TNF-R2 was induced by 4 beta-phorbol 12-myristate 13-acetate but not by TNF-alpha or TNF-beta.


Assuntos
Receptores de Superfície Celular/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Cromatografia de Afinidade , Biblioteca Gênica , Humanos , Cinética , Linfotoxina-alfa/farmacologia , Peso Molecular , Fosforilação , Plasmídeos , RNA Mensageiro/genética , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/isolamento & purificação , Receptores do Fator de Necrose Tumoral , Proteínas Recombinantes/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Transfecção , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA