Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2118: 111-120, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32152974

RESUMEN

Thrombin, a major protein involved in the clotting cascade by the conversion of inactive fibrinogen to fibrin, plays a crucial role in the development of thrombosis. Antithrombin nanoparticles enable site-specific anticoagulation without increasing bleeding risk. Here we outline the process of making and the characterization of bivalirudin and D-phenylalanyl-L-prolyl-L-arginyl-chloromethyl ketone (PPACK) nanoparticles. Additionally, the characterization of these nanoparticles, including particle size, zeta potential, and quantification of PPACK/bivalirudin loading, is also described.


Asunto(s)
Clorometilcetonas de Aminoácidos/síntesis química , Antitrombinas/síntesis química , Fluorocarburos/química , Hirudinas/síntesis química , Fragmentos de Péptidos/síntesis química , Clorometilcetonas de Aminoácidos/química , Clorometilcetonas de Aminoácidos/farmacología , Antitrombinas/química , Antitrombinas/farmacología , Hirudinas/química , Hirudinas/farmacología , Nanopartículas Magnéticas de Óxido de Hierro , Nanopartículas , Tamaño de la Partícula , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Polihidroxietil Metacrilato , Proteínas Recombinantes/síntesis química , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacología
2.
Angew Chem Int Ed Engl ; 56(27): 7803-7807, 2017 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-28504858

RESUMEN

We previously reported an efficient peptide synthesis method, AJIPHASE®, that comprises repeated reactions and isolations by precipitation. This method utilizes an anchor molecule with long-chain alkyl groups as a protecting group for the C-terminus. To further improve this method, we developed a one-pot synthesis of a peptide sequence wherein the synthetic intermediates were isolated by solvent extraction instead of precipitation. A branched-chain anchor molecule was used in the new process, significantly enhancing the solubility of long peptides and the operational efficiency compared with the previous method, which employed precipitation for isolation and a straight-chain aliphatic group. Another prerequisite for this solvent-extraction-based strategy was the use of thiomalic acid and DBU for Fmoc deprotection, which facilitates the removal of byproducts, such as the fulvene adduct.


Asunto(s)
Técnicas de Química Sintética/métodos , Péptidos/síntesis química , Secuencia de Aminoácidos , Cromatografía Líquida de Alta Presión , Fluorenos/química , Hirudinas/análisis , Hirudinas/síntesis química , Hirudinas/química , Fragmentos de Péptidos/análisis , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/química , Péptidos/química , Proteínas Recombinantes/análisis , Proteínas Recombinantes/síntesis química , Proteínas Recombinantes/química , Solubilidad , Solventes/química
3.
Acc Chem Res ; 48(8): 2251-61, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26196117

RESUMEN

Post-translational modification of proteins plays critical roles in regulating structure, stability, localization, and function. Sulfation of the phenolic side chain of tyrosine residues to form sulfotyrosine (sTyr) is a widespread modification of extracellular and integral membrane proteins, influencing the activities of these proteins in cellular adhesion, blood clotting, inflammatory responses, and pathogen infection. Tyrosine sulfation commonly occurs in sequences containing clusters of tyrosine residues and is incomplete at each site, resulting in heterogeneous mixtures of sulfoforms. Purification of individual sulfoforms is typically impractical. Therefore, the most promising approach to elucidate the influence of sulfation at each site is to prepare homogeneously sulfated proteins (or peptides) synthetically. This Account describes our recent progress in both development of such synthetic approaches and application of the resulting sulfopeptides and sulfoproteins to characterize the functional consequences of tyrosine sulfation. Initial synthetic studies used a cassette-based solid-phase peptide synthesis (SPPS) approach in which the side chain sulfate ester was protected to enable it to withstand Fmoc-based SPPS conditions. Subsequently, to address the need for efficient access to multiple sulfoforms of the same peptide, we developed a divergent solid-phase synthetic approach utilizing orthogonally side chain protected tyrosine residues. Using this methodology, we have carried out orthogonal deprotection and sulfation of up to three tyrosine residues within a given sequence, allowing access to all eight sulfoforms of a given target from a single solid-phase synthesis. With homogeneously sulfated peptides in hand, we have been able to probe the influence of tyrosine sulfation on biochemical function. Several of these studies focused on sulfated fragments of chemokine receptors, key mediators of leukocyte trafficking and inflammation. For the receptor CCR3, we showed that tyrosine sulfation enhances affinity and selectivity for binding to chemokine ligands, and we determined the structural basis of these affinity enhancements by NMR spectroscopy. Using a library of CCR5 sulfopeptides, we demonstrated the critical importance of sulfation at one specific site for supporting HIV-1 infection. Demonstrating the feasibility of producing homogeneously tyrosine-sulfated proteins, in addition to smaller peptides, we have used SPPS and native chemical ligation methods to synthesize the leech-derived antithrombotic protein hirudin P6, containing both tyrosine sulfation and glycosylation. Sulfation greatly enhanced inhibitory activity against thrombin, whereas addition of glycans to the sulfated protein decreased inhibition, indicating functional interplay between different post-translational modifications. In addition, the success of the ligation approach suggests that larger sulfoproteins could potentially be obtained by ligation of synthetic sulfopeptides to expressed proteins, using intein-based technology.


Asunto(s)
Péptidos/química , Sulfatos/química , Tirosina/análogos & derivados , Secuencia de Aminoácidos , Sitios de Unión , Infecciones por VIH/patología , VIH-1/fisiología , Hirudinas/síntesis química , Hirudinas/química , Humanos , Espectroscopía de Resonancia Magnética , Datos de Secuencia Molecular , Péptidos/síntesis química , Procesamiento Proteico-Postraduccional , Receptores CCR3/química , Receptores CCR3/metabolismo , Receptores CCR5/química , Receptores CCR5/metabolismo , Técnicas de Síntesis en Fase Sólida , Tirosina/química
4.
Biomater Sci ; 3(1): 41-5, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25589953

RESUMEN

MMP9-responsive bivalirudin-HPMA copolymers were synthesized for direct, local administration in rat spinal cord contusion injury models. Polymer-conjugated bivalirudin peptides maintained activity while demonstrating enzyme-mediated release upon MMP9 exposure and prolonged release from hyaluronic acid/methylcellulose (HAMC) hydrogels compared to free bivalirudin peptide. Localized administration of bivalirudin copolymers in vivo at the site of rat spinal cord injury decreased cellular proliferation and astrogliosis, suggesting the bivalirudin copolymer and HAMC hydrogel system are a promising therapeutic intervention for reducing immediate inflammatory responses and long term scarring.


Asunto(s)
Hirudinas/síntesis química , Ácido Hialurónico/química , Hidrogel de Polietilenoglicol-Dimetacrilato/uso terapéutico , Metaloproteinasa 9 de la Matriz/química , Metilcelulosa/química , Metilcelulosa/uso terapéutico , Fragmentos de Péptidos/síntesis química , Traumatismos de la Médula Espinal/tratamiento farmacológico , Trombina/agonistas , Animales , Hirudinas/química , Ácido Hialurónico/uso terapéutico , Hidrogeles/química , Hidrogeles/uso terapéutico , Metaloproteinasa 9 de la Matriz/metabolismo , Fragmentos de Péptidos/química , Ratas , Proteínas Recombinantes/síntesis química , Proteínas Recombinantes/química , Trombina/química
5.
Angew Chem Int Ed Engl ; 53(15): 3947-51, 2014 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-24615823

RESUMEN

Hirudin P6 is a leech-derived anti-thrombotic protein which possesses two post-translational modifications, O-glycosylation and tyrosine sulfation. In this study we report the ligation-based synthesis of a library of hirudin P6 proteins possessing homogeneous glycosylation and sulfation modifications. The nature of the modifications incorporated was shown to have a drastic effect on inhibition against both the fibrinogenolytic and amidolytic activities of thrombin and thus highlights a potential means for attenuating the biological activity of the protein.


Asunto(s)
Hirudinas/síntesis química , Procesamiento Proteico-Postraduccional/fisiología , Animales , Glicoproteínas , Glicosilación , Hirudinas/química , Estructura Molecular
6.
Protein Pept Lett ; 21(1): 69-74, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24354772

RESUMEN

The RGD sequence was used to design potent hirudin isoform 3 mimetic peptides with both antithrombin activity and antiplatelet aggregation activity. The RGD and proline were inserted between the catalytic active binding domain (D-Phe-Pro-Arg-Pro) on the N-terminus and the anion-binding exosite binding domain (QGDFEPIPEDAYDE) on the Cterminus. Thrombin titration assay and ATP-induced platelet aggregation test revealed that the peptide with the linker RGDWP or RGDGP possessed potent antithrombin and antiplatelet activities, while other peptides without the Pro residue in the linker only showed antithrombin activity. Similar results were obtained in the RGD-containing hirulog-1 variants. Our study indicates that the inserted Pro residue facilitates the exposure of RGD and the binding of the peptide to glycoprotein IIb/IIIa (GPIIb/IIIa). The strategy of combining the RGD sequence and the Pro residue may be used for future designs of bifunctional antithrombotic agents.


Asunto(s)
Proteínas Antitrombina/síntesis química , Antitrombinas/síntesis química , Hirudinas/química , Fragmentos de Péptidos/química , Inhibidores de Agregación Plaquetaria/síntesis química , Animales , Antitrombinas/química , Dominio Catalítico , Hirudinas/síntesis química , Integrina beta3/metabolismo , Masculino , Oligopéptidos , Fragmentos de Péptidos/síntesis química , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Isoformas de Proteínas/síntesis química , Isoformas de Proteínas/química , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/síntesis química , Proteínas Recombinantes/química , Tiempo de Coagulación de la Sangre Total
7.
BMC Biochem ; 14: 6, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23496873

RESUMEN

BACKGROUND: Heparin cofactor II (HCII) is a circulating protease inhibitor, one which contains an N-terminal acidic extension (HCII 1-75) unique within the serpin superfamily. Deletion of HCII 1-75 greatly reduces the ability of glycosaminoglycans (GAGs) to accelerate the inhibition of thrombin, and abrogates HCII binding to thrombin exosite 1. While a minor portion of HCII 1-75 can be visualized in a crystallized HCII-thrombin S195A complex, the role of the rest of the extension is not well understood and the affinity of the HCII 1-75 interaction has not been quantitatively characterized. To address these issues, we expressed HCII 1-75 as a small, N-terminally hexahistidine-tagged polypeptide in E. coli. RESULTS: Immobilized purified HCII 1-75 bound active α-thrombin and active-site inhibited FPR-ck- or S195A-thrombin, but not exosite-1-disrupted γT-thrombin, in microtiter plate assays. Biotinylated HCII 1-75 immobilized on streptavidin chips bound α-thrombin and FPR-ck-thrombin with similar KD values of 330-340 nM. HCII 1-75 competed thrombin binding to chip-immobilized HCII 1-75 more effectively than HCII 54-75 but less effectively than the C-terminal dodecapeptide of hirudin (mean Ki values of 2.6, 8.5, and 0.29 µM, respectively). This superiority over HCII 54-75 was also demonstrated in plasma clotting assays and in competing the heparin-catalysed inhibition of thrombin by plasma-derived HCII; HCII 1-53 had no effect in either assay. Molecular modelling of HCII 1-75 correctly predicted those portions of the acidic extension that had been previously visualized in crystal structures, and suggested that an α-helix found between residues 26 and 36 stabilizes one found between residues 61-67. The latter region has been previously shown by deletion mutagenesis and crystallography to play a crucial role in the binding of HCII to thrombin exosite 1. CONCLUSIONS: Assuming that the KD value for HCII 1-75 of 330-340 nM faithfully predicts that of this region in intact HCII, and that 1-75 binding to exosite 1 is GAG-dependent, our results support a model in which thrombin first binds to GAGs, followed by HCII addition to the ternary complex and release of HCII 1-75 for exosite 1 binding and serpin mechanism inhibition. They further suggest that, in isolated or transferred form, the entire HCII 1-75 region is required to ensure maximal binding of thrombin exosite 1.


Asunto(s)
Cofactor II de Heparina/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Sitios de Unión , Escherichia coli/metabolismo , Cofactor II de Heparina/química , Cofactor II de Heparina/genética , Hirudinas/síntesis química , Hirudinas/química , Hirudinas/metabolismo , Histidina/genética , Histidina/metabolismo , Humanos , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/metabolismo , Cinética , Ratones , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Oligopéptidos/genética , Oligopéptidos/metabolismo , Péptidos/síntesis química , Péptidos/química , Unión Proteica , Estructura Terciaria de Proteína , Conejos , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Alineación de Secuencia , Serpinas/química , Serpinas/metabolismo , Trombina/química , Trombina/metabolismo
9.
Protein Sci ; 15(5): 976-86, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16641485

RESUMEN

3-Nitrotyrosine (NT) is approximately 10(3)-fold more acidic than Tyr, and its absorption properties are strongly pH-dependent. NT absorbs radiation in the wavelength range where Tyr and Trp emit fluorescence (300-450 nm), and it is essentially nonfluorescent. Therefore, NT may function as an energy acceptor in resonance energy transfer (FRET) studies for investigating ligand protein interactions. Here, the potentialities of NT were tested on the hirudin thrombin system, a well-characterized protease inhibitor pair of key pharmacological importance. We synthesized two analogs of the N-terminal domain (residues 1-47) of hirudin: Y3NT, in which Tyr3 was replaced by NT, and S2R/Y3NT, containing the substitutions Ser2 --> Arg and Tyr3 --> NT. The binding of these analogs to thrombin was investigated at pH 8 by FRET and UV/Vis-absorption spectroscopy. Upon hirudin binding, the fluorescence of thrombin was reduced by approximately 50%, due to the energy transfer occurring between the Trp residues of the enzyme (i.e., the donors) and the single NT of the inhibitor (i.e., the acceptor). The changes in the absorption spectra of the enzyme inhibitor complex indicate that the phenate moiety of NT in the free state becomes protonated to phenol in the thrombin-bound form. Our results indicate that the incorporation of NT can be effectively used to detect protein protein interactions with sensitivity in the low nanomolar range, to uncover subtle structural features at the ligand protein interface, and to obtain reliable Kd values for structure activity relationship studies. Furthermore, advances in chemical and genetic methods, useful for incorporating noncoded amino acids into proteins, highlight the broad applicability of NT in biotechnology and pharmacological screening.


Asunto(s)
Hirudinas/química , Hirudinas/síntesis química , Trombina/química , Tirosina/análogos & derivados , Sustitución de Aminoácidos , Sitios de Unión , Transferencia de Energía , Fluorescencia , Hirudinas/metabolismo , Modelos Moleculares , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas , Relación Estructura-Actividad , Trombina/metabolismo , Tirosina/farmacología
10.
J Pept Sci ; 12(2): 116-23, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15952245

RESUMEN

Hirudin variant 1 (HV1), a small protein consisting of 65 amino acids and three disulfide bonds, was synthesized by using Fmoc-based convergent methods on 2-chlorotrityl resin (CLTR). The linear sequence was assembled by the sequential condensation of 7 protected fragments, on the resin-bound 55-65 fragment. The conditions of fragment assembly were carefully studied to determine the most efficient synthetic protocol. Crude reduced [Cys(16, 28)(Acm)]-HV1 thus obtained was easily purified to homogeneity by RP-HPLC. Disulfide bridges were successfully formed by a two-step procedure, involving an oxidative folding step to form Cys(6)-Cys(14) and Cys(22)-Cys(39) linkages, followed by iodine oxidation to form the Cys(16)-Cys(28) bond. The correct disulfide bond alignment was established by peptide mapping using Staphylococcus aureus V8 protease at pH 4.5.


Asunto(s)
Hirudinas/síntesis química , Secuencia de Aminoácidos , Cromatografía Líquida de Alta Presión , Datos de Secuencia Molecular , Oxidación-Reducción , Fragmentos de Péptidos/química
11.
Biochemistry ; 41(46): 13556-69, 2002 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-12427016

RESUMEN

Thrombin is a primary target for the development of novel anticoagulants, since it plays two important and opposite roles in hemostasis: procoagulant and anticoagulant. All thrombin functions are influenced by Na+ binding, which triggers the transition of this enzyme from an anticoagulant (slow) form to a procoagulant (fast) form. In previous studies, we have conveniently produced by chemical synthesis analogues of the N-terminal fragment 1-47 of hirudin HM2 containing noncoded amino acids and displaying up to approximately 2700-fold more potent antithrombin activity, comparable to that of full-length hirudin. In the work presented here, we have exploited the versatility of chemical synthesis to probe the structural and energetic properties of the S3 site of thrombin through perturbations introduced in the structure of hirudin fragment 1-47. In particular, we have investigated the effects of systematic replacement of Tyr3 with noncoded amino acids retaining the aromatic nucleus of Tyr, as well as similar hydrophobic and steric properties, but possessing different electronic (e.g., p-fluoro-, p-iodo-, or p-nitro-Phe), charge (p-aminomethyl-Phe), or conformational (homo-Phe) properties. Our results indicate that the affinity of fragment 1-47 for thrombin is proportional to the desolvation free energy change upon complex formation, and is inversely related to the electric dipole moment of the amino acid side chain at position 3 of hirudin. In this study, we have also identified the key features that are responsible for the preferential binding of hirudin to the procoagulant (fast) form of thrombin. Strikingly, shaving at position 3, by Tyr --> Ala exchange, abolishes the differences in the affinity for thrombin allosteric forms, whereas a bulkier side chain (e.g., beta-naphthylalanine) improves binding preferentially to the fast form. These results provide strong, albeit indirect, evidence that the procoagulant (fast) form of thrombin is in a more open and accessible conformation with respect to the less forgiving structure it acquires in the slow form. This view is also supported by the results of molecular dynamics simulations conducted for 18 ns on free thrombin in full explicit water, showing that after approximately 5 ns thrombin undergoes a significant conformational transition, from a more open conformation (which we propose can be related to the fast form) to a more compact and closed one (which we propose can be related to the slow form). This transition mainly involves the Trp148 and Trp60D loop, the S3 site, and the fibrinogen binding site, whereas the S1 site, the Na+-binding site, and the catalytic pocket remain essentially unchanged. In particular, our data indicate that the S3 site of the enzyme is less accessible to water in the putative slow form. This structural picture provides a reasonable molecular explanation for the fact that physiological substrates related to the procoagulant activity of thrombin (fibrinogen, thrombin receptor 1, and factor XIII) orient a bulky side chain into the S3 site of the enzyme. Taken together, our results can have important implications for the design of novel thrombin inhibitors, of practical utility in the treatment of coagulative disorders.


Asunto(s)
Aminoácidos/metabolismo , Hirudinas/síntesis química , Hirudinas/metabolismo , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/metabolismo , Trombina/metabolismo , Aminoácidos/química , Antitrombinas/farmacología , Dicroismo Circular , Simulación por Computador , Hirudinas/química , Humanos , Cinética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Fragmentos de Péptidos/química , Unión Proteica , Conformación Proteica , Termodinámica , Trombina/antagonistas & inhibidores , Trombina/química
12.
Semin Thromb Hemost ; 27(4): 357-72, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11547358

RESUMEN

This article describes the expression of the hirudin gene heterologously in the methylotrophic yeast Hansenula polymorpha, the establishment of an industrial-scale production process and the subsequent clinical development of polyethylene glycol (PEG)-hirudin. PEGylation increases the molecular weight of hirudin, thereby reducing its kidney filtration rate and immunogenicity and increasing its half-life in the circulation.


Asunto(s)
Hirudinas/síntesis química , Microbiología Industrial , Pichia/genética , Animales , Antitrombinas/síntesis química , Antitrombinas/genética , Antitrombinas/uso terapéutico , Ensayos Clínicos como Asunto , Terapia con Hirudina , Hirudinas/análogos & derivados , Hirudinas/genética , Humanos , Farmacocinética , Transformación Genética
13.
Biochemistry ; 37(39): 13507-15, 1998 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-9753436

RESUMEN

Hirudin is the most potent and specific inhibitor of thrombin, a key enzyme in the coagulation process existing in equilibrium between its procoagulant (fast) and anticoagulant (slow) form. In a previous study, we described the solid-phase synthesis of a Trp3 analogue of fragment 1-47 of hirudin HM2, which displayed approximately 5-fold higher thrombin inhibitory potency relative to that of the natural product [De Filippis, V., et al. (1995) Biochemistry 34, 9552-9564]. By combining automated and manual peptide synthesis, here we have produced in high yields seven analogues of fragment 1-47 containing natural and non-natural amino acids. In particular, we have replaced Val1 with tert-butylglycine (tBug), Ser2 with Arg, and Tyr3 with Phe, cyclohexylalanine (Cha), Trp, alpha-naphthylalanine (alphaNal), and beta-naphthylalanine (betaNal). The crude reduced peptides are able to fold almost quantitatively into the disulfide-cross-linked species, whose unique alignment (Cys6-Cys14, Cys16-Cys28, and Cys22-Cys37) has been shown to be identical to that of the natural fragment. The results of conformational characterization provide evidence that synthetic peptides retain the structural features of the natural species, whereas thrombin inhibition data indicate that the synthetic analogues are all more potent inhibitors of thrombin. In particular, Val --> tBug exchange leads to a 3-fold increase in binding, interpreted as arising from a favorable reduction of the entropy of binding, due to the presence of the more symmetric side chain of tBug relative to that of Val. The S2R analogue binds 24- and 125-fold more tightly than the natural fragment to the fast or slow form of thrombin. These results are explained by considering that Arg2 may favorably couple to Glu192, a key residue involved in the slow to fast transition, thus stabilizing the slow form. Replacement of Tyr3 with more hydrophobic residues having different side chain orientations and electronic structures improves binding by 2-40-fold, suggesting that nonpolar interactions and shape-dependent packing effects strongly influence binding at this position. Overall, these results provide new insights for elucidating the mechanism of hirudin-thrombin recognition at the molecular level and highlight new strategies for designing more potent and selective inhibitors of thrombin.


Asunto(s)
Sustitución de Aminoácidos , Hirudinas/química , Hirudinas/síntesis química , Fragmentos de Péptidos/química , Fragmentos de Péptidos/síntesis química , Alanina/análogos & derivados , Alanina/química , Secuencia de Aminoácidos , Animales , Arginina/química , Bovinos , Dicroismo Circular , Glicina/análogos & derivados , Glicina/química , Hirudinas/farmacología , Humanos , Datos de Secuencia Molecular , Fragmentos de Péptidos/farmacología , Fenilalanina/análogos & derivados , Fenilalanina/química , Serina/química , Espectrometría de Fluorescencia , Trombina/antagonistas & inhibidores , Tirosina/química , Valina/química
14.
J Med Chem ; 39(10): 2008-17, 1996 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-8642559

RESUMEN

We describe here the design, synthesis, and activity of a novel class of alpha-thrombin inhibitors named hirunorms. They were rationally designed to interact through their N-terminal end with the alpha-thrombin active site in a nonsubstrate mode and to specifically bind the fibrinogen recognition exosite. An appropriate spacer that is able to properly orient the N-terminal end in the active site was also selected. This spacer allowed the size of the inhibitors to be reduced to about one-third of the amino acid residues in the hirudin sequence. Hirunorms specifically inhibit the amidolytic action of human alpha-thrombin toward a small chromogenic substrate. The most active compounds of the series, hirunorms IV and V, inhibit alpha-thrombin catalyzed hydrolysis of Tos-Gly-Pro-Arg-p-nitroanilide with K(i) = 0.09 and K(i) = 0.21 nM, respectively. Comparison of the anticoagulant properties of hirunorms, natural hirudin from the European leech Hirudo medicinalis, and the synthetic analog hirulog-1 revealed that hirunorm IV is about 10-fold and 3-fold more active, on a molar base, than hirudin and hirulog-1 in increasing the aPTT, PT, and TT of normal human plasma. The peculiar structure of hirunorms makes them stable to the amidolytic action of thrombin without the introduction of any peptide bond modification. These molecules display long-lasting activity in human plasma, due to the presence of several unnatural amino acids in susceptible positions. Hirunorms are potential candidates for injectable anticoagulants, due to their potency, specificity of action, long-lasting activity, and safety profiles.


Asunto(s)
Hirudinas/química , Trombina/antagonistas & inhibidores , Amidas/metabolismo , Secuencia de Aminoácidos , Coagulación Sanguínea/efectos de los fármacos , Catálisis , Hirudinas/síntesis química , Hirudinas/farmacología , Humanos , Datos de Secuencia Molecular , Estructura Molecular , Trombina/metabolismo
15.
Eur J Pharmacol ; 302(1-3): 69-77, 1996 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-8790994

RESUMEN

The recombinant bifunctional urokinase variant, M23 (rscu-PA-40 kDA/Hir), comprising the kringle and protease domain of single-chain urokinase-type plasminogen activator and a C-terminal fragment of hirudin in one single-chain molecule, was evaluated for its thrombin-inhibitory and fibrinolytic properties in vitro and in vivo. M23 inhibited thrombin-activated coagulation of human blood and thrombin-induced aggregation of human platelet rich plasma in a concentration-dependent manner. The ADP-induced aggregation of human platelet rich plasma was not influenced by M23. In contrast, recombinant single-chain urokinase-type plasminogen activator (saruplase) inhibited neither blood coagulation nor platelet rich plasma aggregation. M23 and saruplase both lysed radiolabelled human thrombi immersed in human plasma (Chandler Loop system) with equal potency. However, there was a significantly lower systemic generation of plasmin (measured as consumption of alpha 2-antiplasmin) by M23 compared to saruplase. In anaesthetized non-heparinized rabbits, experimental femoral artery thrombosis was treated with intravenous bolus injections of M23 or saruplase (6 mg/kg, each). Thrombolytic restoration of arterial blood perfusion was significantly higher in M23- than in saruplase-treated rabbits. Plasma fibrinogen concentrations were decreased markedly in saruplase-treated animals, but remained at significantly higher levels in M23-treated rabbits. In conclusion, the bifunctional molecule, M23, showed thrombin inhibitory and fibrinolytic properties in human in vitro systems and exerted superior thrombolytic effects to saruplase in rabbit femoral artery thrombosis. In vitro and in vivo data indicate that the fibrinolytic activity of M23 is highly clot-specific.


Asunto(s)
Anticoagulantes/uso terapéutico , Arteria Femoral , Fibrinólisis/efectos de los fármacos , Fibrinolíticos/administración & dosificación , Heparina/uso terapéutico , Terapia con Hirudina , Inhibidores de Agregación Plaquetaria/farmacología , Trombosis/tratamiento farmacológico , Activador de Plasminógeno de Tipo Uroquinasa/administración & dosificación , Activador de Plasminógeno de Tipo Uroquinasa/uso terapéutico , Animales , Antitrombinas/uso terapéutico , Quimioterapia Combinada , Hirudinas/síntesis química , Humanos , Masculino , Conejos , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/síntesis química , Proteínas Recombinantes/uso terapéutico , Trombosis/inducido químicamente , Trombosis/fisiopatología , Activador de Plasminógeno de Tipo Uroquinasa/síntesis química
16.
Thromb Haemost ; 71(4): 481-7, 1994 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8052967

RESUMEN

In a continuing effort to create an agent which has both thrombolytic and antithrombotic properties, streptokinase (SK) was covalently bound to the potent antithrombin agent recombinant hirudin (rHir). Linkage of SK to 125I-rHir was accomplished via heterobifunctional crosslinkers in an average molar ratio of 1:1. The 125I-rHir-SK complex was purified from starting components by anion exchange and gel filtration chromatography. The major band containing covalently bound 125I-rHir had a molecular weight of 53 kDa as determined by SDS-PAGE and autoradiography. Biologic activity of each component was then assayed utilizing the chromogenic substrate for each compound. Complex bound 125I-rHir exhibited a 1.2 fold decrease in thrombin inhibition when compared to concentrations of 125I-rHir greater than 3.13 nM. Complex bound 125I-SK, replacing the 125I label on rHir, displayed a 7.9-fold loss in plasminogen activation when compared to 125I-SK. These chromogenic assay results were not adversely altered in the presence of the converse compound's substrate. The 125I-SK-rHir complex (examined at various concentrations) also demonstrated a 0.17- to 17-fold greater affinity for thrombin immobilized onto Sepharose beads as compared to 125I-SK. These findings indicate the rHir-SK complex maintained both thrombolytic and antithrombin properties while also obtaining affinity for immobilized thrombin.


Asunto(s)
Fibrinolíticos/farmacología , Hirudinas/farmacología , Sitios de Unión , Compuestos Cromogénicos/metabolismo , Reactivos de Enlaces Cruzados , Dipéptidos/metabolismo , Fibrinolíticos/síntesis química , Hirudinas/síntesis química , Humanos , Oligopéptidos/metabolismo , Plasminógeno/metabolismo , Unión Proteica , Especificidad por Sustrato , Trombina/antagonistas & inhibidores , Trombina/metabolismo
17.
Thromb Haemost ; 71(3): 320-4, 1994 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8029796

RESUMEN

The specific thrombin inhibitors r-hirudin and a synthetic peptide (I) D-FPRP(G)4-NGDFEEIPEEYL were compared in in vitro tests. r-hirudin proved to be the superior compound with respect to inhibition of amidolytic small substrate turnover that is catalysed by soluble and immobilised thrombin as well as to inhibition of fibrinogen activation. In an in vitro clot model significantly higher molar concentrations of peptide I are needed to achieve fibrin bound thrombin inhibition equivalent to that of r-hirudin. Stable complexes consisting of thrombin and hirudin oppose labile complexes containing the synthetic peptide. The latter leads to a regaining of thrombin activity with subsequent additional fibrin accretion. Analyses of the mixtures of thrombin and peptide I display a time dependent release of amino-terminal D-FPR peptide (III) exhibiting, similar to the residual fragment (peptide II), only weak inhibitory activity. Peptide I and the carboxy-terminal fragment induce, within a certain concentration range, an increase in thrombin activity and clot growth.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Hirudinas/análogos & derivados , Hirudinas/farmacología , Fragmentos de Péptidos/farmacología , Trombina/antagonistas & inhibidores , Secuencia de Aminoácidos , Fibrina/metabolismo , Hirudinas/síntesis química , Humanos , Datos de Secuencia Molecular , Fragmentos de Péptidos/síntesis química , Proteínas Recombinantes/síntesis química , Proteínas Recombinantes/farmacología , Relación Estructura-Actividad , Trombina/metabolismo , Factores de Tiempo
18.
Biochem Biophys Res Commun ; 177(3): 1049-55, 1991 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-2059196

RESUMEN

Using the natural protein hirudin as a model, a novel class of synthetic peptide inhibitors were recently designed. These inhibitors, 'hirulogs', retain the carboxy terminal Hir53-64 domain that interacts with the anion binding exosite of thrombin, connected via an oligoglycyl spacer unit to a catalytic site-directed moiety modeled on the sequence [D]Phe-Pro-Arg-X. The scissile Arg-X bond bond of substrate-like inhibitors has been modified to the proteolytic-resistant functions as beta-homo amino acids Arg psi [CH2CONH] X (2) and reduced bond analogues Arg psi [CH2N]X (3). Both classes of compounds demonstrate inhibition of thrombin amidolytic activity, and this active-site inhibition is highly sensitive to the P1' residue X. Thus these hirulog derivatives are resistant to thrombin proteolysis while maintaining substrate-like interactions with the active center. Finally, hirulog derivatives with non-cleavable replacements of the scissile bond are found to be effective anticoagulant agents.


Asunto(s)
Hirudinas/análogos & derivados , Hirudinas/síntesis química , Trombina/antagonistas & inhibidores , Secuencia de Aminoácidos , Hirudinas/farmacología , Humanos , Indicadores y Reactivos , Cinética , Datos de Secuencia Molecular , Oligopéptidos/síntesis química , Oligopéptidos/farmacología , Relación Estructura-Actividad
19.
J Biol Chem ; 266(11): 6866-71, 1991 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-1849894

RESUMEN

Acidic synthetic peptides corresponding to segments of several nonhomologous proteins (hirudin, residues 54-65; heparin cofactor II, residues 54-75; and fibrinogen, residues 410-427 of the gamma B-chain) inhibit thrombin's cleavage of fibrinogen without blocking the enzyme's active site. Here, we examined effects of these peptides on thrombin's cleavage of protein C and small peptides. Activation of protein C by thrombin in the absence of calcium was inhibited by all of the peptides. Maximal inhibition was 60%, and no greater inhibition was produced by higher peptide concentrations. This differed from progressive inhibition of protein C activation by increasing peptide concentrations in the presence of thrombomodulin and calcium. Potencies of the peptides were in the order hirudin-(54-65) greater than heparin cofactor II-(54-75) greater than gamma B-chain-(410-427). Sulfation of the tyrosine residue in hirudin-(54-65) increased its potency about 10-fold, similar to changes in anticlotting activity. The peptides were activators rather than inhibitors of the cleavage of small chromogenic substrates. In the presence of the peptides, the affinity of thrombin for the substrates S-2366 (pyro-Glu-Pro-Arg-4-nitroanilide), Chromozyme TH (tosyl-Gly-Pro-Arg-4-nitroanilide), and S-2251 (D-Val-Leu-Lys-4-nitroanilide) increased 1.5-2-fold with little change in the Vmax of substrate cleavage. Potencies of peptides in these allosteric effects on thrombin was in the same order as for their other effects. The similar actions of these nonhomologous peptides, which are believed to bind to thrombin's anion-binding exosite, suggest that binding of any peptide to this site exerts the same allosteric effect on thrombin's active site. Interactions of these peptides with thrombin may serve as models for regulation of thrombin's interactions with natural substrates and inhibitors.


Asunto(s)
Fibrinógeno/farmacología , Cofactor II de Heparina/farmacología , Hirudinas/farmacología , Fragmentos de Péptidos/farmacología , Trombina/metabolismo , Secuencia de Aminoácidos , Calcio/farmacología , Fibrinógeno/síntesis química , Cofactor II de Heparina/síntesis química , Hirudinas/síntesis química , Humanos , Cinética , Datos de Secuencia Molecular , Fragmentos de Péptidos/síntesis química , Proteína C/metabolismo , Receptores de Superficie Celular/farmacología , Receptores de Trombina , Trombina/antagonistas & inhibidores
20.
Proc Natl Acad Sci U S A ; 88(4): 1177-81, 1991 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-1996320

RESUMEN

To determine the importance of the thrombin substrate recognition exosite for fibrinogen binding in the formation of both arterial and venous thrombi, we evaluated the antithrombotic effects of the tyrosine-sulfated dodecapeptide from residues 53-64 of hirudin (H peptide) in a nonhuman primate model. This peptide was studied because it inhibits thrombin cleavages of fibrinogen by simple competition without blocking enzyme catalytic-site function. When an exteriorized arteriovenous access shunt model was used in baboons (Papio anubis), thrombus formation was induced by placing a thrombogenic device made of (i) a segment of tubing coated covalently with type I collagen, which generated platelet-rich thrombi under arterial flow conditions, and (ii) two subsequent annular regions of flow expansion that produced fibrin-rich thrombi typically associated with venous valves and veins. Thrombus formation was quantified by measurements of 111In-labeled platelet and 125I-labeled fibrinogen deposition in both arterial-flow and venous-flow portions of the device. Continuous infusion of H peptide (0.5, 15, and 75 mg/kg) proximal to the device for 40 min interrupted, in a dose-response fashion, formation of fibrin-rich thrombus in the regions of disturbed flow and generation of fibrinopeptide A. In contrast, H peptide did not inhibit the capacity of platelets to deposit on the collagen surface (P greater than 0.2 at all doses) or to form hemostatic plugs (as assessed by measurements of bleeding time; P greater than 0.1 at all doses). These findings suggest that, by competitive inhibition of fibrinogen binding to thrombin, fibrin-rich venous-type thrombus formation may be selectively prevented. This strategy may be therapeutically attractive for preserving normal platelet function when conventional anticoagulant therapy is contraindicated.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Fibrina/fisiología , Hirudinas/farmacología , Péptidos/farmacología , Trombosis/prevención & control , Animales , Hemostasis/efectos de los fármacos , Hirudinas/síntesis química , Cinética , Papio , Péptidos/síntesis química , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...