Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
J Biomol Struct Dyn ; 41(9): 3717-3727, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-35343865

RESUMO

Thromboembolic diseases are a major cause of mortality in human and the currently available anticoagulants are associated with various drawbacks, therefore the search for anticoagulants that have better safety profile is highly desirable. Compounds that are part of the dietary routine can be modified to possibly increase their anticoagulant potential. We show mannose 2,3,4,5,6-O-pentasulfate (MPS) as a synthetically modified form of mannose that has appreciable anticoagulation properties. An in silico study identified that mannose in sulfated form can bind effectively to the heparin-binding site of antithrombin (ATIII) and heparin cofactor II (HCII). Mannose was sulfated using a simple sulfation strategy-involving triethylamine-sulfur trioxide adduct. HCII and ATIII were purified from human plasma and the binding analysis using fluorometer and isothermal calorimetry showed that MPS binds at a unique site. A thrombin inhibition analysis using the chromogenic substrate showed that MPS partially enhances the activity of HCII. Further an assessment of in vitro blood coagulation assays using human plasma showed that the activated partial thromboplastin time (APTT) and prothrombin time (PT) were prolonged in the presence of MPS. A molecular dynamics simulation analysis of the HCII-MPS complex showed fluctuations in a N-terminal loop and the cofactor binding site of HCII. The results indicate that MPS is a promising lead due to its effect on the in vitro coagulation rate.Communicated by Ramaswamy H. Sarma.


Assuntos
Cofator II da Heparina , Manose , Humanos , Cofator II da Heparina/química , Cofator II da Heparina/metabolismo , Manose/farmacologia , Coagulação Sanguínea , Anticoagulantes/farmacologia , Anticoagulantes/química , Heparina/farmacologia , Antitrombina III/farmacologia , Antitrombina III/fisiologia , Antitrombinas/farmacologia , Trombina/química
2.
IUBMB Life ; 72(10): 2180-2193, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32827448

RESUMO

Heparin cofactor II (HCII) is predominantly expressed in the liver and inhibits thrombin in blood plasma to influence the blood coagulation cascade. Its deficiency is associated with arterial thrombosis. Its cleavage by neutrophil elastase produces fragment that helps in neutrophil chemotaxis in the acute inflammatory response in human. In the present study, we have identified a novel alternatively spliced transcript of the HCII gene in human liver. This novel transcript includes an additional novel region in continuation with exon 3 called exon 3b. Exon 3b acts like an alternate last exon, and hence its inclusion in the transcript due to alternative splicing removes exon 4 and encodes for a different C-terminal region to give a novel protein, HCII-N. MD simulations of HCII-N and three-dimensional structure showed a unique 51 amino acid sequence at the C-terminal having unique RCL-like structure. The HCII-N protein purified from bacterial culture showed a protein migrating at lower molecular weight (MW 55 kDa) as compared to native HCII (MW 66 kDa). A fluorescence-based analysis revealed a more compact structure of HCII-N that was in a more hydrophilic environment. The HCII-N protein, however, showed no inhibitory activity against thrombin. Due to large conformational variation observed in comparison with native HCII, HCII-N may have alternate protease specificity or a non-inhibitory role. Western blot of HCII purified from large plasma volume showed the presence of a low MW 59 kDa band with no thrombin activity. This study provides the first evidence of alternatively spliced novel isoform of the HCII gene.


Assuntos
Cofator II da Heparina/química , Cofator II da Heparina/genética , Cofator II da Heparina/metabolismo , Fígado/metabolismo , Processamento Alternativo , Fator Xa/metabolismo , Humanos , Modelos Moleculares , Simulação de Dinâmica Molecular , Isoformas de Proteínas , Espectrometria de Fluorescência , Trombina/metabolismo , Ativador de Plasminogênio Tecidual/antagonistas & inibidores , Ativador de Plasminogênio Tecidual/metabolismo
3.
Eur J Med Chem ; 154: 133-143, 2018 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-29787913

RESUMO

Fucosylated chondroitin sulfate (FCS), an unusual glycosaminoglycan with fucose side chains, is a promising anticoagulant agent. To assess the effect of its structure on anticoagulant activity, its derivatives with various degrees of fucosylation (DF), molecular weights (Mw) and sulfation patterns were prepared and characterized. Biological tests showed that their APTT (activated partial thromboplastin time) prolonging activity and intrinsic factor Xase complex (factor IXa-VIIIa-Ca2+-PL complex) inhibitory activity were both reduced in FCS derivatives with lower Mw and DF. However, FCSs with DF at least 16% resulted in greater heparin cofactor II (HCII)-dependent thrombin inhibitory activity in response to decreasing DF, and these activities did not depend on Mw (Mw > 5.2 kDa). Solution competition binding assay further suggested that modulating the DF of FCS derivatives might enhance inhibition of thrombin by activating HCII. These findings imply that FCS derivatives with suitable chain length and DF value may be novel anticoagulants by activating HCII.


Assuntos
Anticoagulantes/farmacologia , Sulfatos de Condroitina/farmacologia , Cofator II da Heparina/metabolismo , Trombina/antagonistas & inibidores , Anticoagulantes/química , Anticoagulantes/isolamento & purificação , Sulfatos de Condroitina/química , Sulfatos de Condroitina/isolamento & purificação , Relação Dose-Resposta a Droga , Cofator II da Heparina/química , Humanos , Estrutura Molecular , Relação Estrutura-Atividade , Trombina/metabolismo
4.
ACS Chem Biol ; 10(6): 1485-94, 2015 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-25742429

RESUMO

The structural diversity of natural sulfated glycosaminoglycans (GAGs) presents major promise for discovery of chemical biology tools or therapeutic agents. Yet, few GAGs have been identified so far to exhibit this promise. We reasoned that a simple approach to identify such GAGs is to explore sequences containing rare residues, for example, 2-O-sulfonated glucuronic acid (GlcAp2S). Genetic algorithm-based computational docking and filtering suggested that GlcAp2S containing heparan sulfate (HS) may exhibit highly selective recognition of antithrombin, a key plasma clot regulator. HS containing only GlcAp2S and 2-N-sulfonated glucosamine residues, labeled as HS2S2S, was chemoenzymatically synthesized in just two steps and was found to preferentially bind antithrombin over heparin cofactor II, a closely related serpin. Likewise, HS2S2S directly inhibited thrombin but not factor Xa, a closely related protease. The results show that a HS containing rare GlcAp2S residues exhibits the unusual property of selective antithrombin activation and direct thrombin inhibition. More importantly, HS2S2S is also the first molecule to activate antithrombin nearly as well as the heparin pentasaccharide although being completely devoid of the critical 3-O-sulfonate group. Thus, this work shows that novel functions and mechanisms may be uncovered by studying rare GAG residues/sequences.


Assuntos
Antitrombinas/química , Ácido Glucurônico/química , Glicosaminoglicanos/química , Bibliotecas de Moléculas Pequenas , Algoritmos , Sítios de Ligação , Fator Xa/química , Cofator II da Heparina/antagonistas & inibidores , Cofator II da Heparina/química , Heparitina Sulfato/química , Cinética , Simulação de Acoplamento Molecular , Ligação Proteica
5.
J Biol Chem ; 289(43): 29790-800, 2014 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-25202017

RESUMO

Antithrombin III (ATIII) is a key antiproteinase involved in blood coagulation. Previous investigations have shown that ATIII is degraded by Staphylococcus aureus V8 protease, leading to release of heparin binding fragments derived from its D helix. As heparin binding and antimicrobial activity of peptides frequently overlap, we here set out to explore possible antibacterial effects of intact and degraded ATIII. In contrast to intact ATIII, the results showed that extensive degradation of the molecule yielded fragments with antimicrobial activity. Correspondingly, the heparin-binding, helix D-derived, peptide FFFAKLNCRLYRKANKSSKLV (FFF21) of human ATIII, was found to be antimicrobial against particularly the Gram-negative bacteria Escherichia coli and Pseudomonas aeruginosa. Fluorescence microscopy and electron microscopy studies demonstrated that FFF21 binds to and permeabilizes bacterial membranes. Analogously, FFF21 was found to induce membrane leakage of model anionic liposomes. In vivo, FFF21 significantly reduced P. aeruginosa infection in mice. Additionally, FFF21 displayed anti-endotoxic effects in vitro. Taken together, our results suggest novel roles for ATIII-derived peptide fragments in host defense.


Assuntos
Anti-Infecciosos/química , Anti-Infecciosos/farmacologia , Antitrombina III/química , Antitrombina III/farmacologia , Sequência de Aminoácidos , Animais , Permeabilidade da Membrana Celular/efeitos dos fármacos , Dicroísmo Circular , Modelos Animais de Doenças , Cofator II da Heparina/química , Cofator II da Heparina/farmacologia , Humanos , Lipopolissacarídeos/metabolismo , Lipossomos/metabolismo , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Dados de Sequência Molecular , Elastase Pancreática/metabolismo , Peptídeos/química , Peptídeos/farmacologia , Estrutura Secundária de Proteína , Proteólise/efeitos dos fármacos , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/patologia , Pseudomonas aeruginosa/efeitos dos fármacos , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/metabolismo , Staphylococcus aureus/ultraestrutura
6.
PLoS One ; 9(7): e102577, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25047075

RESUMO

Sepsis and septic shock remain important medical problems with high mortality rates. Today's treatment is based mainly on using antibiotics to target the bacteria, without addressing the systemic inflammatory response, which is a major contributor to mortality in sepsis. Therefore, novel treatment options are urgently needed to counteract these complex sepsis pathologies. Heparin cofactor II (HCII) has recently been shown to be protective against Gram-negative infections. The antimicrobial effects were mapped to helices A and D of the molecule. Here we show that KYE28, a 28 amino acid long peptide representing helix D of HCII, is antimicrobial against the Gram-negative bacteria Escherichia coli and Pseudomonas aeruginosa, the Gram-positive Bacillus subtilis and Staphylococcus aureus, as well as the fungus Candida albicans. Moreover, KYE28 binds to LPS and thereby reduces LPS-induced pro-inflammatory responses by decreasing NF-κB/AP-1 activation in vitro. In mouse models of LPS-induced shock, KYE28 significantly enhanced survival by dampening the pro-inflammatory cytokine response. Finally, in an invasive Pseudomonas infection model, the peptide inhibited bacterial growth and reduced the pro-inflammatory response, which lead to a significant reduction of mortality. In summary, the peptide KYE28, by simultaneously targeting bacteria and LPS-induced pro-inflammatory responses represents a novel therapeutic candidate for invasive infections.


Assuntos
Anti-Infecciosos/uso terapêutico , Cofator II da Heparina/uso terapêutico , Infecções por Pseudomonas/tratamento farmacológico , Pseudomonas aeruginosa/efeitos dos fármacos , Choque Séptico/tratamento farmacológico , Sequência de Aminoácidos , Animais , Anti-Infecciosos/química , Bactérias/efeitos dos fármacos , Candida albicans/efeitos dos fármacos , Linhagem Celular , Endotoxinas/imunologia , Cofator II da Heparina/química , Humanos , Lipopolissacarídeos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Infecções por Pseudomonas/imunologia , Choque Séptico/imunologia , Choque Séptico/microbiologia
7.
Biomacromolecules ; 15(4): 1337-45, 2014 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-24588750

RESUMO

Effects of poly(ethylene glycol) (PEG) conjugation on peptide interactions with lipid membranes and lipopolysaccharide (LPS) were investigated for KYE28 (KYEITTIHNLFRKLTHRLFRRNFGYTLR), an antimicrobial and anti-inflammatory peptide derived from human heparin cofactor II. In particular, effects of PEG length and localization was investigated by ellipsometry, circular dichroism, nanoparticle tracking analysis, and fluorescence/electron microscopy. PEGylation of KYE28 reduces peptide binding to lipid membranes, an effect accentuated at increasing PEG length, but less sensitive to conjugation site. The reduced binding causes suppressed liposome leakage induction, as well as bacterial lysis. As a result of this, the antimicrobial effects of KYE28 is partially lost with increasing PEG length, but hemolysis also strongly suppressed and selecticity improved. Through this, conditions can be found, at which the PEGylated peptide displays simultaneously efficient antimicrobial affects and low hemolysis in blood. Importantly, PEGylation does not markedly affect the anti-inflammatory effects of KYE28. The combination of reduced toxicity, increased selectivity, and retained anti-inflammatory effect after PEGylation, as well as reduced scavenging by serum proteins, thus shows that PEG conjugation may offer opportunities in the development of effective and selective anti-inflammatory peptides.


Assuntos
Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/farmacologia , Lipídeos de Membrana/metabolismo , Polietilenoglicóis/química , Sequência de Aminoácidos , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/farmacologia , Peptídeos Catiônicos Antimicrobianos/metabolismo , Dicroísmo Circular , Hemólise/efeitos dos fármacos , Cofator II da Heparina/química , Interações Hospedeiro-Patógeno , Humanos , Bicamadas Lipídicas/química , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Lipossomos , Testes de Sensibilidade Microbiana , Dados de Sequência Molecular , Nanopartículas
8.
Carbohydr Polym ; 98(2): 1514-23, 2013 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-24053834

RESUMO

Fucosylated glycosaminoglycans (FGs) are complex glycosaminoglycans that exhibit potent anticoagulant activity. To study the relationship between molecular size and biological activity, oligosaccharides with (2,5)-anhydro-D-talose units at new reducing ends were prepared by hydrazine deacetylation and nitrous acid depolymerization. The product chemical structures were analyzed by one- and two-dimensional NMR methods. Additionally, anticoagulant activities were evaluated by clotting assay and chromogenic substrate cleavage. The results demonstrated that under mild deacetylation and deaminative cleavage conditions, both products were relatively homogeneous and sulfated fucose branch types and sulfate substituents remained stable. These depolymerized FGs with different molecular sizes had potent intrinsic anticoagulant activities, which were similar to those that were obtained by free-radical depolymerization with similar molecular weights. Decreasing molecular weight may weaken activity but not significantly affect factor Xase and heparin cofactor II (HCII)-mediated thrombin inhibition.


Assuntos
Anticoagulantes/química , Coagulação Sanguínea/efeitos dos fármacos , Fucose/química , Glicosaminoglicanos/química , Pepinos-do-Mar/química , Animais , Anticoagulantes/isolamento & purificação , Anticoagulantes/farmacologia , Cisteína Endopeptidases/química , Ensaios Enzimáticos , Glicosaminoglicanos/isolamento & purificação , Glicosaminoglicanos/farmacologia , Cofator II da Heparina/química , Humanos , Hidrazinas/química , Lactonas/química , Espectroscopia de Ressonância Magnética , Peso Molecular , Proteínas de Neoplasias/química , Ácido Nitroso/química , Relação Estrutura-Atividade , Trombina/antagonistas & inibidores , Trombina/química
9.
Biochim Biophys Acta ; 1828(11): 2709-19, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23806651

RESUMO

Lipid membrane and lipopolysaccharide (LPS) interactions were investigated for a series of amphiphilic and cationic peptides derived from human heparin cofactor II (HCII), using dual polarization interferometry, ellipsometry, circular dichroism (CD), cryoTEM, and z-potential measurements. Antimicrobial effects of these peptides were compared to their ability to disorder bacterial lipid membranes, while their capacity to block endotoxic effects of LPS was correlated to the binding of these peptides to LPS and its lipid A moiety, and to charge, secondary structure, and morphology of peptide/LPS complexes. While the peptide KYE28 (KYEITTIHNLFRKLTHRLFRRNFGYTLR) displayed potent antimicrobial and anti-endotoxic effects, its truncated variants KYE21 (KYEITTIHNLFRKLTHRLFRR) and NLF20 (NLFRKLTHRLFRRNFGYTLR) provide some clues on structure-activity relations, since KYE21 retains both the antimicrobial and anti-endotoxic effects of KYE28 (although both attenuated), while NLF20 retains the antimicrobial but only a fraction of the anti-endotoxic effect, hence locating the anti-endotoxic effects of KYE28 to its N-terminus. The antimicrobial effect, on the other hand, is primarily located at the C-terminus of KYE28. While displaying quite different endotoxic effects, these peptides bind to a similar extent to both LPS and lipid A, and also induce comparable LPS scavenging on model eukaryotic membranes. In contrast, fragmentation and densification of LPS aggregates, in turn dependent on the secondary structure in the peptide/LPS aggregates, correlate to the anti-endotoxic effect of these peptides, thus identifying peptide-induced packing transitions in LPS aggregates as key for anti-endotoxic functionality. This aspect therefore needs to be taken into account in the development of novel anti-endotoxic peptide therapeutics.


Assuntos
Endotoxinas/antagonistas & inibidores , Cofator II da Heparina/farmacologia , Lipopolissacarídeos/metabolismo , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Linhagem Celular , Dicroísmo Circular , Cofator II da Heparina/química , Humanos , Camundongos , Dados de Sequência Molecular , Peptídeos/química , Espectrometria de Fluorescência , Relação Estrutura-Atividade
10.
BMC Biochem ; 14: 6, 2013 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-23496873

RESUMO

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.


Assuntos
Cofator II da Heparina/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação , Escherichia coli/metabolismo , Cofator II da Heparina/química , Cofator II da Heparina/genética , Hirudinas/síntese química , Hirudinas/química , Hirudinas/metabolismo , Histidina/genética , Histidina/metabolismo , Humanos , Proteínas Imobilizadas/química , Proteínas Imobilizadas/metabolismo , Cinética , Camundongos , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Peptídeos/síntese química , Peptídeos/química , Ligação Proteica , Estrutura Terciária de Proteína , Coelhos , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Alinhamento de Sequência , Serpinas/química , Serpinas/metabolismo , Trombina/química , Trombina/metabolismo
11.
Methods Enzymol ; 501: 105-37, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22078533

RESUMO

Serpins (serine protease inhibitors) have traditionally been grouped together based on structural homology. They share common structural features of primary sequence, but not all serpins require binding to cofactors in order to achieve maximal protease inhibition. In order to obtain physiologically relevant rates of inhibition of target proteases, some serpins utilize the unbranched sulfated polysaccharide chains known as glycosaminoglycans (GAGs) to enhance inhibition. These GAG-binding serpins include antithrombin (AT), heparin cofactor II (HCII), and protein C inhibitor (PCI). The GAGs heparin and heparan sulfate have been shown to bind AT, HCII, and PCI, while HCII is also able to utilize dermatan sulfate as a cofactor. Other serpins such as PAI-1, kallistatin, and α(1)-antitrypsin also interact with GAGs with different endpoints, some accelerating protease inhibition while others inhibit it. There are many serpins that bind or carry ligands that are unrelated to GAGs, which are described elsewhere in this work. For most GAG-binding serpins, binding of the GAG occurs in a conserved region of the serpin near or involving helix D, with the exception of PCI, which utilizes helix H. The binding of GAG to serpin can lead to a conformational change within the serpin, which can lead to increased or tighter binding to the protease, and can accelerate the rates of inhibition up to 10,000-fold compared to the unbound native serpin. In this chapter, we will discuss three major GAG-binding serpins with known physiological roles in modulating coagulation: AT (SERPINC1), HCII (SERPIND1), and PCI (SERPINA5). We will review methodologies implemented to study the structure of these serpins and those used to study their interactions with GAG's. We discuss novel techniques to examine the serpin-GAG interaction and finally we review the biological roles of these serpins by describing the mouse models used to study them.


Assuntos
Antitrombina III/metabolismo , Técnicas de Química Analítica , Cofator II da Heparina/metabolismo , Biologia Molecular/métodos , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Inibidor da Proteína C/metabolismo , alfa 1-Antitripsina/metabolismo , Animais , Antitrombina III/química , Sítios de Ligação , Coagulação Sanguínea , Dermatan Sulfato/química , Dermatan Sulfato/metabolismo , Heparina/química , Heparina/metabolismo , Cofator II da Heparina/química , Humanos , Cinética , Camundongos , Camundongos Knockout , Modelos Moleculares , Inibidor 1 de Ativador de Plasminogênio/química , Ligação Proteica , Inibidor da Proteína C/química , Estrutura Secundária de Proteína , Especificidade da Espécie , Trombina/química , Trombina/metabolismo , alfa 1-Antitripsina/química
12.
Semin Thromb Hemost ; 37(4): 339-48, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21805439

RESUMO

Heparin cofactor II (HCII) is a serine protease inhibitor (serpin) found in high concentrations in human plasma. Despite its discovery >30 years ago, its physiological function is still poorly understood. It is known to inhibit thrombin, the predominant coagulation protease, and HCII-thrombin complexes have been found in plasma, yet it is thought to contribute little to normal hemostasis. However, thrombin has several other physiological functions, and therefore many biological roles for HCII need consideration. The unique structure and mechanism of action of HCII have helped guide our understanding of HCII. In particular, HCII binds many glycosaminoglycans (GAGs) such as heparin and heparin sulfate as well as several different polyanions to enhance its inhibition of thrombin. Distinctly, HCII is able to use the GAG dermatan sulfate for accelerated thrombin inhibition. Dermatan sulfate is found in high concentrations in the walls of blood vessels as well as in placental tissue. This knowledge has led to research indicating that HCII may play a protective role in atherosclerosis and placental thrombosis. Additionally, pharmaceuticals are being developed that use the dermatan sulfate activation of HCII for anticoagulation. Although much research is still needed to fully understand HCII, this humble protein may have significant impact in our medical future. This article reviews the laboratory history, protein characteristics, structure-activity relationships, protease inhibition, physiological function, and medical relevance of HCII in hopes of regenerating interest in this sometimes forgotten serpin.


Assuntos
Cofator II da Heparina/fisiologia , Animais , Cofator II da Heparina/química , Homeostase/fisiologia , Humanos , Doenças Vasculares/sangue , Doenças Vasculares/tratamento farmacológico
13.
J Inherit Metab Dis ; 34(2): 499-508, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21170681

RESUMO

Early detection of mucopolysaccharidosis (MPS) is an important factor in treatment success; therefore, good disease biomarkers are vital. We evaluate heparin cofactor II-thrombin complex (HCII-T) as a biomarker in serum and dried blood spots (DBS) of MPS patients. Serum HCII-T and urine dermatan sulphate:chondroitin sulphate (DS:CS) ratio are also compared longitudinally against clinical outcomes in MPSI, II and VI patients following treatment. Samples were collected from MPS patients at the Royal Manchester Children's Hospital. DS:CS ratio was obtained by measuring the area density of spots from 2D electrophoresis of urinary glycosaminoglycans. Serum and DBS HCII-T was measured by sandwich ELISA. Serum HCII-T is elevated approximately 25-fold in MPS diseases that store DS, clearly distinguishing untreated MPSI, II and VI patients from unaffected age-matched controls. Serum HCII-T is also elevated in MPSIII, which leads to storage of heparan sulphate, with an increase of approximately 4-fold over unaffected age-matched controls. Urine DS:CS ratio and serum HCII-T decrease in response to treatment of MPSI, II and VI patients. HCII-T appears to respond rapidly to perturbations in treatment, whilst DS:CS ratio responds more slowly. HCII-T is a suitable biomarker for MPSI, II and VI, and it may also be informative for MPS diseases storing HS alone, such as MPSIII, although the elevation observed is smaller. In treated MPS patients, HCII-T and DS:CS ratio appear to measure short-term and long-term treatment outcomes, respectively. The potential value of HCII-T measurement in DBS for newborn screening of MPS diseases warrants further investigation.


Assuntos
Sulfatos de Condroitina/química , Dermatan Sulfato/química , Glicosaminoglicanos/metabolismo , Cofator II da Heparina/química , Trombina/química , Animais , Biomarcadores/metabolismo , Criança , Pré-Escolar , Ensaio de Imunoadsorção Enzimática/métodos , Glicosaminoglicanos/urina , Humanos , Lactente , Recém-Nascido , Camundongos , Triagem Neonatal/métodos , Resultado do Tratamento
14.
Anal Biochem ; 406(2): 166-75, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20670608

RESUMO

Irreversible inactivation of alpha-thrombin (T) by the serpin, heparin cofactor II (HCII), is accelerated by ternary complex formation with the glycosaminoglycans (GAGs) heparin and dermatan sulfate (DS). Low expression of human HCII in Escherichia coli was optimized by silent mutation of 27 rare codons and five secondary Shine-Dalgarno sequences in the cDNA. The inhibitory activities of recombinant HCII, and native and deglycosylated plasma HCII, and their affinities for heparin and DS were compared. Recombinant and deglycosylated HCII bound heparin with dissociation constants (K(D)) of 6+/-1 and 7+/-1 microM, respectively, approximately 6-fold tighter than plasma HCII, with K(D) 40+/-4 microM. Binding of recombinant and deglycosylated HCII to DS, both with K(D) 4+/-1 microM, was approximately 4-fold tighter than for plasma HCII, with K(D) 15+/-4 microM. Recombinant HCII, lacking N-glycosylation and tyrosine sulfation, inactivated alpha-thrombin with a 1:1 stoichiometry, similar to plasma HCII. Second-order rate constants for thrombin inactivation by recombinant and deglycosylated HCII were comparable, at optimal GAG concentrations that were lower than those for plasma HCII, consistent with its weaker GAG binding. This weaker binding may be attributed to interference of the Asn(169)N-glycan with the HCII heparin-binding site.


Assuntos
Escherichia coli/metabolismo , Glicosaminoglicanos/metabolismo , Cofator II da Heparina/metabolismo , Sequência de Aminoácidos , Dermatan Sulfato/metabolismo , Ativação Enzimática , Fluorescência , Glicosilação , Cofator II da Heparina/química , Cofator II da Heparina/isolamento & purificação , Humanos , Cinética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Peptídeos/química , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Trombina/metabolismo
15.
J Biol Chem ; 285(11): 8278-89, 2010 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-20053992

RESUMO

Inactivation of thrombin (T) by the serpins heparin cofactor II (HCII) and antithrombin (AT) is accelerated by a heparin template between the serpin and thrombin exosite II. Unlike AT, HCII also uses an allosteric interaction of its NH(2)-terminal segment with exosite I. Sucrose octasulfate (SOS) accelerated thrombin inactivation by HCII but not AT by 2000-fold. SOS bound to two sites on thrombin, with dissociation constants (K(D)) of 10 +/- 4 microm and 400 +/- 300 microm that were not kinetically resolvable, as evidenced by single hyperbolic SOS concentration dependences of the inactivation rate (k(obs)). SOS bound HCII with K(D) 1.45 +/- 0.30 mm, and this binding was tightened in the T.SOS.HCII complex, characterized by K(complex) of approximately 0.20 microm. Inactivation data were incompatible with a model solely depending on HCII.SOS but fit an equilibrium linkage model employing T.SOS binding in the pathway to higher order complex formation. Hirudin-(54-65)(SO(3)(-)) caused a hyperbolic decrease of the inactivation rates, suggesting partial competitive binding of hirudin-(54-65)(SO(3)(-)) and HCII to exosite I. Meizothrombin(des-fragment 1), binding SOS with K(D) = 1600 +/- 300 microm, and thrombin were inactivated at comparable rates, and an exosite II aptamer had no effect on the inactivation, suggesting limited exosite II involvement. SOS accelerated inactivation of meizothrombin 1000-fold, reflecting the contribution of direct exosite I interaction with HCII. Thrombin generation in plasma was suppressed by SOS, both in HCII-dependent and -independent processes. The ex vivo HCII-dependent process may utilize the proposed model and suggests a potential for oversulfated disaccharides in controlling HCII-regulated thrombin generation.


Assuntos
Anticoagulantes/metabolismo , Coagulação Sanguínea/fisiologia , Cofator II da Heparina/metabolismo , Sacarose/análogos & derivados , Trombina/metabolismo , Regulação Alostérica , Anticoagulantes/química , Anticoagulantes/farmacologia , Ligação Competitiva/efeitos dos fármacos , Ligação Competitiva/fisiologia , Coagulação Sanguínea/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Precursores Enzimáticos/metabolismo , Precursores Enzimáticos/farmacologia , Cofator II da Heparina/química , Hirudinas/metabolismo , Hirudinas/farmacologia , Humanos , Cinética , Modelos Químicos , Plasma , Inibidores de Proteases/metabolismo , Inibidores de Proteases/farmacologia , Protrombina/metabolismo , Espectrometria de Fluorescência , Sacarose/química , Sacarose/metabolismo , Sacarose/farmacologia , Trombina/química , Trombina/farmacologia
16.
Glycobiology ; 18(3): 225-34, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18156656

RESUMO

Dermatan sulfate is a glycosaminoglycan that selectively inhibits the action of thrombin through interaction with heparin cofactor II. Unlike heparin it does not interact with other coagulation factors and is able to inhibit thrombin associated with clots. This property has made dermatan sulfate an attractive candidate as an antithrombotic drug. Previous studies have showed that dermatan sulfate derived from porcine/bovine intestinal mucosa/skin or marine invertebrates is capable of stimulating heparin cofactor II-mediated thrombin inhibition in vitro. This biological activity is reported for the first time in this study using dermatan sulfate derived from mammalian tissues other than intestinal mucosa or skin. Ten different bovine tissues including the aorta, diaphragm, eyes, large and small intestine, esophagus, skin, tendon, tongue, and tongue skin were used to prepare dermatan sulfate-enriched fractions by anion exchange chromatography and acetone precipitation. Heparin cofactor II/dermatan sulfate-mediated thrombin inhibition measured in vitro revealed activity comparable to or higher than the commercial standard with 2-fold differences observed between some tissues. Analysis of the extracted dermatan sulfate using fluorophore-assisted carbohydrate electrophoresis revealed significant differences in the relative percentage of all the mono-sulfated disaccharides, in particular the predominant mammalian disaccharide uronic acid-->N-acetyl-D-galactosamine-4-O-sulfate, confirming previous reports regarding variations in sulfation in dermatan sulfate from different tissues. Overall, these findings demonstrate that dermatan sulfate extracted from a range of bovine tissues exhibits in vitro antithrombin activity equivalent to or higher than that observed for porcine intestinal mucosa, identifying additional sources of dermatan sulfate as potential antithrombotic agents.


Assuntos
Antitrombinas/química , Dermatan Sulfato/química , Dissacarídeos/análise , Animais , Antitrombinas/isolamento & purificação , Aorta/química , Bovinos , Dermatan Sulfato/isolamento & purificação , Diafragma/química , Eletroforese em Gel de Ágar , Esôfago/química , Olho/química , Precipitação Fracionada , Cofator II da Heparina/química , Intestino Grosso/química , Intestino Delgado/química , Especificidade de Órgãos , Pele/química , Tendões/química , Língua/química
17.
Carbohydr Res ; 342(15): 2326-30, 2007 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-17631282

RESUMO

The major acidic polysaccharide from the brown alga Laminaria cichorioides is a complex and heterogeneous sulfated fucan. Its preponderant structure is a 2,3-disulfated, 4-linked alpha-fucose unit. The purified polysaccharide has a potent anticoagulant activity, as estimated by APTT assay ( approximately 40 IU/mg), which is mainly mediated by thrombin inhibition by heparin cofactor II. It also accelerates thrombin and factor Xa inhibition by antithrombin but at a lower potency. Sulfated fucan from L. cichorioides is a promising anticoagulant polysaccharide and a possible alternative for an antithrombotic compound due to its preferential heparin cofactor II-dependent activity.


Assuntos
Anticoagulantes/química , Cofator II da Heparina/química , Heparina/química , Laminaria/metabolismo , Phaeophyceae/metabolismo , Animais , Testes de Coagulação Sanguínea , Cromatografia por Troca Iônica/métodos , Relação Dose-Resposta a Droga , Fator Xa/química , Humanos , Espectroscopia de Ressonância Magnética , Polissacarídeos/química , Frações Subcelulares/química , Trombina/química
18.
Biochemistry ; 45(38): 11444-52, 2006 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-16981704

RESUMO

The conversion of the reactive center bond of the serpin alpha1-proteinase inhibitor (alpha1-PI, also known as alpha1-antitrypsin) from Met-Ser to Arg-Ser decreases the rate at which it inhibits neutrophil elastase and endows it with the ability to inhibit thrombin and activated protein C (APC). Another serpin, heparin cofactor II (HCII), contains a unique N-terminal extension that binds thrombin exosite 1. We fused residues 1-75 of HCII to the N-terminus of alpha1-PI M358R, forming an HCII-alpha1-PI chimera (HAPI M358R). It inhibited alpha-thrombin 21-fold faster than alpha1-PI M358R, with second-order rate constants of 2.3 x 10(8) M(-1) min(-1) versus 1.1 x 10(7) M(-1) min(-1), respectively. When gammaT-thrombin, which lacks an intact exosite 1, was substituted for alpha-thrombin, the kinetic advantage of HAPI M358R over alpha1-PI M358R was reduced to 9-fold, whereas APC and trypsin, proteases lacking exosite 1-like regions, were inhibited only 1.3- and 2-fold more rapidly by HAPI M358R than by alpha1-PI M358R, respectively. Maximal enhancement of alpha1-PI M358R activity required the acidic residues found between HCII residues 55 and 75, because no enhancement was observed either by fusion of residues 1-54 alone or by fusion of a mutated HCII acidic extension in which all Glu and Asp residues between positions 55 and 75 were neutralized by mutation. Fusing residues 55-75 to alpha1-PI M358R yielded a relative rate enhancement of only 6-fold, suggesting a need for the full tail region to achieve maximal enhancement. Our results suggest that transfer of the N-terminal acidic extension of HCII to alpha1-PI M358R enhanced its inhibition of thrombin by conferring the ability to bind exosite 1 on HAPI M358R. This enhancement may aid in efforts to tailor this inhibitor for therapeutic use.


Assuntos
Substituição de Aminoácidos , Arginina/química , Cofator II da Heparina/química , Cofator II da Heparina/metabolismo , Metionina/química , Proteínas Recombinantes de Fusão/metabolismo , Trombina/antagonistas & inibidores , alfa 1-Antitripsina/metabolismo , Fibrina/metabolismo , Humanos , Cinética , Modelos Moleculares , Desnaturação Proteica , Solubilidade , Relação Estrutura-Atividade , Termodinâmica , alfa 1-Antitripsina/química
19.
Trends Biochem Sci ; 31(8): 427-35, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16820297

RESUMO

Serpins use an extraordinary mechanism of protease inhibition that depends on a rapid and marked conformational change and causes destruction of the covalently linked protease. Serpins thus provide stoichiometric, irreversible inhibition, and their dependence on conformational change is exploited for signalling and clearance. The regulatory advantages provided by structural mobility are best illustrated by the heparin activation mechanisms of the plasma serpins antithrombin and heparin cofactor II. This mechanistic complexity, however, renders serpins highly susceptible to disease-causing mutations. Recent crystal structures reveal the intricate conformational rearrangements involved in protease inhibition, activity modulation and the unique molecular pathology of the remarkable shape-shifting serpins.


Assuntos
Conformação Proteica , Serpinas/química , Animais , Antitrombinas/química , Antitrombinas/fisiologia , Cofator II da Heparina/química , Cofator II da Heparina/fisiologia , Humanos , Cinética , Modelos Moleculares , Serina Endopeptidases/química , Serina Endopeptidases/metabolismo , Serpinas/fisiologia
20.
Thromb Res ; 117(4): 447-61, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-15869786

RESUMO

INTRODUCTION: Following thrombin cleavage of the reactive centre (P1-P1'; L444-S445) of the serpin heparin cofactor II (HCII), HCII traps thrombin (IIa) in a stable inhibitory complex. To compare HCII to other serpins we substituted: the P13-P5' residues of HCII with those of alpha(1)-proteinase inhibitor (alpha(1)-PI), alpha(1)-PI (M358R), or antithrombin (AT); the P4-P1, P3-P1, and P2-P1 residues of HCII with those of AT; and made L444A/H/K/M or R point mutations. We also combined L444R with changes in the glycosaminoglycan binding domain collectively termed MutD. MATERIALS AND METHODS: Variants were made by site-directed mutagenesis, expressed in bacteria, purified and characterized electrophoretically and kinetically. RESULTS AND CONCLUSIONS: Of the P13-P5' mutants, only the alpha(1)-PI-loop variant retained anti-IIa activity, but less than the corresponding L444M. Heparin-catalyzed rate constants for IIa inhibition were reduced vs. wild-type (WT) by at most three-fold for all P1 mutants save L444A (reduced 20-fold). L444R and L444K inhibited IIa>50- and >6-fold more rapidly than WT in heparin-free reactions, but stoichiometries of inhibition were increased for all variants. HCII-IIa complexes of all P1 variants were stable in the absence of heparin, but those of the L444K and L444R variants released active IIa over time with heparin. Limited proteolysis of these two groups of HCII-IIa complexes produced different fragmentation patterns consistent with conformational differences. The combination of either substituted AT residues at P2, P3, and P4, or the MutD mutations with L444R resulted in complex instability with or without heparin. This is the first description of HCII-IIa complexes of transient stability forming in the absence of heparin, and may explain the extent to which the reactive centre loop of HCII differs from that of AT.


Assuntos
Escherichia coli/enzimologia , Cofator II da Heparina/química , Cofator II da Heparina/metabolismo , Peptídeo Hidrolases/química , Peptídeo Hidrolases/metabolismo , Serpinas/química , Serpinas/metabolismo , Substituição de Aminoácidos , Sítios de Ligação , Estabilidade Enzimática , Escherichia coli/química , Ligação Proteica , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...