Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Med Chem ; 66(6): 3785-3797, 2023 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-36898159

RESUMO

Anticoagulation therapy is a mainstay of the treatment of thrombotic disorders; however, conventional anticoagulants trade antithrombotic benefits for bleeding risk. Factor (f) XI deficiency, known as hemophilia C, rarely causes spontaneous bleeding, suggesting that fXI plays a limited role in hemostasis. In contrast, individuals with congenital fXI deficiency display a reduced incidence of ischemic stroke and venous thromboembolism, indicating that fXI plays a role in thrombosis. For these reasons, there is intense interest in pursuing fXI/factor XIa (fXIa) as targets for achieving antithrombotic benefit with reduced bleeding risk. To obtain selective inhibitors of fXIa, we employed libraries of natural and unnatural amino acids to profile fXIa substrate preferences. We developed chemical tools for investigating fXIa activity, such as substrates, inhibitors, and activity-based probes (ABPs). Finally, we demonstrated that our ABP selectively labels fXIa in the human plasma, making this tool suitable for further studies on the role of fXIa in biological samples.


Assuntos
Fator XIa , Trombose , Humanos , Fibrinolíticos , Hemostasia , Anticoagulantes/farmacologia , Fator XI/metabolismo
2.
Chem Sci ; 13(23): 6813-6829, 2022 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-35774156

RESUMO

Activated protein C (APC), thrombin, and factor (f) Xa are vitamin K-dependent serine proteases that are key factors in blood coagulation. Moreover, they play important roles in inflammation, apoptosis, fibrosis, angiogenesis, and viral infections. Abnormal activity of these coagulation factors has been related to multiple conditions, such as bleeding and thrombosis, Alzheimer's disease, sepsis, multiple sclerosis, and COVID-19. The individual activities of APC, thrombin, and fXa in coagulation and in various diseases are difficult to establish since these proteases are related and have similar substrate preferences. Therefore, the development of selective chemical tools that enable imaging and discrimination between coagulation factors in biological samples may provide better insight into their roles in various conditions and potentially aid in the establishment of novel diagnostic tests. In our study, we used a large collection of unnatural amino acids, and this enabled us to extensively explore the binding pockets of the enzymes' active sites. Based on the specificity profiles obtained, we designed highly selective substrates, inhibitors, and fluorescent activity-based probes (ABPs) that were used for fast, direct, and simultaneous detection of APC, thrombin, and fXa in human plasma.

3.
Sci Adv ; 8(14): eabm2094, 2022 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-35394846

RESUMO

Misfolding of secretory proteins in the endoplasmic reticulum (ER) features in many human diseases. In α1-antitrypsin deficiency, the pathogenic Z variant aberrantly assembles into polymers in the hepatocyte ER, leading to cirrhosis. We show that α1-antitrypsin polymers undergo a liquid:solid phase transition, forming a protein matrix that retards mobility of ER proteins by size-dependent molecular filtration. The Z-α1-antitrypsin phase transition is promoted during ER stress by an ATF6-mediated unfolded protein response. Furthermore, the ER chaperone calreticulin promotes Z-α1-antitrypsin solidification and increases protein matrix stiffness. Single-particle tracking reveals that solidification initiates in cells with normal ER morphology, previously assumed to represent a healthy pool. We show that Z-α1-antitrypsin-induced hypersensitivity to ER stress can be explained by immobilization of ER chaperones within the polymer matrix. This previously unidentified mechanism of ER dysfunction provides a template for understanding a diverse group of related proteinopathies and identifies ER chaperones as potential therapeutic targets.

4.
Blood ; 139(19): 2972-2982, 2022 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-35148539

RESUMO

The prothrombinase complex processes prothrombin to thrombin through sequential cleavage at Arg320 followed by Arg271 when cofactor, factor (f) Va, protease, fXa, and substrate, prothrombin, are all bound to the same membrane surface. In the absence of the membrane or cofactor, cleavage occurs in the opposite order. For the less favorable cleavage site at Arg320 to be cleaved first, it is thought that prothrombin docks on fVa in a way that presents Arg320 and hides Arg271 from the active site of fXa. Based on the crystal structure of the prothrombinase complex from the venom of the Australian eastern brown snake, pseutarin C, we modeled an initial prothrombin docking mode, which involved an interaction with discrete portions of the A1 and A2 domains of fV and the loop connecting the 2 domains, known as the a1-loop. We interrogated the proposed interface by site-directed PEGylation and by swapping the a1-loop in pseutarin C with that of human fV and fVIII and measuring the effect on rate and pathway of thrombin generation. PEGylation of residues within our proposed binding site greatly reduced the rate of thrombin generation, without affecting the pathway, whereas those outside the proposed interface had no effect. PEGylation of residues within the a1-loop also reduced the rate of thrombin generation. The sequence of the a1-loop was found to play a critical role in prothrombin binding and in the presentation of Arg320 for initial cleavage.


Assuntos
Venenos Elapídicos , Protrombina , Trombina , Austrália , Sítios de Ligação , Fator Va/metabolismo , Fator Xa/metabolismo , Humanos , Protrombina/metabolismo , Trombina/metabolismo , Tromboplastina/metabolismo
5.
Int J Mol Sci ; 22(17)2021 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-34502392

RESUMO

Tick saliva is a rich source of antihemostatic, anti-inflammatory, and immunomodulatory molecules that actively help the tick to finish its blood meal. Moreover, these molecules facilitate the transmission of tick-borne pathogens. Here we present the functional and structural characterization of Iripin-8, a salivary serpin from the tick Ixodes ricinus, a European vector of tick-borne encephalitis and Lyme disease. Iripin-8 displayed blood-meal-induced mRNA expression that peaked in nymphs and the salivary glands of adult females. Iripin-8 inhibited multiple proteases involved in blood coagulation and blocked the intrinsic and common pathways of the coagulation cascade in vitro. Moreover, Iripin-8 inhibited erythrocyte lysis by complement, and Iripin-8 knockdown by RNA interference in tick nymphs delayed the feeding time. Finally, we resolved the crystal structure of Iripin-8 at 1.89 Å resolution to reveal an unusually long and rigid reactive center loop that is conserved in several tick species. The P1 Arg residue is held in place distant from the serpin body by a conserved poly-Pro element on the P' side. Several PEG molecules bind to Iripin-8, including one in a deep cavity, perhaps indicating the presence of a small-molecule binding site. This is the first crystal structure of a tick serpin in the native state, and Iripin-8 is a tick serpin with a conserved reactive center loop that possesses antihemostatic activity that may mediate interference with host innate immunity.


Assuntos
Coagulação Sanguínea/fisiologia , Ativação do Complemento/fisiologia , Ixodes/metabolismo , Serpinas/metabolismo , Animais , Proteínas de Artrópodes/metabolismo , Coagulação Sanguínea/efeitos dos fármacos , Ativação do Complemento/efeitos dos fármacos , Ativação do Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Eritrócitos/metabolismo , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Ixodes/enzimologia , Ixodes/genética , Doença de Lyme , Ninfa , Saliva/química , Glândulas Salivares/metabolismo , Serpinas/ultraestrutura
6.
J Thromb Haemost ; 19(3): 711-718, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33370499

RESUMO

BACKGROUND: The genetically engineered, humanized, bispecific monoclonal antibody emicizumab (Hemlibra) that mimics the cofactor activity of activated factor VIII (FVIII) has been approved for treatment of hemophilia A patients with and without inhibitor. In the pivotal premarketing clinical trials, emicizumab prophylaxis significantly reduced bleeding rates compared with previous treatments and was well tolerated. However, a consequence of this novel therapy may be the host immune response to a foreign protein. OBJECTIVE: Characterization of the neutralizing anti-emicizumab antibody associated with the loss of treatment efficacy. PATIENT: A pediatric hemophilia A patient with inhibitor enrolled in the HAVEN2 (Study of Emicizumab Administered Subcutaneously (SC) in Pediatric Participants With Hemophilia A and Factor VIII (FVIII) Inhibitors) clinical trial. METHODS: The anti-emicizumab antibody has been characterized with Western blot and enzyme-linked immunosorbent assay (ELISA). The antibody was affinity purified and sequenced. Binding affinity to full-length and papain-digested emicizumab was analyzed using surface plasmon resonance and byo-layer interferometry. RESULTS: The neutralizing anti-emicizumab antibody was highly polyclonal with high-affinity binding mainly to the Fab portion of emicizumab with a small amount of binding to the Fc portion. Molecular interaction experiments between emicizumab and the purified antibody indicated the presence of at least two components with similar affinities. CONCLUSIONS: Although the incidence of neutralizing anti-emicizumab antibody is rare, this study highlights the importance of a close monitoring and the need of a simple laboratory assay to promptly detect these antibodies in patients with a history of poor drug efficacy.


Assuntos
Anticorpos Biespecíficos , Hemofilia A , Anticorpos Monoclonais Humanizados , Anticorpos Neutralizantes , Criança , Fator VIII , Hemofilia A/diagnóstico , Hemofilia A/tratamento farmacológico , Humanos
7.
J Dairy Res ; 87(1): 14-22, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32046806

RESUMO

The aims of the study were to determine the long-term effects of dietary supplementation with microalgae (SCIM) on milk and blood fatty acid (FA) composition and reproductive hormones in early lactation dairy cows. Sixty Holstein-Friesian dairy cows (30 per treatment) were unsupplemented (Control) or supplemented with 100 g of SCIM (Schizochytrium limacinum sp.) per cow per day from 25 ± 0.5 d post-partum for 98 d. Intake and milk yield were recorded daily, with milk samples collected at weeks 0, 1, 2, 4, 8 and 14, and blood samples collected from 12 representative pairs per treatment at weeks 0, 2, 4, 8, and 14 for subsequent analysis of FA, ß-hydroxybutyrate, non-esterified fatty acids and glucose. At 33 ± 0.9 d postpartum the oestrus cycle of 24 cows (12 per treatment) were synchronized and plasma 13,14-dihydro-15-keto PGF2α (PGFM) concentrations determined following an oxytocin challenge. Data were analysed by repeated measures analysis of variance. There was no effect of treatment on dry matter intake, milk yield or milk fat content, with mean values across treatments of 22.1 and 40.6, and 37.2 g/kg respectively. Milk fat concentration of C22:6 n-3 increased rapidly in cows receiving SCIM, reaching a maximum of 0.38 g/100 g FA by week 14. Similarly, blood concentration of C22:6 n-3 increased to 1.6 g/100 g FA by week 14 in cows fed SCIM. There was no effect of treatment on plasma metabolites, but plasma glucose was lower in cows fed SCIM compared to the Control at week 2, and higher in week 8. There was no effect of treatment on peak plasma PGFM concentration or area under the curve. It is concluded that feeding SCIM rapidly increases blood and milk concentrations of C22:6 n-3 which are maintained over time, but does not improve plasma PGFM in dairy cows.


Assuntos
Suplementos Nutricionais , Dinoprosta/análogos & derivados , Ácidos Docosa-Hexaenoicos/análise , Microalgas , Leite/química , Animais , Bovinos/sangue , Bovinos/metabolismo , Dinoprosta/sangue , Ácidos Docosa-Hexaenoicos/sangue , Ácidos Graxos/análise , Ácidos Graxos/sangue , Feminino , Lactação , Microalgas/química
9.
Sci Rep ; 9(1): 1605, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30733557

RESUMO

Plasminogen activator inhibitor-1 (PAI-1) is a serine protease inhibitor (serpin) that regulates fibrinolysis, cell adhesion and cell motility via its interactions with plasminogen activators and vitronectin. PAI-1 has been shown to play a role in a number of diverse pathologies including cardiovascular diseases, obesity and cancer and is therefore an attractive therapeutic target. However the multiple patho-physiological roles of PAI-1, and understanding the relative contributions of these in any one disease setting, make the development of therapeutically relevant molecules challenging. Here we describe the identification and characterisation of fully human antibody MEDI-579, which binds with high affinity and specificity to the active form of human PAI-1. MEDI-579 specifically inhibits serine protease interactions with PAI-1 while conserving vitronectin binding. Crystallographic analysis reveals that this specificity is achieved through direct binding of MEDI-579 Fab to the reactive centre loop (RCL) of PAI-1 and at the same exosite used by both tissue and urokinase plasminogen activators (tPA and uPA). We propose that MEDI-579 acts by directly competing with proteases for RCL binding and as such is able to modulate the interaction of PAI-1 with tPA and uPA in a way not previously described for a human PAI-1 inhibitor.


Assuntos
Anticorpos Neutralizantes/imunologia , Inibidor 1 de Ativador de Plasminogênio/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Neutralizantes/química , Especificidade de Anticorpos , Humanos , Camundongos , Modelos Moleculares , Inibidor 1 de Ativador de Plasminogênio/química , Conformação Proteica , Ratos
10.
Sci Rep ; 8(1): 8793, 2018 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-29884816

RESUMO

Activated protein C (APC) is a powerful anticoagulant enzyme that proteolytically inactivates the cofactors of the Xase and prothrombinase complexes, factors VIIIa and Va. A common mutation in factor V, fVLeiden, confers resistance to APC leading to an increased risk of thrombosis in the normal population. However, when coinherited with haemophilia, fVLeiden reduces bleeding severity, suggesting that inhibition of APC may be a useful strategy for treatment of haemophilia. We previously reported on serpins that were rationally designed for improved specificity for APC over other coagulation serine proteases. Based on structural differences in the substrate binding pockets to either side of the P1 Arg, we mutated the P2 and P1' residues to Lys. Although this approach achieved APC specificity, it resulted in a reduction in the rate of APC inhibition relative to the parent containing only the P1 Arg. Here we conduct site-specific random mutagenesis at the P2 and P1' positions to determine if improvements could be made in the rate of APC inhibition. In addition to our original Lys mutations, we found that Arg and Gln also confer specificity for APC. However, in all cases specificity for APC resulted in a reduction in inhibition rate.


Assuntos
Mutagênese Sítio-Dirigida , Proteína C/antagonistas & inibidores , Serpinas/genética , Serpinas/farmacologia , Sítios de Ligação , Coagulação Sanguínea/efeitos dos fármacos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Modelos Moleculares , Proteína C/metabolismo , Conformação Proteica , Serpinas/química , alfa 1-Antitripsina/química , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/farmacologia
11.
Waste Manag ; 75: 251-260, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29459204

RESUMO

Anaerobic digestion was investigated as a potential method for on-farm disposal of fallen stock (pig carcases), degrading the carcase material to produce biogas and digestate. The effects of feedstock (sugar beet pulp or pig carcase material or a 50:50 mix) and organic loading rate (50 g-TS L-1 or 100 g-TS L-1), during mesophilic (35 °C) anaerobic digestion were investigated. Anaerobic digestion was achieved for all experimental treatments, however the pig carcase material at the higher organic loading rate produced the second highest methane yield (0.56 Nm3 kg-VS-1 versus a range of 0.14-0.58 Nm3 kg-VS-1 for other treatments), with the highest percentage of methane in total biogas (61.6% versus a range of 36.1-55.2% for all other treatments). Satisfactory pathogen reduction is a legislative requirement for disposal of carcase material. Pathogens were quantified throughout the anaerobic digestion process. Enterococcus faecalis concentrations decreased to negligible levels (2.8 log10 CFU g-TS-1), whilst Clostridium perfringens levels remained unaffected by treatment throughout the digestion process (5.3 ±â€¯0.2 log10 CFU g-TS-1).


Assuntos
Beta vulgaris , Biocombustíveis , Reatores Biológicos , Anaerobiose , Animais , Fazendas , Metano , Açúcares , Suínos
12.
Curr Opin Hematol ; 24(5): 446-452, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28632502

RESUMO

PURPOSE OF REVIEW: Hemophilia is a debilitating disease, marked by frequent, painful bleeding events, joint deterioration and early death. All current treatments consist of i.v. infusions of replacement factor or other procoagulant factors, and are incompletely effective, due in part to the short half-lives of the proteins. An alternative approach is to rebalance hemostasis by inhibiting natural anticoagulant mechanisms. In this article, we explain why activated protein C (APC) is an appropriate and safe target for the treatment of hemophilia. RECENT FINDINGS: A serpin (serine protease inhibitor) was engineered to specifically inhibit APC and was found to rescue hemostasis in a hemophilia mouse model, even after a severe tail clip injury. However, APC is also anti-inflammatory and has cytoprotective activities, raising safety concerns over the use of an APC inhibitor to treat hemophilia. We summarize the molecular basis of the anticoagulant and signaling activities of APC to assess the potential impact of targeting APC. SUMMARY: We conclude that the signaling and anticoagulant functions of APC are in spatially and kinetically distinct compartments, and that it is possible to specifically inhibit the anticoagulant activity of APC. Targeting APC with a serpin is remarkably effective and may be safe for long-term prophylactic use in the treatment of hemophilia.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Hemofilia A/tratamento farmacológico , Proteína C/antagonistas & inibidores , Serpinas/uso terapêutico , Animais , Modelos Animais de Doenças , Hemofilia A/sangue , Humanos , Camundongos , Proteína C/metabolismo
13.
Br J Haematol ; 178(2): 279-285, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28317092

RESUMO

Existing evidence suggests that in most cases antithrombin deficiency can be explained by mutations in its gene, SERPINC1. We investigated the molecular background of antithrombin deficiency in a single centre family cohort study. We included a total of 21 families comprising 15 original probands and sixty-six relatives, 6 of who were surrogate probands for the genetic analysis. Antithrombin activity and antigen levels were measured. The heparin-antithrombin binding ratio assay was used to distinguish between the different subtypes of type II antithrombin deficiency. SERPINC1 mutations were detected by direct sequencing of all 7 exons and regulatory regions, and multiplex ligation-dependent probe amplification. Eighty-six per cent of the families had a detrimental SERPINC1 gene mutation that segregated in the family. We detected 13 different SERPINC1 gene mutations of which 5 were novel. Among all these mutations, 44% was associated with type I deficiency, whereas the remainder was associated with type II heparin binding site (11%), type II pleiotropic effect (33%), type II reactive site (6%) or had the antithrombin Cambridge II mutation (6%). The current study reports several novel SERPINC1 mutations, thereby adding to our knowledge of the molecular background of antithrombin deficiency. Finally, our results point out the importance of future research outside the conventional SERPINC1 gene approach.


Assuntos
Deficiência de Antitrombina III/genética , Antitrombina III/genética , Mutação/genética , Adolescente , Adulto , Idoso , Proteínas Antitrombina/genética , Pré-Escolar , DNA Recombinante/genética , Éxons/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mutação de Sentido Incorreto/genética , Linhagem , Adulto Jovem
14.
J Biol Chem ; 292(14): 5724-5735, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-28196869

RESUMO

Skewing of the human oral microbiome causes dysbiosis and preponderance of bacteria such as Porphyromonas gingivalis, the main etiological agent of periodontitis. P. gingivalis secretes proteolytic gingipains (Kgp and RgpA/B) as zymogens inhibited by a pro-domain that is removed during extracellular activation. Unraveling the molecular mechanism of Kgp zymogenicity is essential to design inhibitors blocking its activity. Here, we found that the isolated 209-residue Kgp pro-domain is a boomerang-shaped all-ß protein similar to the RgpB pro-domain. Using composite structural information of Kgp and RgpB, we derived a plausible homology model and mechanism of Kgp-regulating zymogenicity. Accordingly, the pro-domain would laterally attach to the catalytic moiety in Kgp and block the active site through an exposed inhibitory loop. This loop features a lysine (Lys129) likely occupying the S1 specificity pocket and exerting latency. Lys129 mutation to glutamate or arginine led to misfolded protein that was degraded in vivo Mutation to alanine gave milder effects but still strongly diminished proteolytic activity, without affecting the subcellular location of the enzyme. Accordingly, the interactions of Lys129 within the S1 pocket are also essential for correct folding. Uniquely for gingipains, the isolated Kgp pro-domain dimerized through an interface, which partially overlapped with that between the catalytic moiety and the pro-domain within the zymogen, i.e. both complexes are mutually exclusive. Thus, pro-domain dimerization, together with partial rearrangement of the active site upon activation, explains the lack of inhibition of the pro-domain in trans. Our results reveal that the specific latency mechanism of Kgp differs from those of Rgps.


Assuntos
Adesinas Bacterianas/química , Cisteína Endopeptidases/química , Precursores Enzimáticos/química , Porphyromonas gingivalis/enzimologia , Porphyromonas gingivalis/patogenicidade , Fatores de Virulência/química , Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Infecções por Bacteroidaceae/enzimologia , Infecções por Bacteroidaceae/genética , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Cisteína Endopeptidases Gingipaínas , Gengivite/enzimologia , Gengivite/genética , Humanos , Microbiota , Boca/microbiologia , Porphyromonas gingivalis/genética , Domínios Proteicos , Multimerização Proteica , Relação Estrutura-Atividade , Fatores de Virulência/metabolismo
15.
Blood Adv ; 1(16): 1224-1237, 2017 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-29296762

RESUMO

Genetics play a significant role in venous thromboembolism (VTE), yet current clinical laboratory-based testing identifies a known heritable thrombophilia (factor V Leiden, prothrombin gene mutation G20210A, or a deficiency of protein C, protein S, or antithrombin) in only a minority of VTE patients. We hypothesized that a substantial number of VTE patients could have lesser-known thrombophilia mutations. To test this hypothesis, we performed whole-exome sequencing (WES) in 64 patients with VTE, focusing our analysis on a novel 55-gene extended thrombophilia panel that we compiled. Our extended thrombophilia panel identified a probable disease-causing genetic variant or variant of unknown significance in 39 of 64 study patients (60.9%), compared with 6 of 237 control patients without VTE (2.5%) (P < .0001). Clinical laboratory-based thrombophilia testing identified a heritable thrombophilia in only 14 of 54 study patients (25.9%). The majority of WES variants were either associated with thrombosis based on prior reports in the literature or predicted to affect protein structure based on protein modeling performed as part of this study. Variants were found in major thrombophilia genes, various SERPIN genes, and highly conserved areas of other genes with established or potential roles in coagulation or fibrinolysis. Ten patients (15.6%) had >1 variant. Sanger sequencing performed in family members of 4 study patients with and without VTE showed generally concordant results with thrombotic history. WES and extended thrombophilia testing are promising tools for improving our understanding of VTE pathogenesis and identifying inherited thrombophilias.

16.
Blood ; 129(1): 105-113, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-27789479

RESUMO

Hemophilia is a bleeding disorder caused by deficiency in factors VIII or IX, the two components of the intrinsic Xase complex. Treatment with replacement factor can lead to the development of inhibitory antibodies, requiring the use of bypassing agents such as factor VIIa and factor concentrates. An alternative approach to bypass the Xase complex is to inhibit endogenous anticoagulant activities. Activated protein C (APC) breaks down the complex that produces thrombin by proteolytically inactivating factor Va. Defects in this mechanism (eg, factor V Leiden) are associated with thrombosis but result in less severe bleeding when co-inherited with hemophilia. Selective inhibition of APC might therefore be effective for the treatment of hemophilia. The endogenous inhibitors of APC are members of the serpin family: protein C inhibitor (PCI) and α1-antitrypsin (α1AT); however, both exhibit poor reactivity and selectivity for APC. We mutated residues in and around the scissile P1-P1' bond in PCI and α1AT, resulting in serpins with the desired specificity profile. The lead candidate was shown to promote thrombin generation in vitro and to restore fibrin and platelet deposition in an intravital laser injury model in hemophilia B mice. The power of targeting APC was further demonstrated by the complete normalization of bleeding after a severe tail clip injury in these mice. These results demonstrate that the protein C anticoagulant system can be successfully targeted by engineered serpins and that administration of such agents is effective at restoring hemostasis in vivo.


Assuntos
Hemofilia B/tratamento farmacológico , Inibidor da Proteína C/farmacologia , Proteína C/antagonistas & inibidores , Serpinas/farmacologia , Animais , Modelos Animais de Doenças , Desenho de Fármacos , Eletroforese em Gel de Poliacrilamida , Humanos , Camundongos
17.
Biochimie ; 122: 235-42, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26365066

RESUMO

Prothrombin is activated to thrombin by the prothrombinase complex through sequential cleavage at two distinct sites. This occurs at sites of vascular injury in a highly regulated cascade of serine protease and cofactor activation, where activated platelets provide a suitable surface for protease/cofactor/substrate assembly. The precise structural and conformational changes undergone during the transition from prothrombin to thrombin have been studied for decades, and several structures of prothrombin fragments along the activation pathway have been solved. Here we present a new structure analyzed in context of other recent structures and biochemical studies. What emerges is an unexpected mechanism that involves a change in the mode of binding of the F2 domain (fragment 2) on the catalytic domain after cleavage at Arg320, and a subsequent reorientation of the linker between the F2 and catalytic domain to present the Arg271 site for cleavage.


Assuntos
Modelos Moleculares , Conformação Proteica , Protrombina/química , Trombina/química , Sequência de Aminoácidos , Arginina/química , Arginina/metabolismo , Sítios de Ligação , Domínio Catalítico , Ativação Enzimática , Fator V/metabolismo , Fator Xa/metabolismo , Humanos , Cinética , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Protrombina/metabolismo , Trombina/metabolismo
18.
Biophys J ; 107(8): 1905-1912, 2014 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-25418171

RESUMO

Emphysema and liver cirrhosis can be caused by the Z mutation (Glu342Lys) in the serine protease inhibitor α1-antitrypsin (α1AT), which is found in more than 4% of the Northern European population. Homozygotes experience deficiency in the lung concomitantly with a massive accumulation of polymers within hepatocytes, causing their destruction. Recently, it was proposed that Z-α1AT polymerizes by a C-terminal domain swap. In this study, small-angle x-ray scattering (SAXS) was used to characterize Z-α1AT polymers in solution. The data show that the Z-α1AT trimer, tetramer, and pentamer all form ring-like structures in strong support of a common domain-swap polymerization mechanism that can lead to self-terminating polymers.


Assuntos
Simulação de Dinâmica Molecular , Multimerização Proteica , alfa 1-Antitripsina/química , Sequência de Aminoácidos , Humanos , Dados de Sequência Molecular , Mutação , Ligação Proteica , Estrutura Terciária de Proteína , Espalhamento a Baixo Ângulo , Difração de Raios X , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/metabolismo
19.
J Biol Chem ; 289(46): 32291-32302, 2014 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-25266723

RESUMO

Cysteine peptidases are key proteolytic virulence factors of the periodontopathogen Porphyromonas gingivalis, which causes chronic periodontitis, the most prevalent dysbiosis-driven disease in humans. Two peptidases, gingipain K (Kgp) and R (RgpA and RgpB), which differ in their selectivity after lysines and arginines, respectively, collectively account for 85% of the extracellular proteolytic activity of P. gingivalis at the site of infection. Therefore, they are promising targets for the design of specific inhibitors. Although the structure of the catalytic domain of RgpB is known, little is known about Kgp, which shares only 27% sequence identity. We report the high resolution crystal structure of a competent fragment of Kgp encompassing the catalytic cysteine peptidase domain and a downstream immunoglobulin superfamily-like domain, which is required for folding and secretion of Kgp in vivo. The structure, which strikingly resembles a tooth, was serendipitously trapped with a fragment of a covalent inhibitor targeting the catalytic cysteine. This provided accurate insight into the active site and suggested that catalysis may require a catalytic triad, Cys(477)-His(444)-Asp(388), rather than the cysteine-histidine dyad normally found in cysteine peptidases. In addition, a 20-Å-long solvent-filled interior channel traverses the molecule and links the bottom of the specificity pocket with the molecular surface opposite the active site cleft. This channel, absent in RgpB, may enhance the plasticity of the enzyme, which would explain the much lower activity in vitro toward comparable specific synthetic substrates. Overall, the present results report the architecture and molecular determinants of the working mechanism of Kgp, including interaction with its substrates.


Assuntos
Adesinas Bacterianas/química , Cisteína Endopeptidases/química , Periodontite/enzimologia , Periodontite/microbiologia , Porphyromonas gingivalis/enzimologia , Sequência de Aminoácidos , Catálise , Domínio Catalítico , Cristalografia por Raios X , Cisteína Endopeptidases Gingipaínas , Humanos , Imunoglobulinas/química , Lisina/química , Modelos Moleculares , Dados de Sequência Molecular , Porphyromonas gingivalis/patogenicidade , Homologia de Sequência de Aminoácidos , Solventes/química , Fatores de Virulência
20.
Biol Chem ; 395(10): 1233-41, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25153592

RESUMO

Thrombin is generated from prothrombin through cleavage at two sites by the prothrombinase complex. Prothrombinase is composed of a protease, factor (f) Xa, and a cofactor, fVa, which interact on negatively charged phospholipid surfaces and cleave prothrombin into thrombin 300 000 times faster than fXa alone. The balance between bleeding and thrombosis depends on the amount of thrombin produced, and this in turn depends on the function of the prothrombinase complex. How fXa and fVa interact and how improved prothrombin processing is conferred are of critical importance for understanding healthy and pathological blood clotting. Until recently, little structural information was available, and molecular models were built on partial structures with assembly guided by biochemical data. Last year our group published a crystal structure of a prothrombinase complex from the venom of the Australian Eastern Brown snake (known as Pseutarin C). Here we use the crystal structure of Pseutarin C as a starting point for homology modelling and assembly of the full human prothrombinase complex. The interface is complementary in shape and charge, and is consistent with much of the published biochemical data. The model of human prothrombinase presented here provides a powerful resource for contextualizing previous data and for designing future experiments.


Assuntos
Venenos Elapídicos/química , Fator V/química , Fator Xa/química , Animais , Cristalografia por Raios X , Fator Va/química , Humanos , Modelos Moleculares , Serpentes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...