Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 237
Filtrar
1.
J Biol Chem ; 300(3): 105683, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38272220

RESUMO

Histidine-rich glycoprotein (HRG) is an abundant plasma protein harboring at least three N-glycosylation sites. HRG integrates many biological processes, such as coagulation, antiangiogenic activity, and pathogen clearance. Importantly, HRG is known to exhibit five genetic variants with minor allele frequencies of more than 10%. Among them, Pro204Ser can induce a fourth N-glycosylation site (Asn202). Considerable efforts have been made to reveal the biological function of HRG, whereas data on HRG glycosylation are scarcer. To close this knowledge gap, we used C18-based LC-MS/MS to study the glycosylation characteristics of six HRG samples from different sources. We used endogenous HRG purified from human plasma and compared its glycosylation to that of the recombinant HRG produced in Chinese hamster ovary cells or human embryonic kidney 293 cells, targeting distinct genotypic isoforms. In endogenous plasma HRG, every N-glycosylation site was occupied predominantly with a sialylated diantennary complex-type glycan. In contrast, in the recombinant HRGs, all glycans showed different antennarities, sialylation, and core fucosylation, as well as the presence of oligomannose glycans, LacdiNAcs, and antennary fucosylation. Furthermore, we observed two previously unreported O-glycosylation sites in HRG on residues Thr273 and Thr274. These sites together showed more than 90% glycan occupancy in all HRG samples studied. To investigate the potential relevance of HRG glycosylation, we assessed the plasmin-induced cleavage of HRG under various conditions. These analyses revealed that the sialylation of the N- and O-glycans as well as the genotype-dependent N-glycosylation significantly influenced the kinetics and specificity of plasmin-induced cleavage of HRG.


Assuntos
Fibrinolisina , Proteínas , Espectrometria de Massas em Tandem , Animais , Cricetinae , Humanos , Células CHO , Cricetulus , Fibrinolisina/química , Genótipo , Glicosilação , Polissacarídeos/química , Isoformas de Proteínas , Cromatografia Líquida de Alta Pressão
2.
Arch Biochem Biophys ; 743: 109671, 2023 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-37336343

RESUMO

Staphylokinase (SAK), a potent fibrin-specific plasminogen activator secreted by Staphylococcus aureus, carries a pair of lysine at the carboxy-terminus that play a key role in plasminogen activation. The underlaying mechanism by which C-terminal lysins of SAK modulate its function remains unknown. This study has been undertaken to unravel role of C-terminal lysins of SAK in plasminogen activation. While deletion of C-terminal lysins (Lys135, Lys136) drastically impaired plasminogen activation by SAK, addition of lysins enhanced its catalytic activity 2-2.5-fold. Circular dichroism analysis revealed that C-terminally modified mutants of SAK carry significant changes in their beta sheets and secondary structure. Structure models and RING (residue interaction network generation) studies indicated that the deletion of lysins has conferred extensive topological alterations in SAK, disrupting vital interactions at the interface of SAK.plasmin complex, thereby leading significant impairment in its functional activity. In contrast, addition of lysins at the C-terminus enhanced its conformational flexibility, creating a stronger coupling at the interface of SAK.plasmin complex and making it more efficient for plasminogen activation. Taken together, these studies provided new insights on the role of C-terminal lysins in establishment of precise intermolecular interactions of SAK with the plasmin for the optimal function of activator complex.


Assuntos
Fibrinolisina , Lisina , Fibrinolisina/química , Plasminogênio/química , Ativadores de Plasminogênio/química
3.
ACS Appl Mater Interfaces ; 15(2): 2617-2629, 2023 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-36596222

RESUMO

The fibrillization and deposition of the human islet amyloid polypeptide (hIAPP) are the pathological hallmark of type 2 diabetes mellitus (T2DM), and these insoluble fibrotic depositions of hIAPP are considered to strongly affect insulin secretion by inducing toxicity toward pancreatic islet ß-cells. The current strategy of preventing amyloid aggregation by nanoparticle-assisted inhibitors can only disassemble fibrotic amyloids into more toxic oligomers and/or protofibrils. Herein, for the first time, we propose a type of cysteine-derived chiral carbon quantum dot (CQD) that targets plasmin, a core natural fibrinolytic protease in humans. These CQDs can serve as fibrinolytic activity regulators for plasmin to cleave hIAPP into nontoxic polypeptides or into even smaller amino acid fragments, thus alleviating hIAPP's fibrotic amyloid-induced cytotoxicity. Our experiments indicate that chiral CQDs have opposing effects on plasmin activity. The l-CQDs promote the cleavage of hIAPP by enhancing plasmin activity at a promotion ratio of 23.2%, thus protecting ß-cells from amyloid-induced toxicity. In contrast, the resultant d-CQDs significantly inhibit proteolysis, decreasing plasmin activity by 31.5% under the same reaction conditions. Second harmonic generation (SHG) microscopic imaging is initially used to dynamically characterize hIAPP before and after proteolysis. The l-CQD promotion of plasmin activity thus provides a promising avenue for the hIAPP-targeted treatment of T2DM to treat low fibrinolytic activity, while the d-CQDs, as inhibitors of plasmin activity, may improve patient survival for hyperfibrinolytic conditions, such as those existing during surgeries and traumas.


Assuntos
Diabetes Mellitus Tipo 2 , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Pontos Quânticos , Humanos , Amiloide/química , Carbono , Cisteína , Diabetes Mellitus Tipo 2/tratamento farmacológico , Fibrinolisina/química , Fibrinolisina/efeitos dos fármacos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Polipeptídeo Amiloide das Ilhotas Pancreáticas/efeitos dos fármacos , Pontos Quânticos/química , Pontos Quânticos/uso terapêutico
4.
ChemMedChem ; 18(6): e202200632, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36710259

RESUMO

Two series of macrocyclic plasmin inhibitors with a C-terminal benzylamine group were synthesized. The substitution of the N-terminal phenylsulfonyl group of a previously described inhibitor provided two analogues with sub-nanomolar inhibition constants. Both compounds possess a high selectivity against all other tested trypsin-like serine proteases. Furthermore, a new approach was used to selectively introduce asymmetric linker segments. Two of these compounds inhibit plasmin with Ki values close to 2 nM. For the first time, four crystal structures of these macrocyclic inhibitors could be determined in complex with a Ser195Ala microplasmin mutant. The macrocyclic core segment of the inhibitors binds to the open active site of plasmin without any steric hindrance. This binding mode is incompatible with other trypsin-like serine proteases containing a sterically demanding 99-hairpin loop. The crystal structures obtained experimentally explain the excellent selectivity of this inhibitor type as previously hypothesized.


Assuntos
Antifibrinolíticos , Fibrinolisina , Fibrinolisina/química , Fibrinolisina/metabolismo , Antifibrinolíticos/química , Antifibrinolíticos/farmacologia , Tripsina/química , Ligação Proteica , Inibidores de Serina Proteinase/farmacologia , Inibidores de Serina Proteinase/química
5.
ChemMedChem ; 17(22): e202200364, 2022 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-36111842

RESUMO

There is an emerging interest in utilizing synthetic multivalent inhibitors that comprise of multiple inhibitor moieties linked on a common scaffold to achieve strong and selective enzyme inhibition. As multivalent inhibition is impacted by valency and linker length, in this study, we explore the effect of multivalent benzamidine inhibitors of varying valency and linker length on plasmin inhibition. Plasmin is an endogenous enzyme responsible for digesting fibrin present in blood clots. Monovalent plasmin(ogen) inhibitors are utilized clinically to treat hyperfibrinolysis-associated bleeding events. Benzamidine is a reversible inhibitor that binds to plasmin's active site. Herein, multivalent benzamidine inhibitors of varying valencies (mono-, bi- and tri-valent) and linker lengths (∼1-12 nm) were synthesized to systematically study their effect on plasmin inhibition. Inhibition assays were performed using a plasmin substrate (S-2251) to determine inhibition constants (Ki). Pentamidine (shortest bivalent) and Tri-AMB (shortest trivalent) were the strongest inhibitors with Ki values of 2.1±0.8 and 3.9±1.7 µM, respectively. Overall, increasing valency and decreasing linker length, increases effective local concentration of the inhibitor and therefore, resulted in stronger inhibition of plasmin via statistical rebinding. This study aids in the design of multivalent inhibitors that can achieve desired enzyme inhibition by means of modulating valency and linker length.


Assuntos
Benzamidinas , Fibrinolisina , Fibrinolisina/química , Fibrinolisina/metabolismo , Benzamidinas/farmacologia
6.
Int J Mol Sci ; 22(21)2021 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-34768908

RESUMO

Ischemic stroke is the most common type of stroke and thrombolytic therapy is the only approved treatment. However, current thrombolytic therapy with tissue plasminogen activator (tPA) is often hampered by the increased risk of hemorrhage. Plasmin, a direct fibrinolytic, has a significantly superior hemostatic safety profile; however, if injected intravenously it becomes rapidly inactivated by anti-plasmin. Nanoformulations have been shown to increase drug stability and half-life and hence could be applied to increase the plasmin therapeutic efficacy. Here in this paper, we report a novel heparin and arginine-based plasmin nanoformulation that exhibits increased plasmin stability and efficacy. In vitro studies revealed significant plasmin stability in the presence of anti-plasmin and efficient fibrinolytic activity. In addition, these particles showed no significant toxicity or oxidative stress effects in human brain microvascular endothelial cells, and no significant blood brain barrier permeability. Further, in a mouse photothrombotic stroke model, plasmin nanoparticles exhibited significant efficacy in reducing stroke volume without overt intracerebral hemorrhage (ICH) compared to free plasmin treatment. The study shows the potential of a plasmin nanoformulation in ischemic stroke therapy.


Assuntos
Arginina/química , Fibrinolisina/administração & dosagem , Heparina/química , AVC Isquêmico/terapia , Nanopartículas/administração & dosagem , Terapia Trombolítica/métodos , Animais , Barreira Hematoencefálica , Fibrinolisina/química , Fibrinolíticos/administração & dosagem , Fibrinolíticos/química , Humanos , Infarto da Artéria Cerebral Média/complicações , AVC Isquêmico/etiologia , AVC Isquêmico/metabolismo , AVC Isquêmico/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas/química
7.
Molecules ; 26(8)2021 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-33920584

RESUMO

Age gelation is a major quality defect in ultra-high-temperature (UHT) pasteurized milk during extended storage. Changes in plasmin (PL)-induced sedimentation were investigated during storage (23 °C and 37 °C, four weeks) of UHT skim milk treated with PL (2.5, 10, and 15 U/L). The increase in particle size and broadening of the particle size distribution of samples during storage were dependent on the PL concentration, storage period, and storage temperature. Sediment analysis indicated that elevated storage temperature accelerated protein sedimentation. The initial PL concentration was positively correlated with the amount of protein sediment in samples stored at 23 °C for four weeks (r = 0.615; p < 0.01), whereas this correlation was negative in samples stored at 37 °C for the same time (r = -0.358; p < 0.01) due to extensive proteolysis. SDS-PAGE revealed that whey proteins remained soluble over storage at 23 °C for four weeks, but they mostly disappeared from the soluble phase of PL-added samples after two weeks' storage at 37 °C. Transmission electron micrographs of PL-containing UHT skim milk during storage at different temperatures supported the trend of sediment analysis well. Based on the Fourier transform infrared spectra of UHT skim milk stored at 23 °C for three weeks, PL-induced particle size enlargement was due to protein aggregation and the formation of intermolecular ß-sheet structures, which contributed to casein destabilization, leading to sediment formation.


Assuntos
Fibrinolisina/química , Conservação de Alimentos , Proteínas do Leite/química , Leite/química , Animais , Caseínas/química , Bovinos , Fibrinolisina/isolamento & purificação , Fibrinolisina/ultraestrutura , Manipulação de Alimentos , Temperatura Alta/efeitos adversos , Humanos , Proteínas do Leite/isolamento & purificação , Proteínas do Leite/ultraestrutura , Tamanho da Partícula , Proteínas do Soro do Leite
8.
Artigo em Inglês | MEDLINE | ID: mdl-33838314

RESUMO

Bee venom is a mixture of bioactive components that include proteases and protease inhibitors. A metalloprotease inhibitor has been predicted to be a bumblebee-specific toxin in the venom proteome of Bombus terrestris; however, the identification and functional roles of bee venom metalloprotease inhibitors have not been previously determined. In this study, we identified a bumblebee (B. ignitus) venom metalloprotease inhibitor (BiVMPI) that exhibits anti-fibrinolytic activity. BiVMPI contains a trypsin inhibitor-like cysteine-rich domain that exhibits similarity to inducible metalloprotease inhibitor. Using an anti-BiVMPI antibody raised against a recombinant BiVMPI protein produced in baculovirus-infected insect cells, the presence of BiVMPI in the venom gland and secreted venom of B. ignitus worker bees was confirmed. The recombinant BiVMPI protein demonstrated inhibitory activity against a metalloprotease, trypsin, chymotrypsin, protease K, and plasmin, but not subtilisin A, elastase, or thrombin. Additionally, the recombinant BiVMPI bound to plasmin and inhibited the plasmin-mediated degradation of fibrin, demonstrating an anti-fibrinolytic role for BiVMPI as a bee venom metalloprotease inhibitor. Our results provide the first evidence for the identification and anti-fibrinolytic activity of a metalloprotease inhibitor from bee venom.


Assuntos
Venenos de Abelha/química , Fibrinogênio/química , Proteínas de Insetos/química , Inibidores de Metaloproteinases de Matriz/química , Proteínas Recombinantes/química , Animais , Abelhas , Fibrinolisina/química , Humanos
9.
Biochem J ; 477(5): 953-970, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-32069359

RESUMO

To understand the role of substrate plasminogen kringles in its differential catalytic processing by the streptokinase - human plasmin (SK-HPN) activator enzyme, Fluorescence Resonance Energy Transfer (FRET) model was generated between the donor labeled activator enzyme and the acceptor labeled substrate plasminogen (for both kringle rich Lys plasminogen - LysPG, and kringle less microplasminogen - µPG as substrates). Different steps of plasminogen to plasmin catalysis i.e. substrate plasminogen docking to scissile peptide bond cleavage, chemical transformation into proteolytically active product, and the decoupling of the nascent product from the SK-HPN activator enzyme were segregated selectively using (1) FRET signal as a proximity sensor to score the interactions between the substrate and the activator during the cycle of catalysis, (2) active site titration studies and (3) kinetics of peptide bond cleavage in the substrate. Remarkably, active site titration studies and the kinetics of peptide bond cleavage have shown that post docking chemical transformation of the substrate into the product is independent of kringles adjacent to the catalytic domain (CD). Stopped-flow based rapid mixing experiments for kringle rich and kringle less substrate plasminogen derivatives under substrate saturating and single cycle turnover conditions have shown that the presence of kringle domains adjacent to the CD in the macromolecular substrate contributes by selectively speeding up the final step, namely the product release/expulsion step of catalysis by the streptokinase-plasmin(ogen) activator enzyme.


Assuntos
Domínio Catalítico/fisiologia , Fibrinolisina/metabolismo , Kringles/fisiologia , Plasminogênio/metabolismo , Estreptoquinase/metabolismo , Catálise , Fibrinolisina/química , Transferência Ressonante de Energia de Fluorescência/métodos , Humanos , Plasminogênio/química , Estrutura Secundária de Proteína , Estreptoquinase/química , Especificidade por Substrato/fisiologia
10.
Thromb Haemost ; 120(3): 400-411, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31940673

RESUMO

Factor XII (FXII) zymogen activation requires cleavage after arginine 353 located in the activation loop. This cleavage can be executed by activated FXII (autoactivation), plasma kallikrein (PKa), or plasmin. Previous studies proposed that the activation loop of FXII is shielded to regulate FXII activation and subsequent contact activation. In this study, we aimed to elucidate this mechanism by expressing and characterizing seven consecutive N-terminally truncated FXII variants as well as full-length wild-type (WT) FXII. As soon as the fibronectin type II domain is lacking (FXII Δ1-71), FXII cleavage products appear on Western blot. These fragments display spontaneous amidolytic activity, indicating that FXII without the fibronectin type II domain is susceptible to autoactivation. Additionally, truncated FXII Δ1-71 is more easily activated by PKa or plasmin than full-length WT FXII. To exclude a contribution of autoactivation, we expressed active-site incapacitated FXII truncation variants (S544A). FXII S544A Δ1-71 is highly susceptible to cleavage by PKa, indicating exposure of the activation loop. In surface binding experiments, we found that the fibronectin type II domain is non-essential for binding to kaolin or polyphosphate, whereas the following epidermal growth factor-like domain is indispensable. Binding of full-length FXII S544A to kaolin or polyphosphate increases its susceptibility to cleavage by PKa. Moreover, the activation of full-length WT FXII by PKa increases approximately threefold in the presence of kaolin. Deletion of the fibronectin type II domain eliminates this effect. Combined, these findings suggest that the fibronectin type II domain shields the activation loop of FXII, ensuring zymogen quiescence.


Assuntos
Precursores Enzimáticos/química , Fator XII/química , Fibrinolisina/química , Fibronectinas/química , Calicreínas/química , Animais , Sítios de Ligação , Coagulação Sanguínea , Bradicinina/química , Domínio Catalítico , Bovinos , Fator XIIa/química , Fibronectinas/sangue , Células HEK293 , Humanos , Calicreínas/sangue , Caulim/química , Polifosfatos/química , Ligação Proteica , Domínios Proteicos
11.
J Med Chem ; 63(4): 1445-1472, 2020 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-31658420

RESUMO

Hyperfibrinolytic situations can lead to life-threatening bleeding, especially during cardiac surgery. The approved antifibrinolytic agents such as tranexamic acid, ε-aminocaproic acid, 4-aminomethylbenzoic acid, and aprotinin were developed in the 1960s without the structural insight of their respective targets. Crystal structures of the main antifibrinolytic targets, the lysine binding sites on plasminogen's kringle domains, and plasmin's serine protease domain greatly contributed to the structure-based drug design of novel inhibitor classes. Two series of ligands targeting the lysine binding sites have been recently described, which are more potent than the most-widely used antifibrinolytic agent, tranexamic acid. Furthermore, four types of promising active site inhibitors of plasmin have been developed: tranexamic acid conjugates targeting the S1 pocket and primed sites, substrate-analogue linear homopiperidylalanine-containing 4-amidinobenzylamide derivatives, macrocyclic inhibitors addressing nonprimed binding regions, and bicyclic 14-mer SFTI-1 analogues blocking both, primed and nonprimed binding sites of plasmin. Furthermore, several allosteric plasmin inhibitors based on heparin mimetics have been developed.


Assuntos
Antifibrinolíticos/uso terapêutico , Fibrinólise/efeitos dos fármacos , Hemorragia/tratamento farmacológico , Hemorragia/prevenção & controle , Animais , Antifibrinolíticos/química , Antifibrinolíticos/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Fibrinolisina/química , Fibrinolisina/metabolismo , Humanos , Ligantes , Estrutura Molecular , Plasminogênio/química , Plasminogênio/metabolismo , Ligação Proteica , Domínios Proteicos
12.
J Thromb Haemost ; 17(12): 2004-2015, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31529593

RESUMO

Fibrinolytic agents including plasmin and plasminogen activators improve outcomes in acute ischemic stroke and thrombosis by recanalizing occluded vessels. In the decades since their introduction into clinical practice, several limitations of have been identified in terms of both efficacy and bleeding risk associated with these agents. Engineered nanoparticles and microparticles address some of these limitations by improving circulation time, reducing inhibition and degradation in circulation, accelerating recanalization, improving targeting to thrombotic occlusions, and reducing off-target effects; however, many particle-based approaches have only been used in preclinical studies to date. This review covers four advances in coupling fibrinolytic agents with engineered particles: (a) modifications of plasminogen activators with macromolecules, (b) encapsulation of plasminogen activators and plasmin in polymer and liposomal particles, (c) triggered release of encapsulated fibrinolytic agents and mechanical disruption of clots with ultrasound, and (d) enhancing targeting with magnetic particles and magnetic fields. Technical challenges for the translation of these approaches to the clinic are discussed.


Assuntos
Portadores de Fármacos , Fibrinolisina/administração & dosagem , Fibrinólise/efeitos dos fármacos , Fibrinolíticos/administração & dosagem , Nanomedicina , Nanopartículas , Ativadores de Plasminogênio/administração & dosagem , Terapia Trombolítica , Animais , Composição de Medicamentos , Fibrinolisina/química , Fibrinolisina/farmacocinética , Fibrinolíticos/química , Fibrinolíticos/farmacocinética , Ondas de Choque de Alta Energia , Humanos , Lipossomos , Nanopartículas de Magnetita , Ativadores de Plasminogênio/química , Ativadores de Plasminogênio/farmacocinética
13.
J Sci Food Agric ; 99(15): 6922-6930, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31393601

RESUMO

BACKGROUND: The potential use of polyphenols to improve the functional characteristics of dairy products has gained much attention. However, the effects of the polyphenols on naturally occurring enzymes in milk have not been studied extensively. Excess plasmin activity in dairy products might result in several quality defects. The objective of this study was to assess the ability of polyphenols to inhibit plasmin in milk using a molecular and kinetic approach. RESULTS: Epicatechin gallate (ECG), epigallocatechin gallate (EGCG), quercetin (QUER), and myricetin (MYR) caused a significant decrease in plasmin activity by 60, 86, 65, and 90%, respectively. The inhibition rates were alleviated in the presence of milk proteins. EGCG, QUER, and MYR, exhibited noncompetitive inhibition against plasmin, whereas ECG caused a mixed-type inhibition. A decrease in the random structure of plasmin upon the complex formation with ECG, EGCG, QUER, and MYR was found. The other phenolics that were evaluated did not cause any significant changes in plasmin conformation. The observed inhibitory phenolic-plasmin interactions were dominated by H-bonds and electrostatic attractions. Green tea extract (GTE) rich in catechins also inhibited plasmin activity in the milk. CONCLUSION: Significant changes in the secondary structure of plasmin upon binding of ECG, EGCG, QUER, and MYR led to diminished plasmin activity both in the absence and presence of milk proteins. These flavonoids with promising plasmin inhibitory potential could be used in new dairy formulations leading to controlled undesired consequences of plasmin activity. © 2019 Society of Chemical Industry.


Assuntos
Antifibrinolíticos/química , Camellia sinensis/química , Leite/enzimologia , Extratos Vegetais/química , Polifenóis/química , Animais , Catequina/análogos & derivados , Catequina/química , Bovinos , Fibrinolisina/química , Cinética , Leite/química
14.
FEBS Open Bio ; 9(7): 1259-1269, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31087538

RESUMO

Streptokinase (SK) is a plasminogen activator which converts inactive plasminogen (Pg) to active plasmin (Pm), which cleaves fibrin clots. SK secreted by groups A, C, and G Streptococcus (SKA/SKC/SKG) is composed of three domains: SKα, SKß and SKγ. Previous domain-swapping studies between SK1/SK2b-cluster variants revealed that SKß plays a major role in the activation of human Pg. Here, we carried out domain-swapping between skcg-SK/SK2-cluster variants to determine the involvement of SKß in several SK functionalities, including specific/proteolytic activity kinetics, fibrinogen-bound Pg activation and α2 -antiplasmin resistance. Our results indicate that SKß has a minor to determining role in these diverse functionalities for skcg-SK and SK2b variants, which might potentially be accompanied by few critical residues acting as hot spots. Our findings enhance our understanding of the roles of SKß and hot spots in different functional characteristics of SK clusters and may aid in the engineering of fibrin-specific variants of SK for breaking down blood clots with potentially higher efficacy and safety.


Assuntos
Domínios Proteicos/fisiologia , Estreptoquinase/metabolismo , Proteínas de Bactérias/química , Fibrinogênio , Fibrinolisina/química , Fibrinolisina/metabolismo , Cinética , Plasminogênio/química , Plasminogênio/metabolismo , Ativadores de Plasminogênio/química , Ativadores de Plasminogênio/metabolismo , Ligação Proteica , Engenharia de Proteínas/métodos , Proteólise , Streptococcus/metabolismo , Estreptoquinase/química , Estreptoquinase/fisiologia
15.
J Thromb Haemost ; 17(9): 1461-1469, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31136076

RESUMO

BACKGROUND: Excessive, plasmin-mediated fibrinolysis augments bleeding and contributes to death in some patients. Current therapies for fibrinolytic bleeding are limited by modest efficacy, low potency, and off-target effects. OBJECTIVES: To determine whether an antibody directed against unique loop structures of the plasmin protease domain may have enhanced specificity and potency for blocking plasmin activity, fibrinolysis, and experimental hemorrhage. METHODS: The binding specificity, affinity, protease cross-reactivity and antifibrinolytic properties of a monoclonal plasmin inhibitor antibody (Pi) were examined and compared with those of epsilon aminocaproic acid (EACA), which is a clinically used fibrinolysis inhibitor. RESULTS: Pi specifically recognized loop 5 of the protease domain, and did not bind to other serine proteases or nine other non-primate plasminogens. Pi was ~7 logs more potent in neutralizing plasmin cleavage of small-molecule substrates and >3 logs more potent in quenching fibrinolysis than EACA. Pi was similarly effective in blocking catalysis of a small-molecule substrate as α2 -antiplasmin, which is the most potent covalent inhibitor of plasmin, and was a more potent fibrinolysis inhibitor. Fab or chimerized Fab fragments of Pi were equivalently effective. In vivo, in a humanized model of fibrinolytic surgical bleeding, Pi significantly reduced bleeding to a greater extent than a clinical dose of EACA. CONCLUSIONS: A mAb directed against unique loop sequences in the protease domain is a highly specific, potent, competitive plasmin inhibitor that significantly reduces experimental surgical bleeding in vivo.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Antifibrinolíticos/uso terapêutico , Fibrinolisina/antagonistas & inibidores , Hemorragia/tratamento farmacológico , Ácido Aminocaproico/farmacologia , Ácido Aminocaproico/uso terapêutico , Animais , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/farmacologia , Afinidade de Anticorpos , Ligação Competitiva , Domínio Catalítico/imunologia , Reações Cruzadas , Avaliação Pré-Clínica de Medicamentos , Feminino , Fibrinolisina/química , Fibrinolisina/imunologia , Fibrinólise/efeitos dos fármacos , Hemorragia/sangue , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Conformação Proteica , Domínios Proteicos , Distribuição Aleatória , Proteínas Recombinantes de Fusão/imunologia , Especificidade da Espécie , Especificidade por Substrato
16.
Int J Biol Macromol ; 120(Pt B): 1591-1600, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30261260

RESUMO

Plasmin (EC 3.4.21.7) is a key enzyme of the fibrinolytic system, responsible for the degradation of fibrin clot and maintaining blood fluidity. Hence, alterations of the fibrinolytic capacity of blood plasma may contribute to thrombotic or bleeding complications. The aim of this study was to determine effects of a bufadienolide-rich fraction, isolated from roots of Kalanchoe daigremontiana (0.05-50 µg/ml) on enzymatic properties of plasmin. Hydrolysis of a synthetic substrate S-2251 (H-D-Valyl-l-leucyl-l-lysine-p-nitroaniline dihydrochloride) by plasmin revealed that the bufadienolide-rich fraction had a diverse effect on this enzyme, dependently on the concentration range. While the lower concentrations of the examined fraction (0.05-2.5 µg/ml) significantly enhanced the amidolytic activity of plasmin, at 25-50 µg/ml concentrations, the enzyme was evidently inhibited (by about 60%). The Lineweaver-Burk plot indicated on an uncompetitive inhibition of plasmin. Inhibitory effects (up to 80%) were also found in the streptokinase-induced plasminogen activation to plasmin. Docking results suggest that only some of compounds (mostly bersaldegenin 1-acetate (10), bryotoxin (13) and hovetrichoside C (17)) were bound to plasminogen/plasmin, depending on the presence or absence of the substrate in the active site. The obtained findings suggest allosteric regulation of plasminogen activation and plasmin activity by components of the examined fraction.


Assuntos
Bufanolídeos/farmacologia , Simulação por Computador , Fibrinolisina/metabolismo , Kalanchoe/química , Bufanolídeos/metabolismo , Relação Dose-Resposta a Droga , Fibrinolisina/química , Humanos , Hidrólise/efeitos dos fármacos , Simulação de Acoplamento Molecular , Conformação Proteica
17.
J Thromb Haemost ; 16(11): 2276-2288, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30176116

RESUMO

Essentials Activated clotting factor X (FXa) acquires fibrinolytic cofactor function after cleavage by plasmin. FXa-mediated plasma fibrinolysis is enabled by active site modification blocking a second cleavage. FXa-directed oral anticoagulants (DOACs) alter FXa cleavage by plasmin. DOACs enhance FX-dependent fibrinolysis and plasmin generation by tissue plasminogen activator. BACKGROUND: When bound to an anionic phospholipid-containing membrane, activated clotting factor X (FXa) is sequentially cleaved by plasmin from the intact form, FXaα, to FXaß and then to Xa33/13. Tissue-type plasminogen activator (t-PA) produces plasmin and is the initiator of fibrinolysis. Both FXaß and Xa33/13 enhance t-PA-mediated plasminogen activation. Although stable in experiments using purified proteins, Xa33/13 rapidly loses t-PA cofactor function in plasma. Bypassing this inhibition, covalent modification of the FXaα active site prevents Xa33/13 formation by plasmin, and the persistent FXaß enhances plasma fibrinolysis. As the direct oral anticoagulants (DOACs) rivaroxaban and apixaban bind to the FXa active site, we hypothesized that they similarly modulate FXa fibrinolytic function. METHODS: DOAC effects on fibrinolysis and the t-PA cofactor function of FXa were studied in patient plasma, normal pooled plasma and purified protein experiments by the use of light scattering, chromogenic assays, and immunoblots. RESULTS: The plasma of patients taking rivaroxaban showed enhanced fibrinolysis correlating with FXaß. In normal pooled plasma, the addition of rivaroxaban or apixaban also shortened fibrinolysis times. This was related to the cleavage product, FXaß, which increased plasmin production by t-PA. It was confirmed that these results were not caused by DOACs affecting activated FXIII-mediated fibrin crosslinking, clot ultrastructure and thrombin-activatable fibrinolysis inhibitor activation in plasma. CONCLUSION: The current study suggests a previously unknown effect of DOACs on FXa in addition to their well-documented anticoagulant role. By enabling the t-PA cofactor function of FXaß in plasma, DOACs also enhance fibrinolysis. This effect may broaden their therapeutic indications.


Assuntos
Fator Xa/química , Pirazóis/farmacologia , Piridonas/farmacologia , Rivaroxabana/farmacologia , Administração Oral , Anticoagulantes/química , Coagulação Sanguínea/efeitos dos fármacos , Domínio Catalítico , Reagentes de Ligações Cruzadas/química , Inibidores do Fator Xa/farmacologia , Fibrina/química , Fibrinolisina/química , Fibrinólise , Humanos , Fosfolipídeos/química , Trombina/química , Terapia Trombolítica , Trombose , Ativador de Plasminogênio Tecidual/química
18.
J Struct Biol ; 203(3): 273-280, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29859328

RESUMO

Fibrin plays a fundamentally important role during hemostasis. To withstand the shear forces of blood flow and prevent embolisation, fibrin monomers form a three-dimensional polymer network that serves as an elastic scaffold for the blood clot. The complex spatial hierarchy of the fibrin meshwork, however, severely complicates the exploration of structural features, mechanical properties and molecular changes associated with the individual fibers of the clot. Here we developed a quasi-two-dimensional nanoscale fibrin matrix that enables the investigation of fibrin properties by topographical analysis using atomic force microscopy. The average thickness of the matrix was ∼50 nm, and structural features of component fibers were accessible. The matrix could be lysed with plasmin following rehydration. By following the topology of the matrix during lysis, we were able to uncover the molecular mechanisms of the process. Fibers became flexible but retained axial continuity for an extended time period, indicating that lateral interactions between protofibrils are disrupted first, but the axial interactions remain stable. Nearby fibers often fused into bundles, pointing at the presence of a cohesional force between them. Axial fiber fragmentation rapidly took place in the final step. Conceivably, the persisting axial integrity and cohesion of the fibrils assist to maintain global clot structure, to prevent microembolism, and to generate a high local plasmin concentration for the rapid, final axial fibril fragmentation. The nanoscale fibrin matrix developed and tested here provides a unique insight into the molecular mechanisms behind the structural and mechanical features of fibrin and its proteolytic degradation.


Assuntos
Produtos de Degradação da Fibrina e do Fibrinogênio/ultraestrutura , Fibrina/ultraestrutura , Fibrinolisina/química , Fibrina/química , Produtos de Degradação da Fibrina e do Fibrinogênio/química , Fibrinólise/genética , Hemostasia , Humanos , Microscopia de Força Atômica , Proteólise , Fluxo Sanguíneo Regional
19.
Food Chem ; 263: 127-134, 2018 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-29784297

RESUMO

Destabilization of UHT milk during its shelf life is mainly promoted by the residual proteolytic activity attributed to the psychrotrophic bacterial proteases and native milk proteases. In this study, we built skim UHT milk-based model systems to which either the major bacterial protease (AprX from Pseudomonas fluorescens), or the major native milk protease (plasmin) was added, to allow a direct comparison between the destabilization of skim UHT milk by both categories of enzymes. The physical and chemical properties were studied during 6 weeks. Our results showed AprX induced compact gels when almost all the κ-casein was hydrolyzed and the degree of hydrolysis (DH) exceeded 1.3%. Plasmin induced soft gels when around 60% of both ß- and αs1-casein were hydrolyzed and the DH reached 2.1%. The knowledge gained from this study may be used for developing diagnostic tests for determining the protease responsible for UHT milk destabilisation.


Assuntos
Proteínas de Bactérias/metabolismo , Fibrinolisina/química , Leite/química , Pasteurização/métodos , Pseudomonas fluorescens/enzimologia , Serina Endopeptidases/metabolismo , Animais , Caseínas/química , Caseínas/metabolismo , Armazenamento de Alimentos , Géis/análise , Leite/metabolismo , Proteólise
20.
Thromb Haemost ; 118(3): 471-479, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29534248

RESUMO

ADAMTS13 (a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13) cleaves von Willebrand Factor (VWF) multimers to control their thrombogenicity. The fibrinolytic enzyme plasmin can cleave VWF in a similar manner. However, plasmin can also cleave ADAMTS13, which ultimately inactivates it. This leaves the overall role of plasmin in primary haemostasis uncertain.We investigated the combined molecular effects of plasmin on VWF and ADAMTS13. We first identified that plasmin destroys FRETS-VWF73 substrate by cleaving the ADAMTS13 binding region in a buffered system. We next investigated how plasmin affects both VWF and ADAMTS13 under static conditions in plasma by western blotting. We found that globular VWF is largely protected from plasmin cleavage. However, ADAMTS13 is rapidly cleaved under these conditions, suggesting inactivation. Surprisingly, we observed that plasmin enhances ADAMTS13 activity in a modified two-stage FRETS-VWF73 assay that protects FRETS-VWF73 substrate from degradation. In direct binding studies under the same conditions, we found that plasmin generates multiple C-terminally truncated forms of ADAMTS13 with VWF-binding capacity. In an effort to seek evidence for this mechanism in vivo, we analysed plasma from patients with systemic amyloidosis, which is hallmarked by a hyperfibrinolytic state. We found that their plasma contained increased levels of C-terminally truncated forms of ADAMTS13, which correlated with their hyperfibrinolytic state.We propose that truncation of ADAMTS13 by plasmin abolishes intramolecular self-association, which improves interaction with unfolded VWF.


Assuntos
Proteína ADAMTS13/química , Amiloidose/imunologia , Fibrinolisina/química , Proteínas Recombinantes/química , Fator de von Willebrand/química , Proteína ADAMTS13/sangue , Motivos de Aminoácidos , Amiloidose/sangue , Sítios de Ligação , Fibrinólise , Humanos , Immunoblotting , Peptídeos/química , Ligação Proteica , Desnaturação Proteica , Domínios Proteicos , Dobramento de Proteína , Proteínas Recombinantes/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA