Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.594
Filtrar
1.
PLoS One ; 19(9): e0310883, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39331637

RESUMO

The management of factor Xa (FXa) inhibitor-associated bleeding remains a clinical challenge. Massive bleeding is often associated with complex coagulopathy and, thus, the sole reversal of FXa inhibitors might not be sufficient to restore hemostasis, requiring instead a multimodal approach. Four-factor prothrombin complex concentrate (4F-PCC) is widely recognized as a viable treatment option for FXa inhibitor-associated bleeding. Here, we applied computational models to explore the effect 4F-PCC has on the coagulation cascade and restoration of thrombin generation in a system that simulates a patient that has received a FXa inhibitor. The coagulation model is largely based on a previously developed model with modifications incorporated from various other published sources. The model was calibrated and validated using data from a phase 3 clinical trial of vitamin K antagonist reversal with 4F-PCC. Using the parameters and initial conditions determined during the calibration and validation process, the prothrombin time (PT) test simulations predicted a PT of 11.4 seconds. The model successfully simulated the effects of rivaroxaban and apixaban on total thrombin concentration and showed that 4F-PCC increased thrombin generation in the presence of rivaroxaban or apixaban.


Assuntos
Fatores de Coagulação Sanguínea , Inibidores do Fator Xa , Hemorragia , Trombina , Humanos , Inibidores do Fator Xa/efeitos adversos , Inibidores do Fator Xa/uso terapêutico , Hemorragia/tratamento farmacológico , Hemorragia/induzido quimicamente , Trombina/metabolismo , Rivaroxabana/efeitos adversos , Pirazóis/efeitos adversos , Piridonas/efeitos adversos , Coagulação Sanguínea/efeitos dos fármacos , Fator Xa/metabolismo , Modelos Biológicos , Tempo de Protrombina
2.
Sci Rep ; 14(1): 22335, 2024 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-39333740

RESUMO

We developed a covalent antithrombin-heparin complex (ATH) with superior In vivo anticoagulant efficacy compared to non-covalent antithrombin (AT) + unfractionated heparin (H). Previous in vitro studies of ATH, investigating the mechanisms behind its efficacy, were done in the absence of endothelium. Since the endothelial surface modulates hemostasis, we investigated its impact on the in vitro anticoagulant properties of ATH and AT+H. Discontinuous second order rate constant enzyme inhibition assays, fibrin formation, and plasma clot generation were performed in the presence of ATH or AT+H, with and without endothelium present. ATH had an increased rate of direct inhibition of IIa and Xa, and increased inhibition of IIa-induced fibrin formation, compared to AT+H. When compared at equal anti-Xa levels, ATH was less effective than AT+H at catalyzing inhibition of plasma clot generation. These results were found in both the presence and absence of endothelium. Endothelium decreased the rate of IIa inhibition, and reduced clot time in IIa-induced fibrin formation and plasma clot generation assays, for both ATH and AT+H. Endothelium did not impact the activity of ATH differently to AT+H. This supports the growing body of evidence suggesting ATH may be a beneficial anticoagulant for potential clinical use.


Assuntos
Anticoagulantes , Antitrombinas , Coagulação Sanguínea , Heparina , Heparina/farmacologia , Heparina/química , Antitrombinas/farmacologia , Antitrombinas/química , Anticoagulantes/farmacologia , Anticoagulantes/química , Humanos , Coagulação Sanguínea/efeitos dos fármacos , Fibrina/metabolismo , Fator Xa/metabolismo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo
3.
Ann Intern Med ; 177(9): JC101, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39222512

RESUMO

SOURCE CITATION: Connolly SJ, Sharma M, Cohen AT, et al; ANNEXA-I Investigators. Andexanet for factor Xa inhibitor-associated acute intracerebral hemorrhage. N Engl J Med. 2024;390:1745-1755. 38749032.


Assuntos
Inibidores do Fator Xa , Trombose , Inibidores do Fator Xa/efeitos adversos , Inibidores do Fator Xa/uso terapêutico , Humanos , Trombose/induzido quimicamente , Hemorragia Cerebral/induzido quimicamente , Masculino , Proteínas Recombinantes , Fator Xa
4.
Hepatol Commun ; 8(8)2024 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-39101776

RESUMO

BACKGROUND: Cholestatic liver diseases induce local and systemic hypercoagulation, with neutrophil extracellular traps (NETs) serving as major drivers. These NETs have been linked to decreased liver function in patients with obstructive jaundice. However, the impact of NETs on liver hypercoagulation in cholestatic liver disease remains unknown. METHODS: We utilized bile duct ligation to create experimental mice and analyzed NETs formation in the liver. Fibrin deposition, tissue factor expression, and inflammation in the liver were visualized through western blot and immunohistochemical techniques. LSECs were incubated with isolated NETs, and we detected endothelial procoagulant activity using coagulation protein production assays and measuring endothelial permeability. In both in vivo and in vitro settings, DNase I was applied to clarify the effect of NETs on intrahepatic hypercoagulability, hepatotoxicity, LSEC, and macrophage activation or injury. RESULTS: Bile duct ligation mice exhibited significantly increased levels of NETs in liver tissue, accompanied by neutrophil infiltration, tissue necrosis, fibrin deposition, and thrombophilia compared to sham mice. Notably, NETs resulted in phosphatidylserine and tissue factor exposure on LSEC, enhancing coagulation Factor Xa and thrombin production. The enhanced procoagulant activity could be reversed by degrading NETs with DNase I. Additionally, NETs-induced permeability changes in LSECs, characterized by increased VE-cadherin expression and F-actin retraction, which could be rescued by DNase I. Meanwhile, NET formation is associated with KC activation and the formation of inflammatory factors. CONCLUSIONS: NETs promote intrahepatic activation of coagulation and inflammation, leading to liver tissue injury. Strategies targeting NET formation may offer a potential therapeutic approach for treating cholestatic liver disease.


Assuntos
Armadilhas Extracelulares , Fígado , Trombose , Armadilhas Extracelulares/metabolismo , Animais , Camundongos , Fígado/patologia , Fígado/metabolismo , Trombose/etiologia , Trombose/patologia , Colestase/patologia , Colestase/complicações , Modelos Animais de Doenças , Masculino , Tromboplastina/metabolismo , Trombofilia/etiologia , Trombofilia/sangue , Fibrina/metabolismo , Camundongos Endogâmicos C57BL , Neutrófilos/metabolismo , Humanos , Infiltração de Neutrófilos , Fator Xa/metabolismo , Trombina/metabolismo
5.
Protein J ; 43(4): 888-909, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39095592

RESUMO

The current investigation focused on separating Cerastes cerastes venom to produce the first Kunitz-type peptide. Based on its anti-trypsin effect, Cerastokunin, a 7.75 kDa peptide, was purified until homogenity by three steps of chromatography. Cerastokunin was found to include 67 amino acid residues that were obtained by de novo sequencing using LC-MALDI-MSMS. Upon alignment with Kunitz-type peptides, there was a high degree of similarity. Cerastokunin's 3D structure had 12% α-helices and 21% ß-strands with pI 8.48. Cerastokunin showed a potent anticoagulant effect by inhibiting the protease activity of thrombin and trypsin as well as blocking the intrinsic and extrinsic coagulation pathways. In both PT and aPPT, Cerastokunin increased the blood clotting time in a dose-dependent way. Using Lys48 and Gln192 for direct binding, Cerastokunin inhibited thrombin, Factor Xa and trypsin as shown by molecular docking. Cerastokunin exhibited a dose-response blockade of PARs-dependent pathway platelet once stimulated by thrombin. An increased concentration of Cerastokunin resulted in a larger decrease of tail thrombus in the mice-carrageenan model in an in vivo investigation when compared to the effects of antithrombotic medications. At all Cerastokunin doses up to 6 mg/kg, no in vivo toxicity was seen in challenged mice over the trial's duration.


Assuntos
Plaquetas , Inibidores do Fator Xa , Trombina , Animais , Humanos , Camundongos , Sequência de Aminoácidos , Anticoagulantes/farmacologia , Anticoagulantes/química , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Fator Xa/química , Fator Xa/metabolismo , Inibidores do Fator Xa/farmacologia , Inibidores do Fator Xa/química , Simulação de Acoplamento Molecular , Trombina/química , Trombina/metabolismo , Masculino
6.
J Theor Biol ; 594: 111925, 2024 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-39142600

RESUMO

Prothrombinase complex, composed of coagulation factors Xa (FXa) and Va (FVa) is a major enzyme of the blood coagulation network that produces thrombin via activation of its inactive precursor prothrombin (FII) on the surface of phospholipid membranes. However, pathways and mechanisms of prothrombinase formation and substrate delivery are still discussed. Here we designed a novel mathematical model that considered different potential pathways of FXa or FII binding (from the membrane or from solution) and analyzed the kinetics of thrombin formation in the presence of a wide range of reactants concentrations. We observed the inhibitory effect of large FVa concentrations and this effect was phospholipid concentration-dependent. We predicted that efficient FII activation occurred via formation of the ternary complex, in which FVa, FXa and FII were in the membrane-bound state. Prothrombin delivery was mostly membrane-dependent, but delivery from solution was predominant under conditions of phospholipid deficiency or FXa/FVa excess. Likewise, FXa delivery from solution was predominant in the case of FVa excess, but high FII did not switch the FXa delivery to the solution-dependent one. Additionally, the FXa delivery pathway did not depend on the phospholipid concentration, being the membrane-dependent one even in case of the phospholipid deficiency. These results suggest a flexible mechanism of prothrombinase functioning which utilizes different complex formation and even inhibitory mechanisms depending on conditions.


Assuntos
Fator Xa , Protrombina , Cinética , Humanos , Fator Xa/metabolismo , Protrombina/metabolismo , Modelos Biológicos , Fosfolipídeos/metabolismo , Coagulação Sanguínea/fisiologia , Trombina/metabolismo , Fator Va/metabolismo , Tromboplastina/metabolismo , Especificidade por Substrato , Fator V
11.
J Thromb Haemost ; 22(10): 2767-2772, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39002733

RESUMO

BACKGROUND: All current X-ray structures of factor (F)Xa are devoid of the γ-carboxyglutamate (Gla) domain and fail to reveal the overall conformation of the free protein. The recent cryogenic electron microscopy (cryo-EM) structure of FXa in the prothrombinase complex is the only structure of full-length FXa and shows that the Gla domain is positioned at an angle relative to the epidermal growth factor 1 domain. OBJECTIVES: Establish if the curved conformation of FXa revealed by cryo-EM is also present in solution. METHODS: The conformation of FXa in solution was studied by single-molecule Förster resonance energy transfer. RESULTS: The conformation of full-length FXa in solution is resolved for the first time. The conformation is curved and extremely sensitive to Ca2+. It does not differ significantly from its zymogen form or from that present in the prothrombinase complex free or bound to the physiologic substrates prothrombin and meizothrombin. CONCLUSION: Measurements by single-molecule Förster resonance energy transfer reveal that FXa has a curved conformation in solution, free or bound to physiologic ligands, and validate the recent cryo-EM structures of prothrombinase. The drastic conformational changes observed in the absence of Ca2+ suggest that the structural architecture of FXa changes upon administration of vitamin K antagonists that perturb the interaction of the Gla domain with divalent cations.


Assuntos
Fator Xa , Conformação Proteica , Humanos , Cálcio/metabolismo , Cálcio/química , Microscopia Crioeletrônica , Fator V , Fator Xa/metabolismo , Fator Xa/química , Transferência Ressonante de Energia de Fluorescência , Modelos Moleculares , Ligação Proteica , Protrombina/química , Protrombina/metabolismo , Imagem Individual de Molécula/métodos , Tromboplastina/metabolismo , Tromboplastina/química
12.
J Immunother Cancer ; 12(7)2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-39060025

RESUMO

BACKGROUND: The high metastasis rate is one of the main reasons for the poor prognosis of patients with hepatocellular carcinoma (HCC). Coagulation factor Xa (FXa) and its receptor proteinase-activated receptor-2 (PAR-2) proven to promote tumor metastasis in other forms of cancer. Here, we explore the role and mechanism of FXa in the regulation of resistance of anoikis and immune escape of HCC. METHODS: In vitro and in vivo experiments were conducted to explore the role of FXa in HCC metastasis and its potential mechanism. The effects of FXa inhibitor rivaroxaban on HCC immunotherapy were evaluated using intrahepatic metastasis animal models and clinical trial (No. ChiCTR20000040540). We investigated the potential of FXa inhibition as a treatment for HCC. RESULTS: FXa was highly expressed in HCC and promoted metastasis by activating PAR-2. Mechanistically, FXa-activated PAR-2 endows HCC cells with the ability of anoikis resistance to survive in the circulating blood by inhibiting the extrinsic apoptosis pathway. Furthermore, suspension stimulation-induced phosphorylation of STAT2, which promotes programmed death-ligand 1 (PD-L1) transcription and inhibits the antitumor effects of immune cells by inhibiting the infiltration of CD8+T cells in tumors and the levels of secreted cytokines. In vivo inhibition of FXa with rivaroxaban reduced HCC metastasis by decreasing PD-L1 expression and exhausting tumor-infiltrating lymphocytes. Notably, the combination of rivaroxaban and anti-programmed death-1 monoclonal antibody (anti-PD-1) programmed Death-1 monoclonal antibody (anti-PD-1) induced synergistic antitumor effects in animal models. Most importantly, rivaroxaban improved the objective response rate of patients with HCC to immune checkpoint inhibitors and prolonged overall survival time. CONCLUSIONS: FXa-activated PAR-2 promotes anoikis resistance and immune escape in HCC, suggesting the potential for combining coagulation inhibitors and PD-1/PD-L1 immune checkpoint blockade to enhance the therapeutic efficacy of HCC.


Assuntos
Anoikis , Antígeno B7-H1 , Carcinoma Hepatocelular , Imunoterapia , Neoplasias Hepáticas , Receptor PAR-2 , Evasão Tumoral , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Humanos , Receptor PAR-2/metabolismo , Animais , Camundongos , Imunoterapia/métodos , Antígeno B7-H1/metabolismo , Antígeno B7-H1/antagonistas & inibidores , Fator Xa/metabolismo , Fator Xa/farmacologia , Fator Xa/uso terapêutico , Masculino , Feminino , Linhagem Celular Tumoral , Inibidores do Fator Xa/farmacologia , Inibidores do Fator Xa/uso terapêutico
13.
J Thromb Haemost ; 22(10): 2889-2899, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39029742

RESUMO

The currently approved direct oral anticoagulants (DOACs) are increasingly used in clinical practice. Although serious bleeding risks are lower with DOACs than with vitamin K antagonists, bleeding remains the most frequent side effect. Andexanet alfa and idarucizumab are the currently approved specific reversal agents for oral factor (F)Xa inhibitors and dabigatran, respectively. Our prior guidance document was published in 2016, but with more information available on the utility and increased use of these reversal agents and other bleeding management strategies, we have updated this International Society on Thrombosis and Haemostasis guidance document on DOAC reversal. In this narrative review, we compare the mechanism of action of specific and nonspecific reversal agents, review the clinical data supporting their use, and provide guidance on when reversal is indicated. In addition, we briefly discuss the reversal of oral FXIa inhibitors, a new class of DOACs currently under clinical development.


Assuntos
Anticorpos Monoclonais Humanizados , Coagulação Sanguínea , Inibidores do Fator Xa , Hemorragia , Humanos , Administração Oral , Hemorragia/induzido quimicamente , Inibidores do Fator Xa/efeitos adversos , Inibidores do Fator Xa/administração & dosagem , Inibidores do Fator Xa/uso terapêutico , Coagulação Sanguínea/efeitos dos fármacos , Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/administração & dosagem , Proteínas Recombinantes/uso terapêutico , Proteínas Recombinantes/administração & dosagem , Anticoagulantes/efeitos adversos , Anticoagulantes/administração & dosagem , Anticoagulantes/uso terapêutico , Dabigatrana/administração & dosagem , Dabigatrana/efeitos adversos , Fator Xa/uso terapêutico , Fatores de Risco , Resultado do Tratamento , Arginina/análogos & derivados , Piperazinas
14.
Subcell Biochem ; 104: 409-423, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38963494

RESUMO

The hemostatic response to vascular injury entails a sequence of proteolytic events where several inactive zymogens of the trypsin family are converted to active proteases. The cascade starts with exposure of tissue factor from the damaged endothelium and culminates with conversion of prothrombin to thrombin in a reaction catalyzed by the prothrombinase complex composed of the enzyme factor Xa, cofactor Va, Ca2+, and phospholipids. This cofactor-dependent activation is paradigmatic of analogous reactions of the blood coagulation and complement cascades, which makes elucidation of its molecular mechanism of broad significance to the large class of trypsin-like zymogens to which prothrombin belongs. Because of its relevance as the most important reaction in the physiological response to vascular injury, as well as the main trigger of pathological thrombotic complications, the mechanism of prothrombin activation has been studied extensively. However, a molecular interpretation of this mechanism has become available only recently from important developments in structural biology. Here we review current knowledge on the prothrombin-prothrombinase interaction and outline future directions for the study of this key reaction of the coagulation cascade.


Assuntos
Coagulação Sanguínea , Protrombina , Tromboplastina , Humanos , Protrombina/metabolismo , Protrombina/química , Tromboplastina/metabolismo , Tromboplastina/química , Coagulação Sanguínea/fisiologia , Animais , Ligação Proteica , Fator Xa/metabolismo , Fator V
15.
Crit Care ; 28(1): 221, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38970010

RESUMO

BACKGROUND: There is currently a lack of evidence for the comparative effectiveness of Andexanet alpha and four-factor prothrombin complex concentrate (4F-PCC) in anticoagulation reversal of direct oral anticoagulants (DOACs). The primary aim of our systematic review was to verify which drug is more effective in reducing short-term all-cause mortality. The secondary aim was to determine which of the two reverting strategies is less affected by thromboembolic events. METHODS: A systematic review and meta-analysis was performed. RESULTS: Twenty-two studies were analysed in the systematic review and quantitative synthesis. In all-cause short-term mortality, Andexanet alpha showed a risk ratio (RR) of 0.71(95% CI 0.37-1.34) in RCTs and PSMs, compared to 4F-PCC (I2 = 81%). Considering the retrospective studies, the pooled RR resulted in 0.84 (95% CI 0.69-1.01) for the common effects model and 0.82 (95% CI 0.63-1.07) for the random effects model (I2 = 34.2%). Regarding the incidence of thromboembolic events, for RCTs and PSMs, the common and the random effects model exhibited a RR of 1.74 (95% CI 1.09-2.77), and 1.71 (95% CI 1.01-2.89), respectively, for Andexanet alpha compared to 4F-PCC (I2 = 0%). Considering the retrospective studies, the pooled RR resulted in 1.21 (95% CI 0.87-1.69) for the common effects model and 1.18 (95% CI 0.86-1.62) for the random effects model (I2 = 0%). CONCLUSION: Considering a large group of both retrospective and controlled studies, Andexanet alpha did not show a statistically significant advantage over 4F-PCC in terms of mortality. In the analysis of the controlled studies alone, Andexanet alpha is associated with an increased risk of thromboembolic events. CLINICAL TRIAL REGISTRATION: PROSPERO: International prospective register of systematic reviews, 2024, CRD42024548768.


Assuntos
Anticoagulantes , Fatores de Coagulação Sanguínea , Humanos , Anticoagulantes/uso terapêutico , Anticoagulantes/efeitos adversos , Fatores de Coagulação Sanguínea/uso terapêutico , Fatores de Coagulação Sanguínea/farmacologia , Fator Xa/uso terapêutico , Inibidores do Fator Xa/uso terapêutico , Inibidores do Fator Xa/efeitos adversos , Proteínas Recombinantes , Tromboembolia/prevenção & controle
16.
Kyobu Geka ; 77(6): 454-456, 2024 Jun.
Artigo em Japonês | MEDLINE | ID: mdl-39009541

RESUMO

The management of patients on direct oral anticoagulants (DOACs) who require an emergency cardiac surgery has been disputed in Japan. Recently, the use of andexanet alfa as an antidote for apixaban and rivaroxaban, is approved in the setting of life-threating or uncontrollable major bleeding. However, the efficacy and safety of andexanet alfa have been investigated. We report a case of 72-year-old man taking rivaroxaban who required the emergency coronary artery bypass grafting. He received andexanet alfa prior to the operation. Heparin resistance was noted before starting cardiopulmonary bypass. Consideration should be given to the use of andexanet alfa before or during cardiopulmonary bypass.


Assuntos
Heparina , Proteínas Recombinantes , Humanos , Idoso , Masculino , Heparina/administração & dosagem , Proteínas Recombinantes/administração & dosagem , Resistência a Medicamentos , Anticoagulantes/administração & dosagem , Anticoagulantes/uso terapêutico , Anticoagulantes/efeitos adversos , Fator Xa , Ponte de Artéria Coronária , Rivaroxabana/administração & dosagem , Rivaroxabana/uso terapêutico
17.
Proc Natl Acad Sci U S A ; 121(24): e2403389121, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38833471

RESUMO

Cell-cell fusion mediated by most paramyxovirus requires fusion protein (F) and attachment protein (H, HN, or G). The F protein is proteolytic cleaved to be fusogenically active. J paramyxovirus (JPV) has a unique feature in the family Paramyxoviridae: It encodes an integral membrane protein, syncytial protein (SP, formerly known as transmembrane protein, TM), which is essential in JPV-promoted cell-cell fusion (i.e., syncytial). In this study, we report that cleavage of SP is essential for its syncytial-promoting activity. We have identified the cleavage site of SP at amino acid residues 172 to 175, LKTG, and deletion of the "LKTG" residues abolished SP protein cleavage and its ability to promote cell-cell fusion. Replacing the cleavage site LKTG with a factor Xa protease cleavage site allows cleavage of the SP with factor Xa protease and restores its ability to promote cell-cell fusion. Furthermore, results from a hemifusion assay indicate that cleavage of SP plays an important role in the progression from the intermediate hemifusion state to a complete fusion. This work indicates that SP has many characteristics of a fusion protein. We propose that SP is likely a cell-cell fusion-promoting protein.


Assuntos
Fusão Celular , Proteínas Virais de Fusão , Animais , Proteínas Virais de Fusão/metabolismo , Chlorocebus aethiops , Proteólise , Células Vero , Internalização do Vírus , Fator Xa/metabolismo , Humanos , Linhagem Celular
18.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 36(5): 461-464, 2024 May.
Artigo em Chinês | MEDLINE | ID: mdl-38845490

RESUMO

The incidence and mortality of venous thromboembolism (VTE) are high in critically ill patients, and there is still a risk of VTE and bleeding after the use of fixed-dose low molecular weight heparin (LMWH) for prophylaxis. The level of anti-factor Xa is not up to standard after LMWH prophylaxis in patients with surgery or trauma. The condition of critically ill patients is complicated, and the proportion of patients with low antithrombin III is high, which can affect the prophylactic efficacy of LMWH and contribute to VTE occurrence. There is currently no consensus on whether adjusting LMWH dose according to anti-factor Xa levels can reduce VTE occurrence in critically ill patients. High-quality multicenter randomized controlled studies are needed in the future to establish new approaches for precise prevention of VTE in critically ill patients.


Assuntos
Estado Terminal , Heparina de Baixo Peso Molecular , Tromboembolia Venosa , Humanos , Tromboembolia Venosa/prevenção & controle , Tromboembolia Venosa/etiologia , Heparina de Baixo Peso Molecular/uso terapêutico , Heparina de Baixo Peso Molecular/administração & dosagem , Anticoagulantes/uso terapêutico , Anticoagulantes/administração & dosagem , Inibidores do Fator Xa/uso terapêutico , Inibidores do Fator Xa/administração & dosagem , Fator Xa
19.
ACS Nano ; 18(26): 17018-17030, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38845136

RESUMO

The advantageous optical properties of quantum dots (QDs) motivate their use in a wide variety of applications related to imaging and bioanalysis, including the detection of proteases and their activity. Recent studies have shown that surface chemistry on QDs is able to modulate protease activity, but only nonspecifically. Here, we present a strategy to selectively accelerate the activity of a particular target protease by as much as two orders of magnitude. Exosite-binding "bait" peptides were derived from proteins that span a range of biological roles─substrate, receptor, and inhibitor─and were used to increase the affinity of the QD-peptide conjugates for either thrombin or factor Xa, resulting in increased rates of proteolysis for coconjugated substrates. Unlike effects from QD surface chemistry, the acceleration was specific to the target protease with negligible acceleration of other proteases. Benefits of this "bait and cleave" sensing approach included detection limits that improved by more than an order of magnitude, reenabled detection of target protease against an overwhelming background of nontarget proteolysis, and mitigation of the action of inhibitors. The cumulative results point to a generalizable strategy, where the mechanism of acceleration, considerations for the design of bait peptides and conjugates, and routes to expanding the scope of this approach are discussed. Overall, this research represents a major step forward in the rational design of nanoparticle-based enzyme sensors that enhance sensitivity and selectivity.


Assuntos
Peptídeos , Pontos Quânticos , Trombina , Pontos Quânticos/química , Peptídeos/química , Peptídeos/metabolismo , Trombina/metabolismo , Trombina/análise , Trombina/química , Fator Xa/metabolismo , Fator Xa/química , Proteólise , Humanos , Propriedades de Superfície , Peptídeo Hidrolases/metabolismo , Peptídeo Hidrolases/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...