Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.944
Filter
1.
Int J Med Mushrooms ; 26(6): 53-68, 2024.
Article in English | MEDLINE | ID: mdl-38801087

ABSTRACT

The purification of a fibrinolytic enzyme from the fruiting bodies of wild-growing medicinal mushroom, Pycnoporus coccineus was achieved through a two-step procedure, resulting in its homogeneity. This purification process yielded a significant 4.13-fold increase in specific activity and an 8.0% recovery rate. The molecular weight of P. coccineus fibrinolytic enzyme (PCFE) was estimated to be 23 kDa using sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis. PCFE demonstrated its optimal activity at a temperature of 40 °C and pH 8. Notably, the enzymatic activity was inhibited by the presence of zinc or copper metal ions, as well as serine protease inhibitors, such as phenylmethylsulfonyl fluoride and 4-amidinophenylmethanesulfonyl fluoride. PCFE exhibited remarkable specificity towards a synthetic chromogenic substrate for thrombin. The enzyme demonstrated the Michaelis-Menten constant (Km), maximal velocity (V ), and catalytic rate constant (Kcat) values of 3.01 mM, 0.33 mM min-1 µg-1, and 764.1 s-1, respectively. In vitro assays showed PCFE's ability to effectively degrade fibrin and blood clots. The enzyme induced alterations in the density and structural characteristics of fibrin clots. PCFE exhibited significant effects on various clotting parameters, including recalcification time, activated partial thromboplastin time, prothrombin time, serotonin secretion from thrombin-activated platelets, and thrombin-induced acute thromboembolism. These findings suggest that P. coccineus holds potential as an antithrombotic biomaterials and resources for cardiovascular research.


Subject(s)
Fibrinolytic Agents , Pycnoporus , Serine Proteases , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/isolation & purification , Fibrinolytic Agents/chemistry , Serine Proteases/isolation & purification , Serine Proteases/pharmacology , Serine Proteases/metabolism , Serine Proteases/chemistry , Animals , Pycnoporus/enzymology , Molecular Weight , Fruiting Bodies, Fungal/chemistry , Hydrogen-Ion Concentration , Temperature , Humans , Fibrin/metabolism , Fungal Proteins/isolation & purification , Fungal Proteins/chemistry , Fungal Proteins/pharmacology
2.
J Vis Exp ; (206)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38709073

ABSTRACT

Fibrinogenolytic agents that can dissolve fibrinogen directly have been widely used in anti-coagulation treatment. Generally, identifying new fibrinogenolytic agents requires the separation of each component first and then checking their fibrinogenolytic activities. Currently, polyacrylamide gel electrophoresis (PAGE) and chromatography are mostly used in the separating stage. Meanwhile, the fibrinogen plate assay and reaction products based PAGE are usually adopted to display their fibrinogenolytic activities. However, because of the spatiotemporal separation of those two stages, it is impossible to separate and display the active fibrinogenolytic agents with the same gel. To simplify the separating and displaying processes of fibrinogenolytic agent identification, we constructed a new fibrinogen-PAGE method to rapidly separate and display the fibrinogenolytic agents of peanut worms (Sipunculus nudus) in this study. This method includes fibrinogen-PAGE preparation, electrophoresis, renaturation, incubation, staining, and decolorization. The fibrinogenolytic activity and molecular weight of the protein can be detected simultaneously. According to this method, we successfully detected more than one active fibrinogenolytic agent of peanut wormhomogenate within 6 h. Moreover, this fibrinogen-PAGE method is time and cost-friendly. Furthermore, this method could be used to study the fibrinogenolytic agents of the other organisms.


Subject(s)
Electrophoresis, Polyacrylamide Gel , Fibrinogen , Fibrinogen/chemistry , Fibrinogen/metabolism , Animals , Electrophoresis, Polyacrylamide Gel/methods , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/isolation & purification
3.
Phytochemistry ; 223: 114109, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38697239

ABSTRACT

A previously undescribed open-loop decarbonizing cembranolide, sarcocinerenolide A, and eight undescribed cembranolides, sarcocinerenolides B-I, characterized by poly-membered oxygen ring fragments were isolated from the soft coral Sarcophyton cinereum collected from the South China Sea. The structures and absolute configurations of these previously undescribed compounds were precisely determined by analysis of NMR data, DP4+ and ECD spectra. The bioactivities of the compounds were evaluated using zebrafish models and sarcocinerenolides C and H exhibited anti-thrombotic activity.


Subject(s)
Anthozoa , Diterpenes , Animals , Anthozoa/chemistry , Diterpenes/chemistry , Diterpenes/pharmacology , Diterpenes/isolation & purification , Molecular Structure , Zebrafish , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/isolation & purification , China , Structure-Activity Relationship
4.
Vet Med Sci ; 10(4): e1448, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38818763

ABSTRACT

BACKGROUND: Recombinant intracameral tissue plasminogen activator (rTPA) administration can aid clearance of fibrin from the anterior chamber. MATERIALS AND METHODS: In this retrospective multicentre case series, the effect of intracameral rTPA administration to treat fibrin in the anterior chamber resulting from trauma or inflammatory ocular disease was evaluated. Clinical data from 30 treatments in 29 horses were obtained from medical records from 2003 to 2022. Association between time from onset of clinical signs and time for rTPA treatment to effect was studied with regression analysis. RESULTS: Twenty-seven horses (93.1%) had no previous history of ophthalmic disease; one had an iridic cyst, and another had equine recurrent uveitis. The majority of cases were related to trauma (79.3%). Median time from the onset of clinical signs to treatment was 12 h (IQR = 4-48 h). rTPA (72% 20 µg; 24% 25 µg; 3.3% 40 µg) was administered once in all but one eye, which was treated twice. Resolution of fibrin was seen in 96.9% (29/30) of treatments. Fibrin accumulation recurred in one case but resolved 14 days after the second treatment. Complications were seen in four treatments (13.3%): moderate pain for 24 h, intracameral debris and mild intracameral haemorrhage in a horse that received 40 µg of tissue plasminogen activator. Recurrence of fibrin accumulation was absent in 96.7% of cases. Median time to effect was 20 min (IQR = 10-45 min). Time for rTPA treatment to effect was not associated with time from fibrin formation (R2 = 0.09; p = 0.11). CONCLUSION: Intracameral rTPA treatment can be considered at 20-25 µg in 0.1 mL solution to aid resolution of fibrin accumulation.


Subject(s)
Anterior Chamber , Fibrin , Horse Diseases , Tissue Plasminogen Activator , Animals , Horses , Tissue Plasminogen Activator/administration & dosage , Horse Diseases/drug therapy , Retrospective Studies , Female , Male , Anterior Chamber/drug effects , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/administration & dosage , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Eye Diseases/veterinary , Eye Diseases/drug therapy
5.
Platelets ; 35(1): 2353582, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38773939

ABSTRACT

Platelets are central to thrombosis. Research at the intersection of biological and physical sciences provides proof-of-concept for shear rate-dependent platelet slip at vascular stenosis and near device surfaces. Platelet slip extends the observed biological "slip-bonds" to the boundary of functional gliding without contact. As a result, there is diminished engagement of the coagulation cascade by platelets at these surfaces. Comprehending platelet slip would more precisely direct antithrombotic regimens for different shear environments, including for percutaneous coronary intervention (PCI). In this brief report we promote translation of the proof-of-concept for platelet slip into improved antithrombotic regimens by: (1) reviewing new supporting basic biological science and clinical research for platelet slip; (2) hypothesizing the principal variables that affect platelet slip; (3) applying the consequent construct model in support of-and in some cases to challenge-relevant contemporary guidelines and their foundations (including for urgent, higher-risk PCI); and (4) suggesting future research pathways (both basic and clinical). Should future research demonstrate, explain and control platelet slip, then a paradigm shift for choosing and recommending antithrombotic regimens based on predicted shear rate should follow. Improved clinical outcomes with decreased complications accompanying this paradigm shift for higher-risk PCI would also result in substantive cost savings.


Subject(s)
Blood Platelets , Humans , Blood Platelets/metabolism , Blood Platelets/drug effects , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use
6.
Int J Biol Macromol ; 270(Pt 1): 132278, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38750856

ABSTRACT

Leeches secrete various biologically active substances which have important medical and pharmaceutical values in antithrombotic treatments. Here, we provide a high quality genome of two Asian medicinal leeches Hirudo nipponia and Hirudo tianjinensis, based on which, we identified 22 antithrombotic gene families, including fourteen coagulation inhibitors, four platelet aggregation inhibitors, three fibrinolysis enhancers, and one tissue penetration enhancer. The total numbers of antithrombotic genes were similar between H. nipponia (N = 86) and H. tianjinensis (N = 83). Molecular evolution analysis showed that no significant differences were detected between the two species in any of the three selection indices (dN, dS, and dN/dS), nor in the number of sites under positive/purifying selection. RNA-Seq based gene expression analysis showed that the overall expression patterns of the antithrombotic gene families were not significantly deviated between the two species. Our results indicated that there were rather close similarities between the two leeches on genomic characteristics, especially for the molecular evolution and expression of antithrombotic genes. Our study provides the most comprehensive collection of antithrombotic biomacromolecules from the two Asian medicinal leeches to date. These results will greatly facilitate the research and application of leech derivatives for medical and pharmaceutical purposes of thrombosis.


Subject(s)
Fibrinolytic Agents , Genomics , Leeches , Animals , Leeches/genetics , Genomics/methods , Fibrinolytic Agents/pharmacology , Phylogeny , Evolution, Molecular , Genome
7.
Int J Mol Sci ; 25(7)2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38612796

ABSTRACT

The development of novel anticoagulants requires a comprehensive investigational approach that is capable of characterizing different aspects of antithrombotic activity. The necessary experiments include both in vitro assays and studies on animal models. The required in vivo approaches include the assessment of pharmacokinetic and pharmacodynamic profiles and studies of hemorrhagic and antithrombotic effects. Comparison of anticoagulants with different mechanisms of action and administration types requires unification of the experiment scheme and its adaptation to existing laboratory conditions. The rodent thrombosis models in combination with the assessment of hemostasis parameters and hematological analysis are the classic methods for conducting preclinical studies. We report an approach for the comparative study of the activity of different anticoagulants in vivo, including the investigation of pharmacodynamics and the assessment of hemorrhagic effects (tail-cut bleeding model) and pathological thrombus formation (inferior vena cava stenosis model of venous thrombosis). The reproducibility and uniformity of our set of experiments were illustrated on unfractionated heparin and dabigatran etexilate (the most common pharmaceuticals in antithrombic therapy) as comparator drugs and an experimental drug variegin from the tick Amblyomma variegatum. Variegin is notorious since it is a potential analogue of bivalirudin (Angiomax, Novartis AG, Basel, Switzerland), which is now being actively introduced into antithrombotic therapy.


Subject(s)
Anticoagulants , Heparin , Animals , Pharmaceutical Preparations , Anticoagulants/pharmacology , Anticoagulants/therapeutic use , Heparin/pharmacology , Heparin/therapeutic use , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use , Reproducibility of Results
8.
Mar Drugs ; 22(4)2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38667765

ABSTRACT

Marine natural products are important sources of novel drugs. In this study, we isolated 4-hydroxyphenylacetic acid (HPA) from the marine-derived fungus Emericellopsis maritima Y39-2. The antithrombotic activity and mechanism of HPA were reported for the first time. Using a zebrafish model, we found that HPA had a strong antithrombotic activity because it can significantly increase cardiac erythrocytes, blood flow velocity, and heart rate, reduce caudal thrombus, and reverse the inflammatory response caused by Arachidonic Acid (AA). Further transcriptome analysis and qRT-PCR validation demonstrated that HPA may regulate autophagy by inhibiting the PI3K/AKT/mTOR signaling pathway to exert antithrombotic effects.


Subject(s)
Autophagy , Fibrinolytic Agents , Phenylacetates , Zebrafish , Animals , Phenylacetates/pharmacology , Autophagy/drug effects , Fibrinolytic Agents/pharmacology , Signal Transduction/drug effects , Biological Products/pharmacology , Thrombosis/drug therapy , Disease Models, Animal , Aquatic Organisms
9.
Thromb Res ; 238: 41-51, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38669962

ABSTRACT

BACKGROUND AND PURPOSE: Hypolipidemia and platelet activation play key roles in atherosclerotic diseases. Pirinixic acid (WY-14643) was originally developed as a lipid-lowering drug. Here we focused on its antiplatelet and antithrombotic abilities and the underlying mechanism. EXPERIMENTAL APPROACH: The effects of WY-14643 on platelet aggregation was measured using a lumi-aggregometer. Clot retraction and spreading on fibrinogen were also assayed. PPARα-/- platelets were used to identify the target of WY-14643. The interaction between WY-14643 and glycoprotein Ibα (GPIbα) was detected using cellular thermal shift assay (CETSA), surface plasmon resonance (SPR) spectroscopy and molecular docking. GPIbα downstream signaling was examined by Western blot. The antithrombotic effect was investigated using mouse mesenteric arteriole thrombosis model. Mouse tail bleeding model was used to study its effect on bleeding side effects. KEY RESULTS: WY-14643 concentration-dependently inhibits human washed platelet aggregation, clot retraction, and spreading. Significantly, WY-14643 inhibits thrombin-induced activation of human washed platelets with an IC50 of 7.026 µM. The antiplatelet effect of WY-14643 is mainly dependent of GPIbα. CESTA, SPR and molecular docking results indicate that WY-14643 directly interacts with GPIbα and acts as a GPIbα antagonist. WY-14643 also inhibits phosphorylation of PLCγ2, Akt, p38, and Erk1/2 induced by thrombin. Noteworthily, 20 mg/kg oral administration of WY-14643 inhibits FeCl3-induced thrombosis of mesenteric arteries in mice similarly to clopidogrel without increasing bleeding. CONCLUSION AND IMPLICATIONS: WY-14643 is not only a PPARα agonist with lipid-lowering effect, but also an antiplatelet agent as a GPIbα antagonist. It may have more significant therapeutic advantages than current antiplatelet agents for the treatment of atherosclerotic thrombosis, which have lipid-lowering effects without bleeding side effects.


Subject(s)
Fibrinolytic Agents , Platelet Aggregation Inhibitors , Platelet Aggregation , Platelet Glycoprotein GPIb-IX Complex , Pyrimidines , Animals , Mice , Platelet Glycoprotein GPIb-IX Complex/metabolism , Humans , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Platelet Aggregation/drug effects , Thrombosis/drug therapy , Blood Platelets/metabolism , Blood Platelets/drug effects , Male , Molecular Docking Simulation , Mice, Inbred C57BL
10.
J Ethnopharmacol ; 329: 118177, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38604510

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Corydalis decumbens (Thunb.) Pers. was used as stasis-eliminating medicine traditionally to treat cardiovascular disease potentially attributed to its antithrombotic effect, but lack of pharmacological research on it. AIM OF THE STUDY: To investigate the antithrombotic effect of C. decumbens and its preliminary mechanism. MATERIALS AND METHODS: A carrageenan-induced mouse thrombus model and adenosine diphosphate stimulated platelet aggregation of rabbits were used to confirm the inhibitory effect of C. decumbens extract and compounds on thrombosis in vivo. Then, H2O2-induced human umbilical vein endothelial cells (HUVECs) injury model was further adopted to verify the effects of bioactive compounds in vitro. Moreover, in silico network pharmacology analyses and molecular docking were performed to predict the underlying mechanisms, targets, and pathways, and which were further confirmed through western blotting assay. RESULTS: The administration of total extract (TE), total alkaloids (TA) and tetrahydropalmatine (TET) resulted in a significant reduction in black tail thrombus and congestion, along with a decreasing in platelet aggregation of rabbits. A superior antithrombotic effect indicated the bioactive fraction, and then the isolated bioactive compounds, TET and protopine (PRO) increased cell survival, and decreased reactive oxygen species (ROS) and lactate dehydrogenase (LDH) release in H2O2-induced HUVECs injury model. Moreover, the two alkaloids targeted 33 major proteins and influenced 153 pathways in network pharmacology prediction. Among these, HSP90AA1, COX-2, NF-κB/p65, MMP1 and HIF-1α were the key proteins and PI3K-Akt emerged as the major signaling pathway. Further western blotting results supported that five key proteins were downregulated by the two bioactive compounds in H2O2-stimulated HUVECs model. CONCLUSION: C. decumbens exerted protective effect on thrombosis through inhibiting PI3K-Akt pathway and related key proteins, which supported the traditional use and presented potential antithrombotic alkaloids for further investigation.


Subject(s)
Corydalis , Fibrinolytic Agents , Human Umbilical Vein Endothelial Cells , Plant Extracts , Proto-Oncogene Proteins c-akt , Signal Transduction , Thrombosis , Animals , Corydalis/chemistry , Rabbits , Humans , Human Umbilical Vein Endothelial Cells/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Thrombosis/drug therapy , Plant Extracts/pharmacology , Mice , Signal Transduction/drug effects , Male , Fibrinolytic Agents/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Platelet Aggregation/drug effects , Molecular Docking Simulation , Berberine Alkaloids/pharmacology , Hydrogen Peroxide/toxicity , Disease Models, Animal , Carrageenan , Reactive Oxygen Species/metabolism
11.
Eur J Pharm Biopharm ; 199: 114281, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38599299

ABSTRACT

Nattokinase (NK) is a thrombolytic enzyme extracted from natto, which can be used to prevent and treat blood clots. However, it is sensitive to the environment, especially the acidic environment of human stomach acid, and its effect of oral ingestion is minimal. This study aims to increase NK's oral and storage stability by embedding NK in microcapsules prepared with chitosan (CS) and γ-polyglutamic acid (γ-PGA). The paper prepared a double-layer NK oral delivery system by layer self-assembly and characterized its stability and in vitro simulated digestion. According to the research results, the bilayer putamen structure has a protective effect on NK, which not only maintains high activity in various environments (such as acid-base, high temperature) and long-term storage (60 days), but also effectively protects the loaded NK from being destroyed in gastric fluid and achieves its slow release. This work has proved the feasibility of the design of bilayer putamen structure in oral administration and has good fibrolytic activity. Therefore, the novel CS/γ-PGA microcapsules are expected to be used in nutraceutical delivery systems.


Subject(s)
Chitosan , Drug Stability , Fibrinolytic Agents , Polyglutamic Acid , Subtilisins , Chitosan/chemistry , Polyglutamic Acid/chemistry , Polyglutamic Acid/analogs & derivatives , Subtilisins/metabolism , Subtilisins/chemistry , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/pharmacology , Administration, Oral , Humans , Digestion/drug effects , Capsules , Drug Delivery Systems/methods , Drug Compounding/methods , Drug Liberation , Drug Carriers/chemistry
12.
Int J Biol Macromol ; 268(Pt 1): 131742, 2024 May.
Article in English | MEDLINE | ID: mdl-38653430

ABSTRACT

Thrombosis is the main cause of catastrophic events including ischemic stroke, myocardial infarction and pulmonary embolism. Acetylsalicylic acid (ASA) therapy offers a desirable approach to antithrombosis through a reduction of platelet reactivity. However, major bleeding complications, severe off-target side effects, and resistance or nonresponse to ASA greatly attenuate its clinical outcomes. Herein, we report a cationic fibrinogen-mimicking nanoparticle, denoted as ASA-RGD-CS@TPP, to achieve activated-platelet-targeted delivery and efficient release of ASA for safer and more effective antithrombotic therapy. This biomimetic antithrombotic system was prepared by one-pot ionic gelation between cationic arginine-glycine-aspartic acid (RGD)-grafted chitosan (RGD-CS) and anionic tripolyphosphate (TPP). The platform exhibited selective binding to activated platelets, leading to efficient release of ASA and subsequent attenuation of platelet functions, including the remarkable inhibition of platelet aggregation through a potent blockage of cyclooxygenase-1 (COX-1). After intravenous administration, ASA-RGD-CS@TPP displayed significantly prolonged circulation time and successful prevention of thrombosis in a mouse model. ASA-RGD-CS@TPP was demonstrated to significantly enhance antithrombotic therapy while showing minimal coagulation and hemorrhagic risks and excellent biocompatibility in vivo as compared to free ASA. This platform provides a simple, safe, effective and targeted strategy for the development of antithrombotic nanomedicines.


Subject(s)
Blood Platelets , Chitosan , Fibrinogen , Fibrinolytic Agents , Nanoparticles , Chitosan/chemistry , Animals , Nanoparticles/chemistry , Blood Platelets/metabolism , Blood Platelets/drug effects , Mice , Fibrinogen/chemistry , Fibrinogen/metabolism , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/chemistry , Thrombosis/drug therapy , Thrombosis/prevention & control , Drug Liberation , Platelet Activation/drug effects , Aspirin/pharmacology , Aspirin/chemistry , Platelet Aggregation/drug effects , Humans , Cations/chemistry , Male
13.
Arterioscler Thromb Vasc Biol ; 44(6): 1283-1301, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38572646

ABSTRACT

BACKGROUND: Glycoursodeoxycholic acid (GUDCA) has been acknowledged for its ability to regulate lipid homeostasis and provide benefits for various metabolic disorders. However, the impact of GUDCA on arterial thrombotic events remains unexplored. The objective of this study is to examine the effects of GUDCA on thrombogenesis and elucidate its underlying mechanisms. METHODS: Plasma samples from patients with arterial thrombotic events and diet-induced obese mice were collected to determine the GUDCA concentrations using mass spectrometry. Multiple in vivo murine thrombosis models and in vitro platelet functional assays were conducted to comprehensively evaluate the antithrombotic effects of GUDCA. Moreover, lipidomic analysis was performed to identify the alterations of intraplatelet lipid components following GUDCA treatment. RESULTS: Plasma GUDCA level was significantly decreased in patients with arterial thrombotic events and negatively correlated with thrombotic propensity in diet-induced obese mice. GUDCA exhibited prominent suppressing effects on platelet reactivity as evidenced by the attenuation of platelet activation, secretion, aggregation, spreading, and retraction (P<0.05). In vivo, GUDCA administration robustly alleviated thrombogenesis (P<0.05) without affecting hemostasis. Mechanistically, GUDCA inhibited DGK (diacylglycerol kinase) activity, leading to the downregulation of the phosphatidic acid-mediated signaling pathway. Conversely, phosphatidic acid supplementation was sufficient to abolish the antithrombotic effects of GUDCA. More importantly, long-term oral administration of GUDCA normalized the enhanced DGK activity, thereby remarkably alleviating the platelet hyperreactivity as well as the heightened thrombotic tendency in diet-induced obese mice (P<0.05). CONCLUSIONS: Our study implicated that GUDCA reduces platelet hyperreactivity and improves thrombotic propensity by inhibiting DGKs activity, which is a potentially effective prophylactic approach and promising therapeutic agent for arterial thrombotic events.


Subject(s)
Blood Platelets , Diacylglycerol Kinase , Disease Models, Animal , Mice, Inbred C57BL , Thrombosis , Animals , Blood Platelets/drug effects , Blood Platelets/enzymology , Blood Platelets/metabolism , Thrombosis/prevention & control , Thrombosis/blood , Thrombosis/enzymology , Thrombosis/drug therapy , Humans , Male , Diacylglycerol Kinase/antagonists & inhibitors , Diacylglycerol Kinase/metabolism , Mice , Platelet Activation/drug effects , Female , Platelet Aggregation/drug effects , Signal Transduction/drug effects , Middle Aged , Fibrinolytic Agents/pharmacology , Case-Control Studies , Mice, Obese , Obesity/drug therapy , Obesity/enzymology , Obesity/blood , Platelet Aggregation Inhibitors/pharmacology
14.
J Ethnopharmacol ; 329: 118158, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38614263

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Trichosanthis pericarpium (TP; Gualoupi, pericarps of Trichosanthes kirilowii Maxim) has been used in traditional Chinese medicine (TCM) to reduce heat, resolve phlegm, promote Qi, and clear chest congestion. It is also an essential herbal ingredient in the "Gualou Xiebai" formula first recorded by Zhang Zhongjing (from the Eastern Han Dynasty) in the famous TCM classic "Jin-Guì-Yào-Lüe" for treating chest impediments. According to its traditional description, Gualou Xiebai is indicated for symptoms of chest impediments, which correspond to coronary heart diseases (CHD). AIM OF THE STUDY: This study aimed to identify the antithrombotic compounds in Gualoupi for the treatment of CHD. MATERIALS AND METHODS: A CHD rat model was established with a combination of high-fat diet and isoproterenol hydrochloride (ISO) administration via subcutaneous multi-point injection in the back of the neck. This model was used to evaluate the antithrombotic effect of two mainstream cultivars of TP ("HaiShi GuaLou" and "WanLou") by analyzing the main components and their effects. Network pharmacology, molecular docking-based studies, and a zebrafish (Danio rerio) thrombosis model induced by phenylhydrazine was used to validate the antithrombosis components of TP. RESULTS: TP significantly reduced the body weight of the CHD rats, improved myocardial ischemia, and reduced collagen deposition and fibrosis around the infarcted tissue. It reduced thrombosis in a dose-dependent manner and significantly reduced inflammation and oxidative stress damage. Cynaroside, isoquercitrin, rutin, citrulline, and arginine were identified as candidate active TP compounds with antithrombotic effects. The key potential targets of TP in thrombosis treatment were initially identified by molecular docking-based analysis, which showed that the candidate active compounds have a strong binding affinity to the potential targets (protein kinase C alpha type [PKCα], protein kinase C beta type [PKCß], von Willebrand factor [vWF], and prostaglandin-endoperoxide synthase 1 [PTGS1], fibrinogen alpha [Fga], fibrinogen beta [Fgb], fibrinogen gamma [Fgg], coagulation factor II [F2], and coagulation factor VII [F7]). In addition, the candidate active compounds reduced thrombosis, improved oxidative stress damage, and down-regulated the expression of thrombosis-related genes (PKCα, PKCß, vWF, PTGS1, Fga, Fgb, Fgg, F2, and F7) in the zebrafish model. CONCLUSION: Cynaroside, isoquercitrin, rutin, citrulline, and arginine were identified as the active antithrombotic compounds of TP used to treat CHD. Mechanistically, the active compounds were found to be involved in oxidative stress injury, platelet activation pathway, and complement and coagulation cascade pathways.


Subject(s)
Coronary Disease , Fibrinolytic Agents , Molecular Docking Simulation , Network Pharmacology , Trichosanthes , Animals , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use , Fibrinolytic Agents/chemistry , Coronary Disease/drug therapy , Rats , Male , Trichosanthes/chemistry , Zebrafish , Rats, Sprague-Dawley , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Plant Extracts/pharmacology , Plant Extracts/chemistry , Plant Extracts/therapeutic use , Medicine, Chinese Traditional/methods
15.
ACS Appl Mater Interfaces ; 16(17): 21438-21449, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38626407

ABSTRACT

Thrombolytic therapy is one of the most effective treatments for thrombus dissolution and recanalization of blocked vessels in thrombotic diseases. However, the application of the thrombolytic strategy has been limited due to unsatisfactory thrombolytic efficacy, relatively higher bleeding complications, and consequently restricted indications. Recombinant staphylokinase (r-SAK) is a third-generation thrombolytic agent produced by genetic engineering technology, which exhibits a better thrombolytic efficacy than urokinase and recombinant streptokinase. Inspired by the natural affinity of platelets in hemostasis and pathological thrombosis, we developed a platelet membrane (PM)-coated r-SAK (PM-r-SAK). Results from animal experiments and human in vitro studies showed that the PM-r-SAK had a thrombolytic efficacy equal to or better than its 4-fold dose of r-SAK. In a totally occluded rabbit femoral artery thrombosis model, the PM-r-SAK significantly shortened the initial recanalization time compared to the same dose and 4-fold dose of r-SAK. Regarding the recanalized vessels, the PM-r-SAK prolonged the time of reperfusion compared to the same dose and 4-fold dose of r-SAK, though the differences were not significant. An in vitro thrombolytic experiment demonstrated that the thrombolytic efficacy of PM-r-SAK could be inhibited by platelet-poor plasma from patients taking aspirin and ticagrelor. PM coating significantly improves the thrombolytic efficacy of r-SAK, which is related to the thrombus-targeting activity of the PM-r-SAK and can be inhibited by aspirin- and ticagrelor-treated plasma.


Subject(s)
Blood Platelets , Fibrinolytic Agents , Metalloendopeptidases , Thrombosis , Animals , Rabbits , Humans , Thrombosis/drug therapy , Blood Platelets/drug effects , Blood Platelets/metabolism , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/therapeutic use , Fibrinolytic Agents/pharmacology , Metalloendopeptidases/metabolism , Thrombolytic Therapy , Recombinant Proteins/therapeutic use , Male , Cell Membrane/metabolism , Cell Membrane/drug effects
16.
J Med Chem ; 67(9): 7487-7503, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38688020

ABSTRACT

Although bactericidal cationic antimicrobial peptides (AMPs) have been well characterized, less information is available about the antibacterial properties and mechanisms of action of nonbactericidal AMPs, especially nonbactericidal anionic AMPs. Herein, a novel anionic antimicrobial peptide (Gy-CATH) with a net charge of -4 was identified from the skin of the frog Glyphoglossus yunnanensis. Gy-CATH lacks direct antibacterial effects but exhibits significantly preventive and therapeutic capacities in mice that are infected with Staphylococcus aureus, Enterobacteriaceae coli, methicillin-resistant Staphylococcus aureus (MRSA), or carbapenem-resistant E. coli (CREC). In vitro and in vivo investigations proved the regulation of Gy-CATH on neutrophils and macrophages involved in the host immune defense against infection. Moreover, Gy-CATH significantly reduced the extent of pulmonary fibrin deposition and prevented thrombosis in mice, which was attributed to the regulatory role of Gy-CATH in physiological anticoagulants and platelet aggregation. These findings show that Gy-CATH is a potential candidate for the treatment of bacterial infection.


Subject(s)
Anti-Bacterial Agents , Antimicrobial Peptides , Animals , Mice , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/therapeutic use , Anura , Bacterial Infections/drug therapy , Bacterial Infections/prevention & control , Escherichia coli/drug effects , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/therapeutic use , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Immunologic Factors/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Thrombosis/prevention & control , Thrombosis/drug therapy
17.
Langmuir ; 40(18): 9661-9668, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38664943

ABSTRACT

Binding of blood components to collagen was proved to be a key step in thrombus formation. Intelligent Design of Protein Matcher (IDProMat), a neural network model, was then developed based on the principle of seq2seq to design an antithrombotic peptide targeting collagen. The encoding and decoding of peptide sequence data and the interaction patterns of peptide chains at the interface were studied, and then, IDProMat was applied to the design of peptides to cover collagen. The 99.3% decrease in seq2seq loss and 58.3% decrease in MLP loss demonstrated that IDProMat learned the interaction patterns between residues at the binding interface. An efficient peptide, LRWNSYY, was then designed using this model. Validations on its binding on collagen and its inhibition of platelet adhesion were obtained using docking, MD simulations, and experimental approaches.


Subject(s)
Collagen , Peptides , Collagen/chemistry , Peptides/chemistry , Peptides/pharmacology , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/pharmacology , Amino Acid Sequence , Drug Design , Humans , Neural Networks, Computer , Molecular Dynamics Simulation , Platelet Adhesiveness/drug effects
18.
Proc Natl Acad Sci U S A ; 121(11): e2314349121, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38442174

ABSTRACT

Thrombosis, induced by abnormal coagulation or fibrinolytic systems, is the most common pathology associated with many life-threatening cardio-cerebrovascular diseases. However, first-line anticoagulant drugs suffer from rapid drug elimination and risk of hemorrhagic complications. Here, we developed an in situ formed depot of elastin-like polypeptide (ELP)-hirudin fusion protein with a prodrug-like feature for long-term antithrombotic therapy. Highly secretory expression of the fusion protein was achieved with the assistance of the Ffu312 tag. Integration of hirudin, ELP, and responsive moiety can customize fusion proteins with properties of adjustable in vivo retention and controllable recovery of drug bioactivity. After subcutaneous injection, the fusion protein can form a reservoir through temperature-induced coacervation of ELP and slowly diffuse into the blood circulation. The biological activity of hirudin is shielded due to the N-terminal modification, while the activated key proteases upon thrombus occurrence trigger the cleavage of fusion protein together with the release of hirudin, which has antithrombotic activity to counteract thrombosis. We substantiated that the optimized fusion protein produced long-term antithrombotic effects without the risk of bleeding in multiple animal thrombosis models.


Subject(s)
Elastin-Like Polypeptides , Thrombosis , Animals , Fibrinolytic Agents/pharmacology , Hirudins/genetics , Hirudins/pharmacology , Anticoagulants , Thrombosis/drug therapy , Thrombosis/prevention & control
19.
Carbohydr Polym ; 334: 122058, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38553243

ABSTRACT

Global public health is seriously threatened by thrombotic disorders because of their high rates of mortality and disability. Most thrombolytic agents, especially protein-based pharmaceuticals, have a short half-life in circulation, reducing their effectiveness in thrombolysis. The creation of an intelligent drug delivery system that delivers medication precisely and releases it under regulated conditions at nearby thrombus sites is essential for effective thrombolysis. In this article, we present a unique medication delivery system (MCRUA) that selectively targets platelets and releases drugs by stimulation from the thrombus' microenvironment. The thrombolytic enzyme urokinase-type plasminogen-activator (uPA) and the anti-inflammatory medication Aspirin (acetylsalicylic acid, ASA) are both loaded onto pH-sensitive CaCO3/cyclodextrin crosslinking metal-organic frameworks (MC) that make up the MCRUA system. c(RGD) is functionalized on the surface of MC, which is functionalized by RGD to an esterification reaction. Additionally, the thrombus site's acidic microenvironment causes MCRUA to disintegrate to release uPA for thrombolysis and aiding in vessel recanalization. Moreover, cyclodextrin-encapsulated ASA enables the treatment of the inflammatory environment within the thrombus, enhancing the antiplatelet aggregation effects and promoting cooperative thrombolysis therapy. When used for thrombotic disorders, our drug delivery system (MCRUA) promotes thrombolysis, suppresses rethrombosis, and enhances biosafety with fewer hemorrhagic side effects.


Subject(s)
Cyclodextrins , Metal-Organic Frameworks , Thrombosis , Humans , Thrombolytic Therapy , Cyclodextrins/therapeutic use , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use , Urokinase-Type Plasminogen Activator/therapeutic use , Thrombosis/drug therapy , Aspirin/pharmacology , Oligopeptides
20.
Nanoscale ; 16(12): 6132-6141, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38444355

ABSTRACT

Thrombosis is the major cause of cardiovascular diseases. Only a small subset of patients could benefit from thrombolytic therapy due to the high bleeding risk brought about by the repeated administration of thrombolytic drugs. Nanoparticles with targeting ligands have been developed as nanocarriers of thrombolytic drugs to deliver the drug to the thrombus through active targeting. However, the passive targeting effect of nanoparticles on the thrombus is yet to be investigated. Herein, we prepared silica cross-linked micelles (SCLMs) with a long blood circulation half-life as drug carriers to target the thrombus through passive targeting. Compared with SCLMs modified with an active targeting ligand cRGD, the SCLMs exhibited similar targeting behavior to the thrombus in vivo. Loaded with the thrombolytic drug tirofiban, the passive targeting SCLMs showed a comparable therapeutic effect to cRGD-modified SCLMs in a mice model with pulmonary embolism and arterial thrombosis.


Subject(s)
Nanoparticles , Thrombosis , Mice , Animals , Humans , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use , Drug Carriers/therapeutic use , Thrombosis/drug therapy , Drug Delivery Systems , Micelles
SELECTION OF CITATIONS
SEARCH DETAIL
...