Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.337
Filter
1.
Sci Rep ; 14(1): 21175, 2024 09 11.
Article in English | MEDLINE | ID: mdl-39256523

ABSTRACT

Various seeds, including sea buckthorn (Hippophae rhamnoides L.) seeds, are sources of different bioactive compounds. They can show anti-inflammatory, hypoglycemic, anti-hyperlipidemic, antibacterial, antioxidant, or other biological properties in in vitro and in vivo models. Our preliminary in vitro results have demonstrated that the extracts from raw (no thermal processing) and roasted (thermally processed) sea buckthorn seeds have antioxidant potential and anticoagulant activity. However, it was unclear which compounds were responsible for these properties. Therefore, in continuation of our previous study, the extracts were fractionated by C18 chromatography. Phytochemical analysis of three fractions (a, b, and c) from raw sea buckthorn seeds and four fractions (d, e, f, and g) from roasted sea buckthorn seeds were performed. Several in vitro assays were also conducted to determine the antioxidant and procoagulant/anticoagulant potential of the fractions and two of their major constituents-isorhamnetin 3-O-ß-glucoside7-O-α-rhamnoside and serotonin. LC-MS analyses showed that serotonin is the dominant constituent of fractions c and f, which was tentatively identified on the basis of its HRMS and UV spectra. Moreover, fractions c and f, as well as b and e, contained different B-type proanthocyanidins. Fractions b and e consisted mainly of numerous glycosides of kaempferol, quercetin, and isorhamnetin. The results of oxidative stress assays (measurements of protein carbonylation, lipid peroxidation, and thiol groups oxidation) showed that out of all the tested fractions, fraction g (isolated from roasted seeds and containing mainly dihexoses, and serotonin) demonstrated the strongest antioxidant properties.


Subject(s)
Antioxidants , Hippophae , Plant Extracts , Seeds , Antioxidants/pharmacology , Antioxidants/chemistry , Seeds/chemistry , Hippophae/chemistry , Plant Extracts/pharmacology , Plant Extracts/chemistry , Phytochemicals/pharmacology , Phytochemicals/chemistry , Serotonin/metabolism , Hemostatics/pharmacology , Hemostatics/isolation & purification , Humans , Anticoagulants/pharmacology , Anticoagulants/chemistry , Animals
2.
Sci Rep ; 14(1): 22335, 2024 09 27.
Article in English | MEDLINE | ID: mdl-39333740

ABSTRACT

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.


Subject(s)
Anticoagulants , Antithrombins , Blood Coagulation , Heparin , Heparin/pharmacology , Heparin/chemistry , Antithrombins/pharmacology , Antithrombins/chemistry , Anticoagulants/pharmacology , Anticoagulants/chemistry , Humans , Blood Coagulation/drug effects , Fibrin/metabolism , Factor Xa/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism
3.
Int J Biol Macromol ; 279(Pt 3): 135255, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39236965

ABSTRACT

A polysaccharide, CZS-0-1, was obtained from the marine green algae Codium fragile using ion-exchange and size-exclusion chromatography. Composition and characteristics analyses showed CZS-0-1 was a sulfated galactoarabinan consisting of arabinose, galactose and a small amount of glucose in a ratio of 9:2:1 with 21% sulfate content and a molecular weight of 810 kDa. Structural properties were determined using desulfation and methylation analyses combined with instrument analysis. The results showed that the backbone of CZS-0-1 was (1 â†’ 3)-ß-L-Arap. Its O-4 and/or O-2 positions showed sulfate modification; additionally, it had 10% of (1 â†’ 3)-ß-D-Galp branches at the O-4 position of the (1 â†’ 3)-ß-L-Arap. The galactose side chains also had sulfate modification at the O-4 or O-6 position. The structure of CZS-0-1 was further confirmed by Top-down analysis of the oligosaccharides after oxidated hydrolysis by mass spectrometry. CZS-0-1 exhibited significant heparin-like anticoagulant activity. It exerted anticoagulant effects by inhibiting FIIa and FXa activities with the presence of heparin cofactors. The anticoagulant activity of CSZ-0-1 was closely related to the molecular weight, and the reduction of molecular weight may lead to a significant decrease in the anticoagulant activity. This study demonstrated that the green algae, Codium fragile can be considered as a useful resource for bioactive polysaccharides.


Subject(s)
Anticoagulants , Chlorophyta , Oligosaccharides , Chlorophyta/chemistry , Anticoagulants/pharmacology , Anticoagulants/chemistry , Anticoagulants/isolation & purification , Oligosaccharides/chemistry , Oligosaccharides/pharmacology , Oligosaccharides/isolation & purification , Polysaccharides/chemistry , Polysaccharides/pharmacology , Polysaccharides/isolation & purification , Molecular Weight , Sulfates/chemistry , Galactose/chemistry , Methylation
4.
BMC Pharmacol Toxicol ; 25(1): 56, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39175081

ABSTRACT

BACKGROUND: Direct oral anticoagulants (DOACs) have high potency against their therapeutic target and are widely used in the treatment of atrial fibrillation (AF). Most DOACs are often claimed to have adverse effects due to off-target inhibition of essential proteins. Human serum paraoxonase 1 (PON1), one of the essential proteins, known for its anti-inflammatory and antioxidant properties, could be affected by DOACs. Thus, a comparative evaluation of DOACs and their effect on PON1 protein will aid in recommending the most effective DOACs for AF treatment. This study aimed to assess the impact of DOACs on PON1 through a combination of computational and experimental analyses. METHODS: We focus on apixaban, dabigatran, and rivaroxaban, the most recommended DOACs in AF treatment, for their impact on PON1 through molecular docking and molecular dynamics (MD) simulation to elucidate the binding affinity and drug-protein structural stability. This investigation revealed the most influential DOACs on the PON1 protein. Then experimental validation was performed in DOAC-treated AF participants (n = 42; 19 treated with dabigatran and 23 treated with rivaroxaban) compared to a healthy control group (n = 22) through gene expression analysis in peripheral blood mononuclear cells (PBMC) and serum enzyme concentration. RESULTS: Our computational investigation showed rivaroxaban (-4.24 kcal/mol) exhibited a lower affinity against the PON1 protein compared to apixaban (-5.97 kcal/mol) and dabigatran (-9.03 kcal/mol) through molecular docking. Dabigatran holds complex interactions with PON1 at GLU53, TYR197, SER193, and ASP269 by forming hydrogen bonds. Additionally, MD simulation revealed that dabigatran disrupts PON1 stability, which may contribute functional changes. Further experimental validation revealed a significant down-regulation (p < 0.05) of PON1 gene expression in PBMC and decreased serum PON1 enzyme concentration on DOAC treatment. Rivaroxaban as about 48% has inhibitory percentage and dabigatran as about 75% of inhibitory percentage compared to healthy control. CONCLUSION: Overall, our computational and experimental results clearly show the higher inhibitory effect of dabigatran than rivaroxaban. Hence, rivaroxaban will be a better drug candidate for improving the outcome of AF.


Subject(s)
Aryldialkylphosphatase , Atrial Fibrillation , Dabigatran , Molecular Docking Simulation , Molecular Dynamics Simulation , Pyridones , Rivaroxaban , Humans , Atrial Fibrillation/drug therapy , Atrial Fibrillation/metabolism , Aryldialkylphosphatase/blood , Rivaroxaban/therapeutic use , Male , Pyridones/therapeutic use , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrazoles/chemistry , Administration, Oral , Anticoagulants/pharmacology , Anticoagulants/chemistry , Female , Aged , Middle Aged
5.
Nat Commun ; 15(1): 6769, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117649

ABSTRACT

Low-molecular-weight heparin (LMWH), derived from unfractionated heparin (UFH), has enhanced anticoagulant efficacy, long duration of action, and extended half-life. Patients receiving LMWH for preventive therapies would strongly benefit from its long-term effects, however, achieving this is challenging. Here, we design and evaluate a nanoengineered LMWH and octadecylamine conjugate (LMHO) that can act for a long time while maintaining close to 97 ± 3% of LMWH activity via end-specific conjugation of the reducing end of LMWH. LMHO can self-assemble into nanoparticles with an average size of 105 ± 1.7 nm in water without any nanocarrier and can be combined with serum albumin, resulting in a lipid-based albumin shuttling effect. Such molecules can circulate in the bloodstream for 4-5 days. We corroborate the self-assembly capability of LMHO and its interaction with albumin through molecular dynamics (MD) simulations and transmission electron microscopy (TEM) analysis. This innovative approach to carrier-free polysaccharide delivery, enhanced by nanoengineered albumin shuttling, represents a promising platform to address limitations in conventional therapies.


Subject(s)
Amines , Anticoagulants , Heparin, Low-Molecular-Weight , Molecular Dynamics Simulation , Nanoparticles , Heparin, Low-Molecular-Weight/chemistry , Amines/chemistry , Humans , Nanoparticles/chemistry , Anticoagulants/chemistry , Anticoagulants/pharmacology , Animals , Serum Albumin/chemistry , Serum Albumin/metabolism , Drug Carriers/chemistry
6.
Int J Pharm ; 664: 124627, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39179008

ABSTRACT

3D-printed dosage forms comprised of Carbopol and Eudragit were fabricated through semi-solid extrusion, combining Enoxaparin (Enox) and the permeation enhancer SNAC in a single-step process without subsequent post-processing. Inks were characterized using rheology and Fourier-transform infrared (FTIR) spectroscopy. The stability of Enox in the fabricated dosage forms was assessed by means of Nuclear Magnetic Resonance (NMR) and Circular Dichroism (CD) analysis. In vitro release studies revealed the release of Enox in a sustained manner, whereas ex vivo experiments demonstrated the mucoadhesive properties of the 3D-printed dosage forms and their ability to enhance Enox permeability across intestinal mucosa. Cellular assays (CCK-8 assay) revealed a dose- and time-dependent response following incubation with the 3D-printed dosage forms. The encapsulation of SNAC in the 3D-printed dosage forms demonstrated their capacity to increase the transcellularly transport of macromolecule across Caco-2 monolayer in a reversible manner, as confirmed by Transepithelial Resistance (TEER) measurements.


Subject(s)
Drug Liberation , Enoxaparin , Printing, Three-Dimensional , Tablets , Caco-2 Cells , Humans , Administration, Oral , Enoxaparin/administration & dosage , Enoxaparin/pharmacokinetics , Enoxaparin/chemistry , Acrylic Resins/chemistry , Animals , Polymethacrylic Acids/chemistry , Intestinal Mucosa/metabolism , Male , Drug Delivery Systems/methods , Adhesiveness , Permeability , Polyvinyls/chemistry , Anticoagulants/administration & dosage , Anticoagulants/pharmacokinetics , Anticoagulants/chemistry
7.
Protein J ; 43(4): 888-909, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39095592

ABSTRACT

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.


Subject(s)
Blood Platelets , Factor Xa Inhibitors , Thrombin , Animals , Humans , Mice , Amino Acid Sequence , Anticoagulants/pharmacology , Anticoagulants/chemistry , Blood Platelets/drug effects , Blood Platelets/metabolism , Factor Xa/chemistry , Factor Xa/metabolism , Factor Xa Inhibitors/pharmacology , Factor Xa Inhibitors/chemistry , Molecular Docking Simulation , Thrombin/chemistry , Thrombin/metabolism , Male
8.
Int J Biol Macromol ; 277(Pt 3): 134490, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39111494

ABSTRACT

Fucoidan is a sulfated polysaccharide that occurs naturally in the cell wall of brown seaweeds and has substantial biological efficacy. Optimizing the extraction of fucoidan from different brown seaweeds was the primary goal of this research. The optimization of fucoidan extraction was applied on the brown macroalga Turbinaria turbinata using a Box-Behnken Design (BBD) to inspect the impacts of different pH (3, 5, 7), temperature (70, 80, 90 °C) and extraction duration (60, 120, 180 min) on both the yield and sulfate content of fucoidan. The optimized parameters recorded to maximize the fucoidan yield and its sulfate content were a pH of 3.44 and a temperature of 82.26 °C for 60 min. The optimal conditions obtained from BBD were used for fucoidan extraction from T. turbinata, Sargassum cinereum, Padina pavonica, and Dictyota dichotoma. The highest average of fucoidan yield was derived from P. pavonica (40.76 ± 4.04 % DW). FTIR, 1H NMR, and HPLC were used to characterize extracted fucoidan. The extracted fucoidan's Physical characteristics, biochemical composition, antioxidant potential, antitumor effect against breast cancer cells (MCF-7), and antimicrobial and anticoagulant activity were assessed. The extracted fucoidan from D. dichotoma, followed by that extracted from S. cinereum, which had the highest sulphate content, depicted the highest antioxidant, anticancer, and anticoagulant activities. Fucoidan has demonstrated a strong antimicrobial action against some pathogenic microorganisms; Bacillus subtilis, Staphylococcus aureus, Escherichia coli, Klebsiella pneumonia, and Candida albicans. The anticoagulant properties of fucoidan from D. dichotoma were stronger than those of fucoidan from S. cinereum, T. turbinata, and P. pavonica due to its higher sulphate content. These findings could be used for various biomedical applications to improve the pharmaceutical industry.


Subject(s)
Polysaccharides , Polysaccharides/pharmacology , Polysaccharides/chemistry , Polysaccharides/isolation & purification , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Antioxidants/pharmacology , Antioxidants/chemistry , Antioxidants/isolation & purification , MCF-7 Cells , Hydrogen-Ion Concentration , Temperature , Seaweed/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Phaeophyceae/chemistry , Microbial Sensitivity Tests , Anticoagulants/pharmacology , Anticoagulants/chemistry , Anticoagulants/isolation & purification
9.
Carbohydr Polym ; 343: 122469, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39174090

ABSTRACT

Examining the critical role of anticoagulants in medical practice, particularly their central function in preventing abnormal blood clotting, is of the utmost importance. However, the study of interactions between blood proteins and alternative anticoagulant nano-surfaces is still understood poorly. In this study, novel approach involving direct functionalisation of magnetic iron oxide nanoparticles (MNPs) as carriers with sulphated dextran (s-dext) is presented, with the aim of evaluating the potential of magnetically-responsive MNPs@s-dext as anticoagulants. The physicochemical characterisation of the synthesised MNPs@s-dext includes crystal structure analysis, morphology study, surface and electrokinetic properties, thermogravimetric analysis and magnetic properties` evaluation, which confirms the successful preparation of the nanocomposite with sulfonate groups. The anticoagulant potential of MNPs@s-dext was investigated using a standardised activated partial thromboplastin time (APTT) test and a modified APTT test with a quartz crystal microbalance with dissipation (QCM-D) which confirmed the anticoagulant effect. Time-resolved solid-liquid interactions between the MNPs@s-dext and model blood proteins bovine serum albumin and fibrinogen were also investigated, to gain insight into their hemocompatibility, and revealed protein-repellence of MNPs@s-dext against blood proteins. The study also addressed comprehensive cytotoxicity studies of prepared nanocomposites, and provided valuable insights into potential applicability of MNPs@s-dext as a promising magnetic anticoagulant in biomedical contexts.


Subject(s)
Anticoagulants , Dextran Sulfate , Nanocomposites , Anticoagulants/pharmacology , Anticoagulants/chemistry , Humans , Nanocomposites/chemistry , Nanocomposites/toxicity , Dextran Sulfate/chemistry , Serum Albumin, Bovine/chemistry , Blood Coagulation/drug effects , Magnetic Iron Oxide Nanoparticles/chemistry , Magnetic Iron Oxide Nanoparticles/toxicity , Animals , Ferric Compounds/chemistry , Ferric Compounds/pharmacology , Fibrinogen/chemistry , Cell Survival/drug effects , Partial Thromboplastin Time , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/toxicity
10.
Toxins (Basel) ; 16(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39057940

ABSTRACT

Envenoming resulting from snakebites is recognized as a priority neglected tropical disease by The World Health Organization. The Bothrops genus, consisting of different pitviper species, is considered the most medically significant taxa in Central and South America. Further research into Bothrops venom composition is important to aid in the development of safer and more effective snakebite treatments. In addition, the discovery of Bothrops toxins that could potentially be used for medical or diagnostic purposes is of interest to the pharmaceutical industry. This study aimed to employ high-throughput (HT) venomics to qualitatively analyze venom composition while utilizing coagulation bioassays for identifying coagulopathic toxins and characterizing coagulopathic activity in various Bothrops venoms. Using the recently demonstrated HT venomics workflow in combination with post-column coagulopathic bioassaying, focus was placed at anticoagulant toxins. Well-known procoagulant toxins were also investigated, taking into account that using the HT venomics workflow, procoagulant toxins are especially prone to denaturation during the reversed-phase chromatographic separations performed in the workflow. The findings revealed that the venoms of B. atrox and B. jararaca harbored procoagulant toxins, whereas those of B. alternatus and B. neuwiedi contained both procoagulant and anticoagulant toxins. In general, anticoagulation was associated with phospholipases A2s, while procoagulation was associated with snake venom metalloproteinases and snake venom serine proteases. These results showed the identification of coagulopathic venom toxins in the Bothrops venoms analyzed using multiple analytical methods that complement each other. Additionally, each venom underwent qualitative characterization of its composition.


Subject(s)
Blood Coagulation , Bothrops , Crotalid Venoms , High-Throughput Screening Assays , Animals , Crotalid Venoms/chemistry , Blood Coagulation/drug effects , Biological Assay , Anticoagulants/pharmacology , Anticoagulants/chemistry , Anticoagulants/analysis , Humans
11.
Luminescence ; 39(7): e4824, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39004773

ABSTRACT

Dabigatran (DBG), marketed as Pradaxa, is an anticoagulant medication prescribed for the treatment and mitigation of blood clots and to lower the risk of stroke in individuals with the heart condition known as atrial fibrillation. This medication is specifically indicated for preventing blood clots post hip or knee replacement surgeries and in patients with a prior history of clots. Compared to warfarin, dabigatran serves as a viable alternative that does not necessitate routine blood monitoring tests. The complimentary benefits associated with SALL (salting-out assisted liquid-liquid extraction) and the fluorogenic capabilities of benzofurazan. These methods were combined to provide an affordable and sensitive DBG assaying method. The spectral strength of the yellow luminous product was examined at 533.8 nm and by adjustment of a wavelength of 474.7 nm for excitation. To assess its linearity, the calibration chart was tested across a DBG concentration range of 30-500 ng/ml. Via accurate computation based on ICH, the detection limit (LD) was determined to be 9.5 ng/ml, and the strategy can quantify the DBG to a limit of 28 ng/ml. To ensure success, various crucial parameters for method implementation have been extensively studied and adapted. The validation of the strategy adhered to the policies outlined by ICH, affirming its precision in quantifying DBG in capsules. Furthermore, the inclusion of SALLE steps facilitated accurate monitoring of DBG in plasma samples, introducing a unique and advanced methodology for analyzing this compound in biological samples.


Subject(s)
Anticoagulants , Capsules , Dabigatran , Dabigatran/blood , Dabigatran/chemistry , Dabigatran/pharmacology , Humans , Anticoagulants/chemistry , Anticoagulants/blood , Anticoagulants/pharmacology , Fluorescent Dyes/chemistry , Liquid-Liquid Extraction , Spectrometry, Fluorescence , Limit of Detection , 4-Chloro-7-nitrobenzofurazan
12.
Int J Biol Macromol ; 275(Pt 2): 133743, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38986975

ABSTRACT

Due to wonderful taste, rich nutrition and biological functions, many marine green algae in the genus Caulerpa have been recently developed as candidates for green caviar. A novel water-soluble sulfated xylogalactomannan CO-0-1 was obtained from the green algae Caulerpa okamurae. CO-0-1 was mainly composed of mannose (Man), galactose (Gal), and xylose (Xyl) at the ratio of 4.4:4.0:1.4 with the molecular weight at 470 kDa and the sulfate content at 12.78 %. The sulfated xylogalactomannan had Man at the backbone with →4)-ß-D-Manp-(1→ and →2)-ß-D-Manp-(1→ as the main chain and branches at O-3 position. The side chains contained →3)-ß-D-Galp-(1→ and minor →2)-ß-D-Xylp(1→. The sulfate groups only distributed at the side chains and at O-6 position of →3)-ß-D-Galp-(1→ and O-4 position of (1→2)-ß-D-Xylp. The anticoagulant activity indicated that CO-0-1 displayed intrinsic anticoagulant and specific anti-thrombin activities. The investigation expanded the utilization and development scene and scope of the green algae Caulerpa okamurae.


Subject(s)
Anticoagulants , Caulerpa , Mannans , Anticoagulants/chemistry , Anticoagulants/pharmacology , Anticoagulants/isolation & purification , Caulerpa/chemistry , Mannans/chemistry , Mannans/pharmacology , Mannans/isolation & purification , Molecular Weight , Sulfates/chemistry , Humans
13.
Proc Natl Acad Sci U S A ; 121(29): e2401136121, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38985762

ABSTRACT

Hemostasis relies on a reaction network of serine proteases and their cofactors to form a blood clot. Coagulation factor IXa (protease) plays an essential role in hemostasis as evident from the bleeding disease associated with its absence. RNA aptamers specifically targeting individual coagulation factors have potential as anticoagulants and as probes of the relationship between structure and function. Here, we report X-ray structures of human factor IXa without a ligand bound to the active site either in the apo-form or in complex with an inhibitory aptamer specific for factor IXa. The aptamer binds to an exosite in the catalytic domain and allosterically distorts the active site. Our studies reveal a conformational ensemble of IXa states, wherein large movements of Trp215 near the active site drive functional transitions between the closed (aptamer-bound), latent (apo), and open (substrate-bound) states. The latent state of the apo-enzyme may bear on the uniquely poor catalytic activity of IXa compared to other coagulation proteases. The exosite, to which the aptamer binds, has been implicated in binding VIIIa and heparin, both of which regulate IXa function. Our findings reveal the importance of exosite-driven allosteric modulation of IXa function and new strategies to rebalance hemostasis for therapeutic gain.


Subject(s)
Aptamers, Nucleotide , Factor IXa , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/metabolism , Factor IXa/metabolism , Factor IXa/chemistry , Factor IXa/antagonists & inhibitors , Humans , Allosteric Regulation , Catalytic Domain , Crystallography, X-Ray , Models, Molecular , Protein Binding , Anticoagulants/chemistry , Anticoagulants/metabolism , Anticoagulants/pharmacology
14.
Int J Biol Macromol ; 275(Pt 1): 133631, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38964688

ABSTRACT

Controllable heparin-release is of great importance and necessity for the precise anticoagulant regulation. Efforts have been made on designing heparin-releasing systems, while, it remains a great challenge for gaining the external-stimuli responsive heparin-release in either intravenous or catheter delivery. In this study, an azobenzene-containing ammonium surfactant is designed and synthesized for the fabrication of photoresponsive heparin ionic complexes through the electrostatic complexation with heparin. Under the assistance of photoinduced trans-cis isomerization of azobenzene, the obtained heparin materials perform reversible athermal phase transition between ordered crystalline and isotropic liquid state at room temperature. Compared to the ordered state, the formation of isotropic state can effectively improve the dissolving of heparin from ionic materials in aqueous condition, which realizes the photo-modulation on the concentration of free heparin molecules. With good biocompatibility, such a heparin-releasing system addresses photoresponsive anticoagulation in both in vitro and in vivo biological studies, confirming its great potential clinical values. This work provides a new designing strategy for gaining anticoagulant regulation by light, also opening new opportunities for the development of photoresponsive drugs and biomedical materials based on biomolecules.


Subject(s)
Anticoagulants , Drug Delivery Systems , Heparin , Photosensitizing Agents , Heparin/chemistry , Heparin/pharmacology , Photosensitizing Agents/chemistry , Anticoagulants/chemistry , Anticoagulants/pharmacology , Ions/chemistry , Animals , Mice , Cell Line , Female , Mice, Inbred BALB C , Drug Delivery Systems/methods , Cell Survival/drug effects , Azo Compounds/chemistry , Surface-Active Agents/chemistry
15.
Mar Drugs ; 22(6)2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38921576

ABSTRACT

Three polysaccharides (SnNG, SnFS and SnFG) were purified from the body wall of Stichopus naso. The physicochemical properties, including monosaccharide composition, molecular weight, sulfate content, and optical rotation, were analyzed, confirming that SnFS and SnFG are sulfated polysaccharides commonly found in sea cucumbers. The highly regular structure {3)-L-Fuc2S-(α1,}n of SnFS was determined via a detailed NMR analysis of its oxidative degradation product. By employing ß-elimination depolymerization of SnFG, tri-, penta-, octa-, hendeca-, tetradeca-, and heptadeca-saccharides were obtained from the low-molecular-weight product. Their well-defined structures confirmed that SnFG possessed the backbone of {D-GalNAc4S6S-ß(1,4)-D-GlcA}, and each GlcA residue was branched with Fuc2S4S. SnFS and SnFG are both structurally the simplest version of natural fucan sulfate and fucosylated glycosaminoglycan, facilitating the application of low-value sea cucumbers S. naso. Bioactivity assays showed that SnFG and its derived oligosaccharides exhibited potent anticoagulation and intrinsic factor Xase (iXase) inhibition. Moreover, a comparative analysis with the series of oligosaccharides solely branched with Fuc3S4S showed that in oligosaccharides with lower degrees of polymerization, such as octasaccharides, Fuc2S4S led to a greater increase in APTT prolongation and iXase inhibition. As the degree of polymerization increases, the influence from the sulfation pattern diminishes, until it is overshadowed by the effects of molecular weight.


Subject(s)
Anticoagulants , Molecular Weight , Oligosaccharides , Polysaccharides , Animals , Anticoagulants/pharmacology , Anticoagulants/chemistry , Anticoagulants/isolation & purification , Polysaccharides/pharmacology , Polysaccharides/chemistry , Polysaccharides/isolation & purification , Oligosaccharides/pharmacology , Oligosaccharides/chemistry , Oligosaccharides/isolation & purification , Stichopus/chemistry , Sea Cucumbers/chemistry , Sulfates/chemistry , Magnetic Resonance Spectroscopy , Blood Coagulation/drug effects
16.
Int J Mol Sci ; 25(12)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38928208

ABSTRACT

Unfractionated heparin (UFH) and its low-molecular-weight fragments (LMWH) are widely used as anticoagulants for surgical procedures and extracorporeal blood purification therapies such as cardiovascular surgery and dialysis. The anticoagulant effect of heparin is essential for the optimal execution of extracorporeal blood circulation. However, at the end of these procedures, to avoid the risk of bleeding, it is necessary to neutralize it. Currently, the only antidote for heparin neutralization is protamine sulphate, a highly basic protein which constitutes a further source of serious side events and is ineffective in neutralizing LMWH. Furthermore, dialysis patients, due to the routine administration of heparin, often experience serious adverse effects, among which HIT (heparin-induced thrombocytopenia) is one of the most severe. For this reason, the finding of new heparin antagonists or alternative methods for heparin removal from blood is of great interest. Here, we describe the synthesis and characterization of a set of biocompatible macroporous cryogels based on poly(2-hydroxyethyl methacrylate) (pHEMA) and L-lysine with strong filtering capability and remarkable neutralization performance with regard to UFH and LMWH. These properties could enable the design and creation of a filtering device to rapidly reverse heparin, protecting patients from the harmful consequences of the anticoagulant.


Subject(s)
Anticoagulants , Cryogels , Heparin , Lysine , Heparin/chemistry , Heparin/adverse effects , Humans , Cryogels/chemistry , Anticoagulants/chemistry , Lysine/chemistry , Heparin, Low-Molecular-Weight/chemistry , Heparin Antagonists/chemistry
17.
Carbohydr Polym ; 339: 122216, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38823901

ABSTRACT

Low Molecular Weight Heparins (LMWHs) are well-established for use in the prevention and treatment of thrombotic diseases, and as a substitute for unfractionated heparin (UFH) due to their predictable pharmacokinetics and subcutaneous bioavailability. LMWHs are produced by various depolymerization methods from UFH, resulting in heterogeneous compounds with similar biochemical and pharmacological properties. However, the delicate supply chain of UFH and potential contamination from animal sources require new manufacturing approaches for LMWHs. Various LMWH preparation methods are emerging, such as chemical synthesis, enzymatic or chemical depolymerization and chemoenzymatic synthesis. To establish the sameness of active ingredients in both innovator and generic LMWH products, the Food and Drug Administration has implemented a stringent scientific method of equivalence based on physicochemical properties, heparin source material and depolymerization techniques, disaccharide composition and oligosaccharide mapping, biological and biochemical properties, and in vivo pharmacodynamic profiles. In this review, we discuss currently available LMWHs, potential manufacturing methods, and recent progress for manufacturing quality control of these LMWHs.


Subject(s)
Heparin, Low-Molecular-Weight , Quality Control , Heparin, Low-Molecular-Weight/chemistry , Humans , Animals , Anticoagulants/chemistry , Anticoagulants/pharmacology
18.
J Med Chem ; 67(13): 10946-10966, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38913497

ABSTRACT

Thrombo-inflammation is closely associated with a few severe cardiovascular and infectious diseases. Factor XIIa (FXIIa) in the intrinsic coagulation pathway plays a pivotal role in the development of thrombo-inflammation and its inhibition has emerged as a potential therapeutic approach for thrombo-inflammatory disorders. Nonetheless, as of now, few small-molecule FXIIa inhibitors have demonstrated notable effectiveness against thrombo-inflammation, with none progressing into clinical stages. Herein, we present potent, covalent, reversible, and selective small-molecule FXIIa inhibitors such as 4a and 4j obtained through structure-based drug design. Compounds 4a and 4j showed significant anticoagulation and substantial anti-inflammatory effects in vitro, coupled with exceptional plasma stability. Furthermore, in carrageenan-induced thrombosis models, 4a and 4j demonstrated remarkable dual antithrombotic and anti-inflammatory activity when administered orally. Compound 4j exhibited a favorable safety profile without obvious tissue toxicity in mice, suggesting its potential as an oral therapeutic option for thrombo-inflammation.


Subject(s)
Factor XIIa , Thrombosis , Animals , Thrombosis/drug therapy , Mice , Humans , Factor XIIa/antagonists & inhibitors , Factor XIIa/metabolism , Administration, Oral , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacokinetics , Structure-Activity Relationship , Carrageenan , Drug Discovery , Inflammation/drug therapy , Male , Anticoagulants/pharmacology , Anticoagulants/therapeutic use , Anticoagulants/chemistry , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use , Fibrinolytic Agents/chemistry , Biological Availability
19.
Carbohydr Polym ; 339: 122220, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38823902

ABSTRACT

Natural heparin, a glycosaminoglycan consisting of repeating hexuronic acid and glucosamine linked by 1 â†’ 4 glycosidic bonds, is the most widely used anticoagulant. To subvert the dependence on animal sourced heparin, alternative methods to produce heparin saccharides, i.e., either heterogenous sugar chains similar to natural heparin, or structurally defined oligosaccharides, are becoming hot subjects. Although the success by chemical synthesis of the pentasaccharide, fondaparinux, encourages to proceed through a chemical approach generating homogenous product, synthesizing larger oligos is still cumbersome and beyond reach so far. Alternatively, the chemoenzymatic pathway exhibited exquisite stereoselectivity of glycosylation and regioselectivity of modification, with the advantage to skip the tedious protection steps unavoidable in chemical synthesis. However, to a scale of drug production needed today is still not in sight. In comparison, a procedure of de novo biosynthesis in an organism could be an ultimate goal. The main purpose of this review is to summarize the current available/developing strategies and techniques, which is expected to provide a comprehensive picture for production of heparin saccharides to replenish or eventually to replace the animal derived products. In chemical and chemoenzymatic approaches, the methodologies are discussed according to the synthesis procedures: building block preparation, chain elongation, and backbone modification.


Subject(s)
Anticoagulants , Heparin , Animals , Anticoagulants/chemical synthesis , Anticoagulants/chemistry , Glycosylation , Heparin/chemistry , Heparin/chemical synthesis , Oligosaccharides/chemical synthesis , Oligosaccharides/chemistry
20.
Carbohydr Polym ; 341: 122297, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38876729

ABSTRACT

The biological activities of heparan sulfate (HS) are intimately related to their molecular weights, degree and pattern of sulfation and homogeneity. The existing methods for synthesizing homogeneous sugar chains of low dispersity involve multiple steps and require stepwise isolation and purification processes. Here, we designed a mesoporous metal-organic capsule for the encapsulation of glycosyltransferase and obtained a microreactor capable of enzymatically catalyzing polymerization reactions to prepare homogeneous heparosan of low dispersity, the precursor of HS and heparin. Since the sugar chain extension occurs in the pores of the microreactor, low molecular weight heparosan can be synthesized through space-restricted catalysis. Moreover, the glycosylation co-product, uridine diphosphate (UDP), can be chelated with the exposed metal sites of the metal-organic capsule, which inhibits trans-cleavage to reduce the molecular weight dispersity. This microreactor offers the advantages of efficiency, reusability, and obviates the need for stepwise isolation and purification processes. Using the synthesized heparosan, we further successfully prepared homogeneous 6-O-sulfated HS of low dispersity with a molecular weight of approximately 6 kDa and a polydispersity index (PDI) of 1.032. Notably, the HS generated exhibited minimal anticoagulant activity, and its binding affinity to fibroblast growth factor 1 was comparable to that of low molecular weight heparins.


Subject(s)
Heparitin Sulfate , Polymerization , Heparitin Sulfate/chemistry , Anticoagulants/chemistry , Anticoagulants/pharmacology , Anticoagulants/chemical synthesis , Molecular Weight , Porosity , Humans , Disaccharides/chemistry , Glycosyltransferases/metabolism , Glycosyltransferases/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL