Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.712
Filter
1.
Int J Mol Sci ; 25(17)2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39273200

ABSTRACT

Thrombosis is a key process that determines acute coronary syndrome and ischemic stroke and is the leading cause of morbidity and mortality in the world, together with cancer. Platelet adhesion and subsequent activation and aggregation are critical processes that cause thrombus formation after endothelial damage. To date, high hopes are associated with compounds of natural origin, which show anticoagulant action without undesirable effects and can be proposed as supportive therapies. We investigated the effect of the new combination of four natural compounds, escin-bromelain-ginkgo biloba-sage miltiorrhiza (EBGS), on the initial process of the coagulation cascade, which is the adhesion of platelets to activated vascular endothelium. Our results demonstrated that EBGS pretreatment of endothelial cells reduces platelet adhesion even in the presence of the monocyte-lymphocyte population. Our data indicate that EBGS exerts its effects by inhibiting the transcription of adhesion molecules, including P-selectin, platelet membrane glycoprotein GP1b, integrins αV and ß3, and reducing the secretion of the pro-inflammatory cytokines interleukin 6, interleukin 8, and the metalloproteinases MMP-2 and MMP-9. Furthermore, we demonstrated that EBGS inhibited the expression of focal adhesion kinase (FAK), strictly involved in platelet adhesion, and whose activity is correlated with that of integrin ß3. The results shown in this manuscript suggest a possible inhibitory role of the new combination EBGS in the reduction in platelet adhesion to activated endothelium, thus possibly preventing coagulation cascade initiation.


Subject(s)
Endothelium, Vascular , Platelet Adhesiveness , Signal Transduction , Tumor Necrosis Factor-alpha , Humans , Platelet Adhesiveness/drug effects , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Blood Platelets/metabolism , Blood Platelets/drug effects , Salvia miltiorrhiza/chemistry , Focal Adhesion Kinase 1/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Plant Extracts/pharmacology
2.
Carbohydr Polym ; 343: 122409, 2024 Nov 01.
Article in English | MEDLINE | ID: mdl-39174076

ABSTRACT

The study focuses on developing a bioactive shape memory sponge to address the urgent demand for short-term rapid hemostasis and long-term wound healing in noncompressible hemorrhage cases. A composite sponge was created by spontaneously generating pores and double cross-linking under mild conditions using biomimetic collagen fibril (BCF) and oxidized alginate (OA) as natural backbone, combined with an inert calcium source (Ca) from CaCO3-GDL slow gelation mechanism. The optimized BCF/OACa (5/5) sponge efficiently absorbed blood after compression and recovered to its original state within 11.2 ± 1.3 s, achieving physical hemostatic mechanism. The composite sponge accelerated physiological coagulation by promoting platelet adhesion and activation through BCF, as well as enhancing endogenous and exogenous hemostatic pathways by Ca2+. Compared to commercial PVA expanding hemostatic sponge, the composite sponge reduced bleeding volume and shortened hemostasis time in rat liver injury pick and perforation wound models. Additionally, it stimulated fibroblast migration and differentiation, thus promoting wound healing. It is biodegradable with low inflammatory response and promotes granulation tissue regeneration. In conclusion, this biocomposite sponge provides multiple hemostatic pathways and biochemical support for wound healing, is biologically safe and easy to fabricate, process and use, with significant potential for clinical translation and application.


Subject(s)
Alginates , Biomimetic Materials , Collagen , Hemorrhage , Hemostatics , Wound Healing , Alginates/chemistry , Alginates/pharmacology , Animals , Wound Healing/drug effects , Collagen/chemistry , Rats , Hemorrhage/drug therapy , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Hemostatics/pharmacology , Hemostatics/chemistry , Male , Rats, Sprague-Dawley , Hemostasis/drug effects , Oxidation-Reduction , Platelet Adhesiveness/drug effects
3.
Int J Biol Macromol ; 277(Pt 1): 134064, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39048012

ABSTRACT

The clinical utility of small-diameter vascular grafts (SDVGs) is limited due to the possibility of thrombosis and intimal hyperplasia. These features can delay the development of a functional endothelial cell (EC) monolayer on the luminal surface of grafts. Therefore, the development and fabrication of vascular grafts (VGs) with comparable extracellular matrix (ECM) functions are mandatory to elicit hemocompatible confluent EC monolayers, and angiogenesis behavior inside the body. To promote the interactions between ECs and the surface of electrospun polyacrylic acid-grafted polyhedral oligomeric silsesquioxane-poly(carbonate-urea)-urethane (PAAc-POSS-PCUU), in this research, the surface of nanofibers was modified by covalently immobilizing extracted soluble proteins from aorta (ESPA) using EDC/NHS chemistry. The ATR-FTIR spectroscopy, WCA, and SEM microscopy confirmed the binding of acrylic acid and soluble vascular proteins on the surface of electrospun fibers. The PAAc-POSS-PCUU nanofibers and engineered biomimetic Pro-PAAc-POSS-PCUU nanofibers exhibited excellent biocompatibility indicated by increased survival rate (p < 0.05). Western blotting revealed the increase of VE-cadherin, Tie-2, vWF, and VEGFR-2 in HUVECs after being plated on PAAc-POSS-PCUU and Pro-PAAc-POSS-PCUU scaffolds, indicating appropriate angiogenesis behavior (p < 0.05). Besides, the antioxidant capacity was induced by the increase of SOD and GPx activity (p < 0.05). Additionally, blood compatibility tests revealed that Pro-PAAc-POSS-PCUU nanofibers accelerate the formation of a single EC layer without hemolysis and platelet adhesion. Taken together, Pro-PAAc-POSS-PCUU nanofibers exhibited excellent blood compatibility, and angiogenesis behavior, making them a promising candidate for clinical applications.


Subject(s)
Biocompatible Materials , Blood Vessel Prosthesis , Neovascularization, Physiologic , Organosilicon Compounds , Polyurethanes , Humans , Organosilicon Compounds/chemistry , Organosilicon Compounds/pharmacology , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Neovascularization, Physiologic/drug effects , Polyurethanes/chemistry , Polyurethanes/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Nanofibers/chemistry , Materials Testing , Animals , Tissue Engineering/methods , Platelet Adhesiveness/drug effects , Angiogenesis
4.
Int J Biol Macromol ; 276(Pt 2): 133829, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39002904

ABSTRACT

In this investigation, the electrospun nanocomposite scaffolds were developed utilizing poly-3-hydroxybutyrate (PHB), zein, and multiwalled carbon nanotubes (MWCNTs) at varying concentrations of MWCNTs including 0.5 and 1 wt%. Based on the SEM evaluations, the scaffold containing 1 wt% MWCNTs (PZ-1C) exhibited the lowest fiber diameter (384 ± 99 nm) alongside a suitable porosity percentage. The presence of zein and MWCNT in the chemical structure of the scaffold was evaluated by FTIR. Furthermore, TEM images revealed the alignment of MWCNTs with the fibers. Adding 1 % MWCNTs to the PHB-zein scaffold significantly enhanced tensile strength by about 69 % and reduced elongation by about 31 %. Hydrophilicity, surface roughness, crystallinity, and biomineralization were increased by incorporating 1 wt% MWCNTs, while weight loss after in vitro degradation was decreased. The MG-63 cells exhibited enhanced attachment, viability, ALP secretion, calcium deposition, and gene expression (COLI, RUNX2, and OCN) when cultivated on the scaffold containing MWCNTs compared to the scaffolds lacking MWCNTs. Moreover, the study found that MWCNTs significantly reduced platelet adhesion and hemolysis rates below 4 %, indicating their favorable anti-hemolysis properties. Regarding the aforementioned results, the PZ-1C electrospun composite scaffold is a promising scaffold with osteogenic properties for bone tissue engineering applications.


Subject(s)
Hydroxybutyrates , Nanotubes, Carbon , Osteogenesis , Polyesters , Tissue Engineering , Tissue Scaffolds , Zein , Nanotubes, Carbon/chemistry , Zein/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Osteogenesis/drug effects , Humans , Polyesters/chemistry , Hydroxybutyrates/chemistry , Hydroxybutyrates/pharmacology , Bone and Bones/drug effects , Bone and Bones/metabolism , Hemolysis/drug effects , Prohibitins , Cell Survival/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Nanocomposites/chemistry , Cell Adhesion/drug effects , Platelet Adhesiveness/drug effects , Tensile Strength , Osteoblasts/drug effects , Osteoblasts/cytology , Porosity , Polyhydroxybutyrates
5.
Nutrients ; 16(13)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38999733

ABSTRACT

Natural and synthetic colorants present in food can modulate hemostasis, which includes the coagulation process and blood platelet activation. Some colorants have cardioprotective activity as well. However, the effect of genipin (a natural blue colorant) and synthetic blue colorants (including patent blue V and brilliant blue FCF) on hemostasis is not clear. In this study, we aimed to investigate the effects of three blue colorants-genipin, patent blue V, and brilliant blue FCF-on selected parameters of hemostasis in vitro. The anti- or pro-coagulant potential was assessed in human plasma by measuring the following coagulation times: thrombin time (TT), prothrombin time (PT), and activated partial thromboplastin time (APTT). Moreover, we used the Total Thrombus formation Analysis System (T-TAS, PL-chip) to evaluate the anti-platelet potential of the colorants in whole blood. We also measured their effect on the adhesion of washed blood platelets to fibrinogen and collagen. Lastly, the cytotoxicity of the colorants against blood platelets was assessed based on the activity of extracellular lactate dehydrogenase (LDH). We observed that genipin (at all concentrations (1-200 µM)) did not have a significant effect on the coagulation times (PT, APTT, and TT). However, genipin at the highest concentration (200 µM) and patent blue V at the concentrations of 1 and 10 µM significantly prolonged the time of occlusion measured using the T-TAS, which demonstrated their anti-platelet activity. We also observed that genipin decreased the adhesion of platelets to fibrinogen and collagen. Only patent blue V and brilliant blue FCF significantly shortened the APTT (at the concentration of 10 µM) and TT (at concentrations of 1 and 10 µM), demonstrating pro-coagulant activity. These synthetic blue colorants also modulated the process of human blood platelet adhesion, stimulating the adhesion to fibrinogen and inhibiting the adhesion to collagen. The results demonstrate that genipin is not toxic. In addition, because of its ability to reduce blood platelet activation, genipin holds promise as a novel and valuable agent that improves the health of the cardiovascular system and reduces the risk of cardiovascular diseases. However, the mechanism of its anti-platelet activity remains unclear and requires further studies. Its in vivo activity and interaction with various anti-coagulant and anti-thrombotic drugs, including aspirin and its derivatives, should be examined as well.


Subject(s)
Blood Coagulation , Blood Platelets , Food Coloring Agents , Iridoids , Humans , Iridoids/pharmacology , Blood Coagulation/drug effects , Food Coloring Agents/pharmacology , Blood Platelets/drug effects , Blood Platelets/metabolism , Hemostasis/drug effects , Partial Thromboplastin Time , Platelet Adhesiveness/drug effects , Fibrinogen/metabolism , Benzenesulfonates/pharmacology , Prothrombin Time , Rosaniline Dyes/pharmacology , Hemostatics/pharmacology , Platelet Activation/drug effects , Thrombin Time
6.
ACS Appl Mater Interfaces ; 16(30): 39104-39116, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39036941

ABSTRACT

Surface modification using zwitterionic 2-methacryloyloxyethylphosphorylcholine (MPC) polymers is one of the most reasonable ways to prepare medical devices that can suppress undesired biological reactions such as blood coagulation. Usable MPC polymers are hydrophilic and water soluble, and their surface modification strategy involves exploiting the copolymer structures by adding physical or chemical bonding moieties. In this study, we developed copolymers composed of MPC, hydrophobic anchoring moiety, and chemical cross-linking unit to clarify the role of hydrophobic interactions in achieving biocompatible and long-term stable coatings. The four kinds of MPC copolymers with cross-linking units, such as 3-methacryloxypropyl trimethoxysilane (MPTMSi), and four different hydrophobic anchoring moieties, such as 3-(methacryloyloxy)propyltris(trimethylsiloxy)silane (MPTSSi) named as PMMMSi, n-butyl methacrylate (BMA) as PMBSi, 2-ethylhexyl methacrylate (EHMA) as PMESi, and lauryl methacrylate as PMLSi, were synthesized and coated on polydimethylsiloxane, polypropylene (PP), and polymethyl pentene. These copolymers were uniformly coated on the substrate materials PP and poly(methyl pentene) (PMP), to achieve hydrophilic and electrically neutral coatings. The results of the antibiofouling test showed that PMBSi repelled the adsorption of fluorescence-labeled bovine serum albumin the most, whereas PMLSi repelled it the least. Notably, all four copolymers suppressed platelet adhesion similarly. The variations in protein adsorption quantities among the four copolymer coatings were attributed to their distinct swelling behaviors in aqueous environments. Further investigations, including 3D scanning force microscopy and neutron reflectivity measurements, revealed that the PMLSi coating exhibited a higher water intake under aqueous conditions in comparison to the other coatings. Consequently, all copolymer coatings effectively prevented the invasion of platelets but the proteins penetrated the PMLSi network. Subsequently, the dynamic stability required to induce shear stress was evaluated using a circulation system. The results demonstrated that the PMMMSi and PMLSi coatings on PMP and PP exhibited exceptional platelet repellency and maintained high stability during circulation. This study highlights the potential of hydrophobic moieties to improve hemocompatibility and stability, offering potential applications in medical devices.


Subject(s)
Hydrophobic and Hydrophilic Interactions , Phosphorylcholine/chemistry , Phosphorylcholine/analogs & derivatives , Polymers/chemistry , Animals , Cross-Linking Reagents/chemistry , Coated Materials, Biocompatible/chemistry , Platelet Adhesiveness/drug effects , Surface Properties , Serum Albumin, Bovine/chemistry , Humans , Methacrylates/chemistry , Phospholipids/chemistry , Cattle
7.
Sci Rep ; 14(1): 14109, 2024 06 19.
Article in English | MEDLINE | ID: mdl-38898080

ABSTRACT

Developing a reliable method to predict thrombocytopenia is imperative in drug discovery. Here, we establish an assay using a microphysiological system (MPS) to recapitulate the in-vivo mechanisms of platelet aggregation and adhesion. This assay highlights the role of shear stress on platelet aggregation and their interactions with vascular endothelial cells. Platelet aggregation induced by soluble collagen was detected under agitated, but not static, conditions using a plate shaker and gravity-driven flow using MPS. Notably, aggregates adhered on vascular endothelial cells under gravity-driven flow in the MPS, and this incident increased in a concentration-dependent manner. Upon comparing the soluble collagen-induced aggregation activity in platelet-rich plasma (PRP) and whole blood, remarkable platelet aggregate formation was observed at concentrations of 30 µg/mL and 3 µg/mL in PRP and whole blood, respectively. Moreover, ODN2395, an oligonucleotide, induced platelet aggregation and adhesion to vascular endothelial cells. SYK inhibition, which mediated thrombogenic activity via glycoprotein VI on platelets, ameliorated platelet aggregation in the system, demonstrating that the mechanism of platelet aggregation was induced by soluble collagen and oligonucleotide. Our evaluation system partially recapitulated the aggregation mechanisms in blood vessels and can contribute to the discovery of safe drugs to mitigate the risk of thrombocytopenia.


Subject(s)
Blood Platelets , Platelet Aggregation , Thrombocytopenia , Platelet Aggregation/drug effects , Humans , Thrombocytopenia/chemically induced , Blood Platelets/drug effects , Blood Platelets/metabolism , Collagen/metabolism , Collagen/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Platelet Adhesiveness/drug effects , Syk Kinase/metabolism , Syk Kinase/antagonists & inhibitors , Platelet-Rich Plasma/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Microphysiological Systems
8.
Int J Biol Macromol ; 274(Pt 1): 133265, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38909732

ABSTRACT

Hemostasis is the first step of emergency medical treatment. It is particularly important to develop rapid-acting and efficacious hemostatic materials. Carboxymethyl chitosan (CMCS), sodium alginate (SA) and Resina Draconis (RD) were composited uniformly by polyelectrolyte blending. Their composite sponges (CMCS/SA/RD) were prepared by freeze-induced phase separation. CMCS/SA/RD sponges were characterized by Fourier transform infrared spectroscopy and scanning electron microscopy, and their blood absorption and hemolysis ratio were analyzed. The hemostatic effect of the composite sponges was evaluated by coagulation in vitro and in vivo. The composite sponges had a porous network structure. The water absorption ratio was >8000 %, and hemolysis ratio was <5 %. CMCS/SA/RD-II and CMCS/SA/RD-III composite sponges shortened the coagulation time in vitro by 11.33 s and 9.66 s, the hepatic hemostasis time by 13.8 % and 23.3 %, and the hemostasis time after mouse-tail amputation by 28.9 % and 23.9 %, respectively. A preliminary study on its coagulation mechanism showed that CMCS/SA/RD had significant effects on erythrocyte adsorption, platelet adhesion, and shortening of the activated partial thromboplastin time.


Subject(s)
Alginates , Blood Coagulation , Chitosan , Hemostasis , Hemostatics , Chitosan/chemistry , Chitosan/analogs & derivatives , Alginates/chemistry , Animals , Hemostasis/drug effects , Mice , Hemostatics/chemistry , Hemostatics/pharmacology , Blood Coagulation/drug effects , Hemolysis/drug effects , Platelet Adhesiveness/drug effects , Spectroscopy, Fourier Transform Infrared
9.
Int J Biol Macromol ; 272(Pt 2): 132747, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38821301

ABSTRACT

Degradable magnesium alloy stents are considered to be ideal candidates to replace the traditional non-degradable stents for the treatment of cardiovascular diseases. However, bare magnesium alloy stents usually degrade too fast and show poor hemocompatibility and cytocompatibility, which seriously affects their clinical use. In this study, surface modification based on the MgF2 layer, polydopamine (PDA) coating, fucoidan and CAG peptides was performed on the Mg-Zn-Y-Nd (ZE21B) magnesium alloy with the purpose of improving its corrosion resistance, hemocompatibility and cytocompatibility for vascular stent application. After modification, the ZE21B alloy showed better corrosion resistance. Moreover, the lower hemolysis rate, platelet adhesion and activation, and fibrinogen adsorption and denaturation proved the improved hemocompatibility of modified ZE21B alloy in in vitro blood experiments. Furthermore, the co-immobilization of fucoidan and CAG peptides significantly promoted the adhesion, proliferation, migration and NO release of endothelial cells (ECs) on the modified ZE21B alloy, and meanwhile the modification with fucoidan and CAG peptides inhibited the adhesion and proliferation of smooth muscle cells (SMCs) and suppressed the expression of proinflammatory factors in the macrophages (MAs). The surface modification obviously enhanced the corrosion resistance, hemocompatibility and cytocompatibility of ZE21B alloy, and provided an effective strategy for the development of degradable vascular stents.


Subject(s)
Alloys , Cell Adhesion , Magnesium , Materials Testing , Peptides , Polysaccharides , Alloys/chemistry , Alloys/pharmacology , Polysaccharides/chemistry , Polysaccharides/pharmacology , Humans , Peptides/chemistry , Peptides/pharmacology , Magnesium/chemistry , Cell Adhesion/drug effects , Animals , Cell Proliferation/drug effects , Hemolysis/drug effects , Corrosion , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Platelet Adhesiveness/drug effects , Mice , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Surface Properties , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Aquatic Organisms/chemistry , Indoles , Polymers
10.
Catheter Cardiovasc Interv ; 104(1): 10-20, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38769726

ABSTRACT

BACKGROUND: During the transition from dual antiplatelet therapy (DAPT) to single antiplatelet therapy (SAPT), previous studies have raised concerns about a rebound effect. We compared platelet and inflammatory cell adhesion on different types of stents in the setting of clopidogrel presence and withdrawal. METHODS: In Experiment 1, three pigs were administered with DAPT, that is, clopidogrel and acetylsalicylic acid (ASA), for 7 days. Each animal underwent an extracorporeal carotid arteriovenous shunt model implanted with fluoropolymer-coated everolimus-eluting stent (FP-EES), biodegradable-polymer sirolimus-eluting stent (BP-SES), and biodegradable-polymer everolimus-eluting stents (BP-EES). In Experiment 2, two pigs were administered DAPT, clopidogrel was then withdrawn at day 7, and SAPT with ASA was continued for next 21 days. Then flow-loop experiments with the drawn blood from each time point were performed for FP-EES, BioLinx-polymer zotarolimus-eluting stents (BL-ZES), and BP-EES. The rebound effect was defined as the statistical increase of inflammation and platelet adhesion assessed with immunohistochemistry on the stent-strut level basis from baseline to day-14 or 28. RESULTS: Both experiments showed platelet adhesion value was highest in BP-EES, while the least in FP-EES during DAPT therapy. There was no increase in platelet or inflammatory cell adhesion above baseline values (i.e., no therapy) due to the cessation of clopidogrel on the stent-strut level. Monocyte adhesion was the least for FP-EES with the same trend observed for neutrophil adhesion. CONCLUSIONS: No evidence of rebound effect was seen after the transition from DAPT to SAPT. FP-EES demonstrated the most favorable antithrombotic and anti-inflammatory profile regardless of the different experimental designs.


Subject(s)
Aspirin , Clopidogrel , Drug-Eluting Stents , Dual Anti-Platelet Therapy , Everolimus , Platelet Adhesiveness , Platelet Aggregation Inhibitors , Prosthesis Design , Sirolimus , Thrombosis , Animals , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/adverse effects , Clopidogrel/administration & dosage , Clopidogrel/pharmacology , Time Factors , Sirolimus/analogs & derivatives , Sirolimus/administration & dosage , Sirolimus/pharmacology , Everolimus/administration & dosage , Everolimus/pharmacology , Thrombosis/prevention & control , Thrombosis/etiology , Aspirin/administration & dosage , Platelet Adhesiveness/drug effects , Arteriovenous Shunt, Surgical/adverse effects , Sus scrofa , Blood Platelets/drug effects , Blood Platelets/metabolism , Drug Administration Schedule , Disease Models, Animal
11.
ACS Appl Mater Interfaces ; 16(22): 28134-28146, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38768602

ABSTRACT

Vessel transplantation is currently considered the "gold standard" treatment for cardiovascular disease. However, ideal artificial vascular grafts should possess good biocompatibility and mechanical strength that match those of native autologous vascular tissue to promote in vivo tissue regeneration. In this study, a series of dynamic cross-linking double-network hydrogels and the resultant hydrogel tubes were prepared. The hydrogels (named PCO), composed of rigid poly(vinyl alcohol) (PVA), flexible carboxymethyl chitosan (CMCS), and a cross-linker of aldehyde-based ß-cyclodextrin (OCD), were formed in a double-network structure with multiple dynamical cross-linking including dynamic imine bonds, hydrogen bonds, and microcrystalline regions. The PCO hydrogels exhibited superior mechanical strength, good network stability, and fatigue resistance. Additionally, it demonstrated excellent cell and blood compatibility. The results showed that the introduction of CMCS/OCD led to a significant increase in the proliferation rate of endothelial cells seeded on the surface of the hydrogel. The hemolysis rate in the test was lower than 0.3%, and both protein adsorption and platelet adhesion were reduced, indicating an excellent anticoagulant function. The plasma recalcification time test results showed that endogenous coagulation was alleviated to some extent. When formed into blood vessels and incubated with blood, no thrombus formation was observed, and there was minimal red blood cell aggregation. Therefore, this novel hydrogel tube, with excellent mechanical properties, exhibits antiadhesive characteristics toward blood cells and proteins, as well as antithrombotic properties, making it hold tremendous potential for applications in the biomedical and engineering fields.


Subject(s)
Biocompatible Materials , Chitosan , Hydrogels , Polyvinyl Alcohol , Hydrogels/chemistry , Hydrogels/pharmacology , Hydrogels/chemical synthesis , Chitosan/chemistry , Chitosan/analogs & derivatives , Chitosan/pharmacology , Humans , Polyvinyl Alcohol/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Biocompatible Materials/chemical synthesis , Blood Vessel Prosthesis , Materials Testing , beta-Cyclodextrins/chemistry , Human Umbilical Vein Endothelial Cells/drug effects , Cell Proliferation/drug effects , Hemolysis/drug effects , Animals , Platelet Adhesiveness/drug effects , Cross-Linking Reagents/chemistry
12.
Colloids Surf B Biointerfaces ; 239: 113927, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38714078

ABSTRACT

Humans often have bleeding, which exerts substantial selective pressure on the coagulation system to optimize hemostasis in a variety of situations. Uncontrolled hemorrhage due to severe trauma leads to morbidity and mortality. Although nonbiological surfaces such as silicates can activate coagulation factor XII (FXII), the presence of Zn (Zinc) in the material stimulates and activates the various steps in the coagulation cascade. This results in blood clotting. The Zn@SiO2 nanocomposite has an excellent hemostatic property that establishes hemostasis by activating the factors responsible for the formation of a stable clot called fibrin mesh. This can be used as a hemostatic agent during surgeries and in any other trauma condition related to bleeding. Zn@SiO2 was synthesized and characterized with XRD, FTIR and HRTEM. It is analyzed for its RBC (Red Blood Corpuscles) aggregation and Platelet adhesion ability, fibrin formation, thrombus formation and prothrombin time (PT), Activated Partial Thromboplastin Time (aPTT), D-dimer for its ability to activate the coagulation cascade to achieve stable clotting.


Subject(s)
Blood Coagulation , Nanocomposites , Silicon Dioxide , Zinc , Nanocomposites/chemistry , Blood Coagulation/drug effects , Zinc/chemistry , Zinc/pharmacology , Silicon Dioxide/chemistry , Humans , Partial Thromboplastin Time , Prothrombin Time , Platelet Adhesiveness/drug effects , Animals , Particle Size , Fibrin Fibrinogen Degradation Products/metabolism , Hemostatics/chemistry , Hemostatics/pharmacology
13.
Int J Biol Macromol ; 270(Pt 2): 132181, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38740155

ABSTRACT

Nowadays, developing vascular grafts (e.g., vascular patches and tubular grafts) is challenging. Bacterial cellulose (BC) with 3D fibrous network has been widely investigated for vascular applications. In this work, different from BC vascular patch cultured with the routine culture medium, dopamine (DA)-containing culture medium is employed to in situ synthesize dense BC fibrous structure with significantly increased fiber diameter and density. Simultaneously, BC fibers are modified by DA during in situ synthesis process. Then DA on BC fibers can self-polymerize into polydopamine (PDA) accompanied with the removal of bacteria in NaOH solution, obtaining PDA-modified dense BC (PDBC) vascular patch. Heparin (Hep) is subsequently covalently immobilized on PDBC fibers to form Hep-immobilized PDBC (Hep@PDBC) vascular patch. The obtained results indicate that Hep@PDBC vascular patch exhibits remarkable tensile and burst strength due to its dense fibrous structure. More importantly, compared with BC and PDBC vascular patches, Hep@PDBC vascular patch not only displays reduced platelet adhesion and improved anticoagulation activity, but also promotes the proliferation, adhesion, spreading, and protein expression of human umbilical vein endothelial cells, contributing to the endothelialization process. The combined strategy of in situ densification and Hep immobilization provides a feasible guidance for the construction of BC-based vascular patches.


Subject(s)
Blood Vessel Prosthesis , Cellulose , Heparin , Human Umbilical Vein Endothelial Cells , Cellulose/chemistry , Heparin/chemistry , Heparin/pharmacology , Humans , Platelet Adhesiveness/drug effects , Cell Proliferation/drug effects , Polymers/chemistry , Polymers/pharmacology , Indoles/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology
14.
Int J Biol Macromol ; 271(Pt 2): 132487, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38768910

ABSTRACT

Due to its biofunctions similar to NO, the CO gas signaling molecule has gradually shown great potential in cardiovascular biomaterials for regulating the in vivo performances after the implantation and has received increasing attention. To construct a bioactive surface with CO-releasing properties on the surface of magnesium-based alloy to augment the anticorrosion and biocompatibility, graphene oxide (GO) was firstly modified using carboxymethyl chitosan (CS), and then CO-releasing molecules (CORM401) were introduced to synthesize a novel biocompatible nanomaterial (GOCS-CO) that can release CO in the physiological environments. The GOCS-CO was further immobilized on the magnesium alloy surface modified by polydopamine coating with Zn2+ (PDA/Zn) to create a bioactive surface capable of releasing CO in the physiological environment. The outcomes showed that the CO-releasing coating can not only significantly enhance the anticorrosion and abate the corrosion degradation rate of the magnesium alloy in a simulated physiological environment, but also endow it with good hydrophilicity and a certain ability to adsorb albumin selectively. Owing to the significant enhancement of anticorrosion and hydrophilicity, coupled with the bioactivity of GOCS, the modified sample not only showed excellent ability to prevent platelet adhesion and activation and reduce hemolysis rate but also can promote endothelial cell (EC) adhesion, proliferation as well as the expression of nitric oxide (NO) and vascular endothelial growth factor (VEGF). In the case of CO release, the hemocompatibility and EC growth behaviors were further significantly improved, suggesting that CO molecules released from the surface can significantly improve the hemocompatibility and EC growth. Consequently, the present study provides a novel surface modification method that can simultaneously augment the anticorrosion and biocompatibility of magnesium-based alloys, which will strongly promote the research and application of CO-releasing bioactive coatings for surface functionalization of cardiovascular biomaterials and devices.


Subject(s)
Alloys , Chitosan , Coated Materials, Biocompatible , Graphite , Magnesium , Graphite/chemistry , Chitosan/chemistry , Chitosan/analogs & derivatives , Chitosan/pharmacology , Magnesium/chemistry , Magnesium/pharmacology , Alloys/chemistry , Alloys/pharmacology , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Humans , Carbon Monoxide/chemistry , Carbon Monoxide/pharmacology , Stents , Hemolysis/drug effects , Platelet Adhesiveness/drug effects , Corrosion , Cell Adhesion/drug effects , Materials Testing , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Proliferation/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Animals , Endothelial Cells/drug effects
15.
Int J Biol Macromol ; 269(Pt 1): 132040, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38702003

ABSTRACT

Decellularized vascular tissue has high potential as a tissue-engineered vascular graft because of its similarity to native vessels in terms of mechanical strength. However, exposed collagen on the tissue induces blood coagulation, and low hemocompatibility is a major obstacle to its vascular application. Here we report that freeze-drying and ethanol treatment effectively modify collagen fiber structure and drastically reduce blood coagulation on the graft surface without exogenous chemical modification. Decellularized carotid artery of ostrich was treated with freeze-drying and ethanol solution at concentrations ranging between 5 and 99.5 %. Collagen fiber distance in the graft was narrowed by freeze-drying, and the non-helical region increased by ethanol treatment. Although in vitro blood coagulation pattern was similar on the grafts, platelet adhesion on the grafts was largely suppressed by freeze-drying and ethanol treatments. Ex vivo blood circulation tests also indicated that the adsorption of platelets and Von Willebrand Factor was largely reduced to approximately 80 % by ethanol treatment. These results indicate that structural modification of collagen fibers in decellularized tissue reduces blood coagulation on the surface by inhibiting platelet adhesion.


Subject(s)
Blood Coagulation , Collagen , Platelet Adhesiveness , Animals , Platelet Adhesiveness/drug effects , Blood Coagulation/drug effects , Collagen/chemistry , Tissue Engineering/methods , Materials Testing , Freeze Drying , Blood Vessel Prosthesis , Tissue Scaffolds/chemistry , Blood Platelets/metabolism , Blood Platelets/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Carotid Arteries/drug effects , Humans , Ethanol/chemistry
16.
Acta Biomater ; 180: 372-382, 2024 05.
Article in English | MEDLINE | ID: mdl-38614415

ABSTRACT

Catheter-induced thrombosis is a major contributor to infectious and mechanical complications of biomaterials that lead to device failure. Herein, a dualfunction submicron textured nitric oxide (NO)-releasing catheter was developed. The hemocompatibility and antithrombotic activity of vascular catheters were evaluated in both 20 h in vitro blood loop and 7 d in vivo rabbit model. Surface characterization assessments via atomic force microscopy show the durability of the submicron pattern after incorporation of NO donor S-nitroso-N-acetylpenicillamine (SNAP). The SNAP-doped catheters exhibited prolonged and controlled NO release mimicking the levels released by endothelium. Fabricated catheters showed cytocompatibility when evaluated against BJ human fibroblast cell lines. After 20h in vitro evaluation of catheters in a blood loop, textured-NO catheters exhibited a 13-times reduction in surface thrombus formation compared to the control catheters, which had 83% of the total area covered by clots. After the 7 d in vivo rabbit model, analysis on the catheter surface was examined via scanning electron microscopy, where significant reduction of platelet adhesion, fibrin mesh, and thrombi can be observed on the NO-releasing textured surfaces. Moreover, compared to relative controls, a 63% reduction in the degree of thrombus formation within the jugular vein was observed. Decreased levels of fibrotic tissue decomposition on the jugular vein and reduced platelet adhesion and thrombus formation on the texture of the NO-releasing catheter surface are indications of mitigated foreign body response. This study demonstrated a biocompatible and robust dual-functioning textured NO PU catheter in limiting fouling-induced complications for longer-term blood-contacting device applications. STATEMENT OF SIGNIFICANCE: Catheter-induced thrombosis is a major contributor to infectious and mechanical complications of biomaterials that lead to device failure. This study demonstrated a robust, biocompatible, dual-functioning textured nitric oxide (NO) polyurethane catheter in limiting fouling-induced complications for longer-term blood-contacting device applications. The fabricated catheters exhibited prolonged and controlled NO release that mimics endothelium levels. After the 7 d in vivo model, a significant reduction in platelet adhesion, fibrin mesh, and thrombi was observed on the NO-releasing textured catheters, along with decreased levels of fibrotic tissue decomposition on the jugular vein. Results illustrate that NO-textured catheter surface mitigates foreign body response.


Subject(s)
Catheters , Nitric Oxide , S-Nitroso-N-Acetylpenicillamine , Animals , Rabbits , Nitric Oxide/metabolism , Humans , S-Nitroso-N-Acetylpenicillamine/pharmacology , S-Nitroso-N-Acetylpenicillamine/chemistry , Thrombosis/pathology , Materials Testing , Cell Line , Platelet Adhesiveness/drug effects , Disease Models, Animal
17.
Colloids Surf B Biointerfaces ; 238: 113908, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677153

ABSTRACT

In response to the critical demand for advancements in coronary artery stents, this study addresses the challenges associated with arterial recoil and restenosis post-angioplasty and the imperative to encourage rapid re-endothelialization for minimizing thrombosis risks. We employed an innovative approach inspired by mussel adhesion, incorporating placental anticoagulant protein (AnnexinV) on stent design. The introduction of a post-translationally modified catecholic amino acid L-3,4-dihydroxyphenylalanine (L-Dopa), mimicking mussel characteristics, allowed for effective surface modification of Stainless steel stents through genetic code engineering in AnnexinV (AnxDopa). The efficacy of AnxDopa was analyzed through microscale thermophoresis and flow cytometry, confirming AnxDopa's exceptional binding with phosphatidylserine and activated platelets. AnxDopa coated stainless steel demonstrates remarkable bio-, hemo-, and immuno-compatibility, preventing smooth muscle cell proliferation, platelet adhesion, and fibrin formation. It acts as an interface between the stent and biological fluid, which facilitates the anticoagulation and rapid endothelialization. Surface modification of SS verified through XPS analysis and contact angle measurement attests to the efficacy of AnxDopa mediated surface modification. The hydrophilic nature of the AnxDopa-coated surface enhanced the endothelialization through increased protein absorption. This approach represents a significant stride in developing coronary stents with improved biocompatibility and reduced restenosis risks, offering valuable contributions to scientific and clinical realms alike.


Subject(s)
Coated Materials, Biocompatible , Stents , Humans , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Coronary Vessels/drug effects , Platelet Adhesiveness/drug effects , Anticoagulants/pharmacology , Anticoagulants/chemistry , Surface Properties , Cell Proliferation/drug effects , Stainless Steel/chemistry , Blood Platelets/drug effects , Blood Platelets/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/cytology , Animals , Levodopa/chemistry , Levodopa/pharmacology
18.
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
19.
Phytomedicine ; 128: 155420, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38547619

ABSTRACT

BACKGROUND: Within the pro-metastatic hemato-microenvironment, interaction between platelets and tumor cells provides essential support for tumor cells by inducing Epithelial-Mesenchymal Transition (EMT), which greatly increases the stemness of colon cancer cells. Pharmacologically, although platelet deactivation has proved to be benefit against metastasis, its wide application is severely restricted due to the bleeding risk. Spatholobi Caulis, a traditional Chinese herb with circulatory promotion and blood stasis removal activity, has been proved to be clinically effective in malignant medication, leaving its mechanistic relevance to tumor-platelet interaction largely unknown. METHODS: Firstly, MC38-Luc cells were injected into tail-vein in C57BL/6 mice to establish hematogenous metastasis model and the anti-metastasis effects of SEA were evaluated by using a small-animal imaging system. Then, we evaluated the anti-tumor-platelet interaction efficacy of SEA using a tumor-specific induced platelet aggregation model. Platelet aggregation was specifically induced by tumor cells in vitro. Furthermore, to clarify the anti-metastatic effects of SEA is mainly attributed to its blockage on tumor-platelet interaction, after co-culture with tumor cells and platelets (with or without SEA), MC38-Luc cells were injected into the tail-vein and finally count the total of photons quantitatively. Besides, to clarify the blocking pattern of SEA within the tumor-platelet complex, the dependence of SEA on different fractions from activated platelets was tested. Lastly, molecular docking screening were performed to screen potential effective compounds and we used ß-catenin blockers to verify the pathways involved in SEA blocking tumor-platelet interaction. RESULTS: Our study showed that SEA was effective in blocking tumor-platelet specific interaction: (1) Through CCK-8 and LDH assays, SEA showed no cytotoxic effects on tumor cells and platelets. On this basis, by the tail vein injection model, the photon counts in the SEA group was significantly lower than model group, indicating that SEA effectively reduced metastasis. (2) In the "tumor-platelet" co-culture model, SEA effectively inhibited the progression of EMT and cancer stemness signatures of MC38 cells in the model group. (3) In mechanism study, by using the specific inhibitors for galectin-3 (GB1107) andWNT (IWR) respectively, we proved that SEA inhibits the activation of the galectin-3-mediated ß-catenin activation. CONCLUSION: By highlighting the pro-metastatic effects of galectin-3-mediated tumor-platelet adhesion, our study provided indicative evidence for Spatholobi Caulis as the representative candidate for anti-metastatic therapy.


Subject(s)
Colonic Neoplasms , Mice, Inbred C57BL , Tumor Microenvironment , Animals , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Tumor Microenvironment/drug effects , Cell Line, Tumor , Blood Platelets/drug effects , Mice , Platelet Aggregation/drug effects , Platelet Adhesiveness/drug effects , Epithelial-Mesenchymal Transition/drug effects , Humans , Plant Extracts/pharmacology , Neoplasm Metastasis
20.
Blood ; 143(19): 1992-2004, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38290109

ABSTRACT

ABSTRACT: Activation of von Willebrand factor (VWF) is a tightly controlled process governed primarily by local elements around its A1 domain. Recent studies suggest that the O-glycosylated sequences flanking the A1 domain constitute a discontinuous and force-sensitive autoinhibitory module (AIM), although its extent and conformation remains controversial. Here, we used a targeted screening strategy to identify 2 groups of nanobodies. One group, represented by clone 6D12, is conformation insensitive and binds the N-terminal AIM (NAIM) sequence that is distal from A1; 6D12 activates human VWF and induces aggregation of platelet-rich plasma at submicromolar concentrations. The other group, represented by clones Nd4 and Nd6, is conformation sensitive and targets the C-terminal AIM (CAIM). Nd4 and Nd6 inhibit ristocetin-induced platelet aggregation and reduce VWF-mediated platelet adhesion under flow. A crystal structure of Nd6 in complex with AIM-A1 shows a novel conformation of both CAIM and NAIM that are primed to interact, providing a model of steric hindrance stabilized by the AIM as the mechanism for regulating GPIbα binding to VWF. Hydrogen-deuterium exchange mass spectrometry analysis shows that binding of 6D12 induces the exposure of the GPIbα-binding site in the A1 domain, but binding of inhibitory nanobodies reduces it. Overall, these results suggest that the distal portion of NAIM is involved in specific interactions with CAIM, and binding of nanobodies to the AIM could either disrupt its conformation to activate VWF or stabilize its conformation to upkeep VWF autoinhibition. These reported nanobodies could facilitate future studies of VWF functions and related pathologies.


Subject(s)
Single-Domain Antibodies , von Willebrand Factor , von Willebrand Factor/metabolism , von Willebrand Factor/chemistry , Humans , Single-Domain Antibodies/pharmacology , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/metabolism , Platelet Aggregation/drug effects , Protein Conformation , Protein Domains , Protein Binding , Platelet Adhesiveness/drug effects , Crystallography, X-Ray , Animals , Blood Platelets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL