Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.703
Filtrar
1.
Sci Rep ; 14(1): 14109, 2024 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-38898080

RESUMEN

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.


Asunto(s)
Plaquetas , Agregación Plaquetaria , Trombocitopenia , Agregación Plaquetaria/efectos de los fármacos , Humanos , Trombocitopenia/inducido químicamente , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Colágeno/metabolismo , Colágeno/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Adhesividad Plaquetaria/efectos de los fármacos , Quinasa Syk/metabolismo , Quinasa Syk/antagonistas & inhibidores , Plasma Rico en Plaquetas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Sistemas Microfisiológicos
2.
ACS Appl Mater Interfaces ; 16(22): 28134-28146, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38768602

RESUMEN

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.


Asunto(s)
Materiales Biocompatibles , Quitosano , Hidrogeles , Alcohol Polivinílico , Hidrogeles/química , Hidrogeles/farmacología , Hidrogeles/síntesis química , Quitosano/química , Quitosano/análogos & derivados , Quitosano/farmacología , Humanos , Alcohol Polivinílico/química , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/síntesis química , Prótesis Vascular , Ensayo de Materiales , beta-Ciclodextrinas/química , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Hemólisis/efectos de los fármacos , Animales , Adhesividad Plaquetaria/efectos de los fármacos , Reactivos de Enlaces Cruzados/química
3.
Colloids Surf B Biointerfaces ; 239: 113927, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38714078

RESUMEN

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.


Asunto(s)
Coagulación Sanguínea , Nanocompuestos , Dióxido de Silicio , Zinc , Nanocompuestos/química , Coagulación Sanguínea/efectos de los fármacos , Zinc/química , Zinc/farmacología , Dióxido de Silicio/química , Humanos , Tiempo de Tromboplastina Parcial , Tiempo de Protrombina , Adhesividad Plaquetaria/efectos de los fármacos , Animales , Tamaño de la Partícula , Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Hemostáticos/química , Hemostáticos/farmacología
4.
Int J Biol Macromol ; 271(Pt 2): 132487, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38768910

RESUMEN

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.


Asunto(s)
Aleaciones , Quitosano , Materiales Biocompatibles Revestidos , Grafito , Magnesio , Grafito/química , Quitosano/química , Quitosano/análogos & derivados , Quitosano/farmacología , Magnesio/química , Magnesio/farmacología , Aleaciones/química , Aleaciones/farmacología , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Humanos , Monóxido de Carbono/química , Monóxido de Carbono/farmacología , Stents , Hemólisis/efectos de los fármacos , Adhesividad Plaquetaria/efectos de los fármacos , Corrosión , Adhesión Celular/efectos de los fármacos , Ensayo de Materiales , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Proliferación Celular/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Animales , Células Endoteliales/efectos de los fármacos
5.
Int J Biol Macromol ; 272(Pt 2): 132747, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38821301

RESUMEN

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.


Asunto(s)
Aleaciones , Adhesión Celular , Magnesio , Ensayo de Materiales , Péptidos , Polisacáridos , Aleaciones/química , Aleaciones/farmacología , Polisacáridos/química , Polisacáridos/farmacología , Humanos , Péptidos/química , Péptidos/farmacología , Magnesio/química , Adhesión Celular/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Hemólisis/efectos de los fármacos , Corrosión , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Adhesividad Plaquetaria/efectos de los fármacos , Ratones , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Propiedades de Superficie , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Organismos Acuáticos/química , Indoles , Polímeros
6.
Int J Biol Macromol ; 270(Pt 2): 132181, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38740155

RESUMEN

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.


Asunto(s)
Prótesis Vascular , Celulosa , Heparina , Células Endoteliales de la Vena Umbilical Humana , Celulosa/química , Heparina/química , Heparina/farmacología , Humanos , Adhesividad Plaquetaria/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Polímeros/química , Polímeros/farmacología , Indoles/química , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología
7.
Int J Biol Macromol ; 269(Pt 1): 132040, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38702003

RESUMEN

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.


Asunto(s)
Coagulación Sanguínea , Colágeno , Adhesividad Plaquetaria , Animales , Adhesividad Plaquetaria/efectos de los fármacos , Coagulación Sanguínea/efectos de los fármacos , Colágeno/química , Ingeniería de Tejidos/métodos , Ensayo de Materiales , Liofilización , Prótesis Vascular , Andamios del Tejido/química , Plaquetas/metabolismo , Plaquetas/química , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Arterias Carótidas/efectos de los fármacos , Humanos , Etanol/química
8.
Acta Biomater ; 180: 372-382, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38614415

RESUMEN

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.


Asunto(s)
Catéteres , Óxido Nítrico , S-Nitroso-N-Acetilpenicilamina , Animales , Conejos , Óxido Nítrico/metabolismo , Humanos , S-Nitroso-N-Acetilpenicilamina/farmacología , S-Nitroso-N-Acetilpenicilamina/química , Trombosis/patología , Ensayo de Materiales , Línea Celular , Adhesividad Plaquetaria/efectos de los fármacos , Modelos Animales de Enfermedad
9.
Colloids Surf B Biointerfaces ; 238: 113908, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38677153

RESUMEN

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.


Asunto(s)
Materiales Biocompatibles Revestidos , Stents , Humanos , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Vasos Coronarios/efectos de los fármacos , Adhesividad Plaquetaria/efectos de los fármacos , Anticoagulantes/farmacología , Anticoagulantes/química , Propiedades de Superficie , Proliferación Celular/efectos de los fármacos , Acero Inoxidable/química , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/citología , Animales , Levodopa/química , Levodopa/farmacología
10.
Langmuir ; 40(18): 9661-9668, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38664943

RESUMEN

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.


Asunto(s)
Colágeno , Péptidos , Colágeno/química , Péptidos/química , Péptidos/farmacología , Fibrinolíticos/química , Fibrinolíticos/farmacología , Secuencia de Aminoácidos , Diseño de Fármacos , Humanos , Redes Neurales de la Computación , Simulación de Dinámica Molecular , Adhesividad Plaquetaria/efectos de los fármacos
11.
Phytomedicine ; 128: 155420, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38547619

RESUMEN

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.


Asunto(s)
Neoplasias del Colon , Ratones Endogámicos C57BL , Microambiente Tumoral , Animales , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Microambiente Tumoral/efectos de los fármacos , Línea Celular Tumoral , Plaquetas/efectos de los fármacos , Ratones , Agregación Plaquetaria/efectos de los fármacos , Adhesividad Plaquetaria/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Extractos Vegetales/farmacología , Metástasis de la Neoplasia
12.
Blood ; 143(19): 1992-2004, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38290109

RESUMEN

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.


Asunto(s)
Anticuerpos de Dominio Único , Factor de von Willebrand , Factor de von Willebrand/metabolismo , Factor de von Willebrand/química , Humanos , Anticuerpos de Dominio Único/farmacología , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Conformación Proteica , Dominios Proteicos , Unión Proteica , Adhesividad Plaquetaria/efectos de los fármacos , Cristalografía por Rayos X , Animales , Plaquetas/metabolismo
13.
Int J Mol Sci ; 23(1)2022 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-35008919

RESUMEN

Understanding the pathways involved in the formation and stability of the core and shell regions of a platelet-rich arterial thrombus may result in new ways to treat arterial thrombosis. The distinguishing feature between these two regions is the absence of fibrin in the shell which indicates that in vitro flow-based assays over thrombogenic surfaces, in the absence of coagulation, can be used to resemble this region. In this study, we have investigated the contribution of Syk tyrosine kinase in the stability of platelet aggregates (or thrombi) formed on collagen or atherosclerotic plaque homogenate at arterial shear (1000 s-1). We show that post-perfusion of the Syk inhibitor PRT-060318 over preformed thrombi on both surfaces enhances thrombus breakdown and platelet detachment. The resulting loss of thrombus stability led to a reduction in thrombus contractile score which could be detected as early as 3 min after perfusion of the Syk inhibitor. A similar loss of thrombus stability was observed with ticagrelor and indomethacin, inhibitors of platelet adenosine diphosphate (ADP) receptor and thromboxane A2 (TxA2), respectively, and in the presence of the Src inhibitor, dasatinib. In contrast, the Btk inhibitor, ibrutinib, causes only a minor decrease in thrombus contractile score. Weak thrombus breakdown is also seen with the blocking GPVI nanobody, Nb21, which indicates, at best, a minor contribution of collagen to the stability of the platelet aggregate. These results show that Syk regulates thrombus stability in the absence of fibrin in human platelets under flow and provide evidence that this involves pathways additional to activation of GPVI by collagen.


Asunto(s)
Resistencia al Corte , Quinasa Syk/metabolismo , Trombosis/enzimología , Colágeno/metabolismo , Humanos , Fosforilación , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria , Glicoproteínas de Membrana Plaquetaria/metabolismo , Anticuerpos de Dominio Único/metabolismo , Quinasa Syk/antagonistas & inhibidores , Temperatura , Trombina/farmacología
14.
Am J Physiol Cell Physiol ; 322(3): C370-C381, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35080922

RESUMEN

Cannabis usage has steadily increased as acceptance is growing for both medical and recreational reasons. Medical cannabis is administered for treatment of chronic pain based on the premise that the endocannabinoid system signals desensitize pain sensor neurons and produce anti-inflammatory effects. The major psychoactive ingredient of cannabis is Δ9-tetrahydrocannabinol (THC) that signals mainly through cannabinoid receptor-1 (CBr), which is also present on nonneuron cells including blood platelets of the circulatory system. In vitro, CBr-mediated signaling has been shown to acutely inhibit platelet activation downstream of the platelet collagen receptor glycoprotein (GP)VI. The systemic effects of chronic THC administration on platelet activity and function remain unclear. This study investigates the effects of chronic THC administration on platelet function using a nonhuman primate (NHP) model. Our results show that female and male NHPs consuming a daily THC edible had reduced platelet adhesion, aggregation, and granule secretion in response to select platelet agonists. Furthermore, a change in bioactive lipids (oxylipins) was observed in the female cohort after THC administration. These results indicate that chronic THC edible administration desensitized platelet activity and function in response to GPVI- and G-protein coupled receptor-based activation by interfering with primary and secondary feedback signaling pathways. These observations may have important clinical implications for patients who use medical marijuana and for providers caring for these patients.


Asunto(s)
Plaquetas/efectos de los fármacos , Agonistas de Receptores de Cannabinoides/administración & dosificación , Dronabinol/administración & dosificación , Marihuana Medicinal/administración & dosificación , Administración Oral , Animales , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/metabolismo , Femenino , Macaca mulatta , Masculino , Oxilipinas/sangre , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Vesículas Secretoras/efectos de los fármacos , Vesículas Secretoras/metabolismo , Transducción de Señal , Tromboxanos/sangre , Factores de Tiempo
15.
Platelets ; 33(3): 404-415, 2022 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-34097573

RESUMEN

Several Janus kinase (JAK) inhibitors (jakinibs) have recently been approved to treat inflammatory, autoimmune and hematological conditions. Despite emerging roles for JAKs and downstream signal transducer and activator of transcription (STAT) proteins in platelets, it remains unknown whether jakinibs affect platelet function. Here, we profile platelet biochemical and physiological responses in vitro in the presence of five different clinically relevant jakinibs, including ruxolitinib, upadacitinib, oclacitinib, baricitinib and tofacitinib. Flow cytometry, microscopy and other assays found that potent JAK1/2 inhibitors baricitinib and ruxolitinib reduced platelet adhesion to collagen, as well as platelet aggregation, secretion and integrin αIIbß3 activation in response to the glycoprotein VI (GPVI) agonist collagen-related peptide (CRP-XL). Western blot analysis demonstrated that jakinibs reduced Akt phosphorylation and activation following GPVI activation, where ruxolitinib and baricitinib prevented DAPP1 phosphorylation. In contrast, jakinibs had no effects on platelet responses to thrombin. Inhibitors of GPVI and JAK signaling also abrogated platelet STAT5 phosphorylation following CRP-XL stimulation. Additional pharmacologic experiments supported roles for STAT5 in platelet secretion, integrin activation and cytoskeletal responses. Together, our results demonstrate that ruxolitinib and baricitinib have inhibitory effects on platelet function in vitro and support roles for JAK/STAT5 pathways in GPVI/ITAM mediated platelet function.


Asunto(s)
Azetidinas/uso terapéutico , Plaquetas/metabolismo , Inhibidores de las Cinasas Janus/uso terapéutico , Nitrilos/uso terapéutico , Activación Plaquetaria/efectos de los fármacos , Adhesividad Plaquetaria/efectos de los fármacos , Glicoproteínas de Membrana Plaquetaria/efectos de los fármacos , Purinas/uso terapéutico , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Sulfonamidas/uso terapéutico , Azetidinas/farmacología , Humanos , Inhibidores de las Cinasas Janus/farmacología , Nitrilos/farmacología , Glicoproteínas de Membrana Plaquetaria/metabolismo , Purinas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Sulfonamidas/farmacología
16.
Mol Med ; 27(1): 149, 2021 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-34837956

RESUMEN

BACKGROUND: Thrombocytopenia is one of the most common hematological disease that can be life-threatening caused by bleeding complications. However, the treatment options for thrombocytopenia remain limited. METHODS: In this study, giemsa staining, phalloidin staining, immunofluorescence and flow cytometry were used to identify the effects of 3,3'-di-O-methylellagic acid 4'-glucoside (DMAG), a natural ellagic acid derived from Sanguisorba officinalis L. (SOL) on megakaryocyte differentiation in HEL cells. Then, thrombocytopenia mice model was constructed by X-ray irradiation to evaluate the therapeutic action of DMAG on thrombocytopenia. Furthermore, the effects of DMAG on platelet function were evaluated by tail bleeding time, platelet aggregation and platelet adhesion assays. Next, network pharmacology approaches were carried out to identify the targets of DMAG. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed to elucidate the underling mechanism of DMAG against thrombocytopenia. Finally, molecular docking simulation, molecular dynamics simulation and western blot analysis were used to explore the relationship between DAMG with its targets. RESULTS: DMAG significantly promoted megakaryocyte differentiation of HEL cells. DMAG administration accelerated platelet recovery and megakaryopoiesis, shortened tail bleeding time, strengthened platelet aggregation and adhesion in thrombocytopenia mice. Network pharmacology revealed that ITGA2B, ITGB3, VWF, PLEK, TLR2, BCL2, BCL2L1 and TNF were the core targets of DMAG. GO and KEGG pathway enrichment analyses suggested that the core targets of DMAG were enriched in PI3K-Akt signaling pathway, hematopoietic cell lineage, ECM-receptor interaction and platelet activation. Molecular docking simulation and molecular dynamics simulation further indicated that ITGA2B, ITGB3, PLEK and TLR2 displayed strong binding ability with DMAG. Finally, western blot analysis evidenced that DMAG up-regulated the expression of ITGA2B, ITGB3, VWF, p-Akt and PLEK. CONCLUSION: DMAG plays a critical role in promoting megakaryocytes differentiation and platelets production and might be a promising medicine for the treatment of thrombocytopenia.


Asunto(s)
Ácido Elágico/análogos & derivados , Ácido Elágico/uso terapéutico , Trombocitopenia/tratamiento farmacológico , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Ácido Elágico/farmacología , Femenino , Humanos , Masculino , Ratones , Simulación del Acoplamiento Molecular , Fosfatidilinositol 3-Quinasas/metabolismo , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Trombocitopenia/metabolismo
17.
Int J Mol Sci ; 22(22)2021 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-34830063

RESUMEN

This study describes a method for the modification of polyurethane small-diameter (5 mm) vascular prostheses obtained with the phase inversion method. The modification process involves two steps: the introduction of a linker (acrylic acid) and a peptide (REDV and YIGSR). FTIR and XPS analysis confirmed the process of chemical modification. The obtained prostheses had a porosity of approx. 60%, Young's Modulus in the range of 9-11 MPa, and a water contact angle around 40°. Endothelial (EC) and smooth muscle (SMC) cell co-culture showed that the surfaces modified with peptides increase the adhesion of ECs. At the same time, SMCs adhesion was low both on unmodified and peptide-modified surfaces. Analysis of blood-materials interaction showed high hemocompatibility of obtained materials. The whole blood clotting time assay showed differences in the amount of free hemoglobin present in blood contacted with different materials. It can be concluded that the peptide coating increased the hemocompatibility of the surface by increasing ECs adhesion and, at the same time, decreasing platelet adhesion. When comparing both types of peptide coatings, more promising results were obtained for the surfaces coated with the YISGR than REDV-coated prostheses.


Asunto(s)
Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Prótesis Vascular , Poliuretanos/química , Poliuretanos/farmacología , Animales , Materiales Biocompatibles/síntesis química , Coagulación Sanguínea/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Ensayo de Materiales , Fenómenos Mecánicos , Ratones , Microscopía Electrónica de Rastreo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Adhesividad Plaquetaria/efectos de los fármacos , Poliuretanos/síntesis química , Porosidad , Propiedades de Superficie
18.
Sci Rep ; 11(1): 22694, 2021 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-34811441

RESUMEN

Increased blood platelet activation plays an important role in cardiovascular diseases (CVDs). Recent experiments indicate that certain fruits and vegetables, including onion, garlic, and beetroot, have anti-platelet potential and therefore may reduce the likelihood of CVDs. While vegetables from the Cucuritaceae family are known to exerting beneficial antioxidant and anti-inflammatory effects, their effects on blood platelet activation are poorly understood. Therefore, the aim of the present study was to determine the effect on platelet adhesion of preparations from selected cucurbits: pumpkin (Cucurbita pepo; fruit without seeds), zucchini (Cucurbita pepo convar. giromontina; fruit with seeds), cucumber (Cucumis sativus; fruit with seeds), white pattypan squash (Cucurbita pepo var. patisoniana; fruit without seeds) and yellow pattypan squash (Cucurbita pepo var. patisoniana, fruit without seeds). It also evaluates the activity of these preparations on enzymatic lipid peroxidation in thrombin-activated washed blood platelets by TBARS assay. The study also determines the anti-platelet properties of these five cucurbit preparations in whole blood by flow cytometry and with the total thrombus-formation analysis system (T-TAS) and evaluates the cytotoxicity of the tested preparations against platelets based on LDH activity. The results indicate that the yellow Cucurbita pepo var. patisoniana preparation demonstrated stronger anti-platelet properties than the other tested preparations, reducing the adhesion of thrombin-activated platelets to collagen/fibrinogen, and inhibiting arachidonic acid metabolism and GPIIb/IIIa expression on 10 µM ADP-activated platelets. None of the preparations was found to cause platelet lysis. Our findings provide new information on the anti-platelet activity of the tested cucurbit preparations and their potential for treating CVDs associated with platelet hyperactivity.


Asunto(s)
Cucumis sativus/química , Cucurbita/química , Frutas/química , Extractos Vegetales/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Semillas/química , Verduras/química , Donantes de Sangre , Plaquetas/metabolismo , Supervivencia Celular/efectos de los fármacos , Voluntarios Sanos , Hemostasis/efectos de los fármacos , Humanos , L-Lactato Deshidrogenasa/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Adhesividad Plaquetaria/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Trombosis/prevención & control
19.
Front Immunol ; 12: 729951, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34527000

RESUMEN

Circulating platelets establish a variety of immunological programs and orchestrate inflammatory responses at the endothelium. Platelets express the innate immunity family of Toll-like receptors (TLRs). While TLR2/TLR1 ligands are known to activate platelets, the effects of TLR2/TLR6 ligands on platelet function remain unclear. Here, we aim to determine whether the TLR2/TLR6 agonists Pam2CSK4 and FSL-1 activate human platelets. In addition, human umbilical vein endothelial cells (HUVECs) and platelets were co-cultured to analyze the role of platelet TLR2/TLR6 on inflammation and adhesion to endothelial cells. Pam2CSK4, but not FSL-1, induced platelet granule secretion and integrin αIIbß3 activation in a concentration-dependent manner. Moreover, Pam2CSK4 promoted platelet aggregation and increased platelet adhesion to collagen-coated surfaces. Mechanistic studies with blocking antibodies and pharmacologic inhibitors demonstrated that the TLR2/Nuclear factor-κB axis, Bruton's-tyrosine kinase, and a secondary ADP feedback loop are involved in Pam2CSK4-induced platelet functional responses. Interestingly, Pam2CSK4 showed cooperation with immunoreceptor tyrosine-based activation motif (ITAM)-mediated signaling to enhance platelet activation. Finally, the presence of platelets increased inflammatory responses in HUVECs treated with Pam2CSK4, and platelets challenged with Pam2CSK4 showed increased adhesion to HUVECs under static and physiologically relevant flow conditions. Herein, we define a functional role for platelet TLR2-mediated signaling, which may represent a druggable target to dampen excessive platelet activation in thrombo-inflammatory diseases.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/metabolismo , Plaquetas/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , FN-kappa B/metabolismo , Oligopéptidos/farmacología , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 9/agonistas , Adenosina Difosfato/metabolismo , Plaquetas/enzimología , Células Cultivadas , Diglicéridos/farmacología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Transducción de Señal , Receptor Toll-Like 2/metabolismo
20.
ACS Appl Mater Interfaces ; 13(33): 38947-38958, 2021 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-34433245

RESUMEN

Although dressing blood-contacting devices with robust and synergistic antibacterial and antithrombus properties has been explored for several decades, it still remains a great challenge. In order to endow materials with remarkable antibacterial and antithrombus abilities, a stable and antifouling hydrogel coating was developed via surface-initiated polymerization of sulfobetaine methacrylate and acrylic acid on a polymeric substrate followed by embedding of antimicrobial peptides (AMPs), including WR (sequence: WRWRWR-NH2) or Bac2A (sequence: RLARIVVIRVAR-NH2) AMPs. The chemical composition of the AMP-embedded hydrogel coating was determined through XPS, zeta potential, and SEM-EDS measurements. The AMP-embedded antifouling hydrogel coating showed not only good hemocompatibility but also excellent bactericidal and antiadhesion properties against Gram-positive and Gram-negative bacteria. Moreover, the hydrogel coating could protect the AMPs with long-term bioactivity and cover the positive charge of the dotted distributed AMPs, which in turn well retained the hemocompatibility and antifouling capacity of the bulk hydrogels. Furthermore, the microbiological results of animal experiments also verified the anti-infection performance in vivo. Histological and immunological data further indicated that the hydrogel coating had an excellent anti-inflammatory function. Therefore, the present study might provide a promising approach to prevent bacterial infections and thrombosis in clinical applications of blood-contacting devices and related implants.


Asunto(s)
Antibacterianos/química , Materiales Biocompatibles Revestidos/química , Fibrinolíticos/química , Hidrogeles/química , Proteínas Citotóxicas Formadoras de Poros/química , Resinas Acrílicas/química , Antibacterianos/farmacología , Vendajes , Sangre/metabolismo , Supervivencia Celular , Materiales Biocompatibles Revestidos/metabolismo , Eritrocitos , Fibrinolíticos/farmacología , Hemólisis , Humanos , Hidrogeles/metabolismo , Metacrilatos/química , Adhesividad Plaquetaria/efectos de los fármacos , Polimerizacion , Proteínas Citotóxicas Formadoras de Poros/farmacología , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...