Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 9.093
Filtrar
1.
Sci Rep ; 14(1): 14109, 2024 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-38898080

RESUMO

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.


Assuntos
Plaquetas , Agregação Plaquetária , Trombocitopenia , Agregação Plaquetária/efeitos dos fármacos , Humanos , Trombocitopenia/induzido quimicamente , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Colágeno/metabolismo , Colágeno/farmacologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Adesividade Plaquetária/efeitos dos fármacos , Quinase Syk/metabolismo , Quinase Syk/antagonistas & inibidores , Plasma Rico em Plaquetas/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Sistemas Microfisiológicos
2.
ACS Appl Mater Interfaces ; 16(22): 28134-28146, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38768602

RESUMO

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.


Assuntos
Materiais Biocompatíveis , Quitosana , Hidrogéis , Álcool de Polivinil , Hidrogéis/química , Hidrogéis/farmacologia , Hidrogéis/síntese química , Quitosana/química , Quitosana/análogos & derivados , Quitosana/farmacologia , Humanos , Álcool de Polivinil/química , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Materiais Biocompatíveis/síntese química , Prótese Vascular , Teste de Materiais , beta-Ciclodextrinas/química , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Animais , Adesividade Plaquetária/efeitos dos fármacos , Reagentes de Ligações Cruzadas/química
3.
Int J Mol Sci ; 25(9)2024 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-38732019

RESUMO

Thrombosis is the pathological clot formation under abnormal hemodynamic conditions, which can result in vascular obstruction, causing ischemic strokes and myocardial infarction. Thrombus growth under moderate to low shear (<1000 s-1) relies on platelet activation and coagulation. Thrombosis at elevated high shear rates (>10,000 s-1) is predominantly driven by unactivated platelet binding and aggregating mediated by von Willebrand factor (VWF), while platelet activation and coagulation are secondary in supporting and reinforcing the thrombus. Given the molecular and cellular level information it can access, multiscale computational modeling informed by biology can provide new pathophysiological mechanisms that are otherwise not accessible experimentally, holding promise for novel first-principle-based therapeutics. In this review, we summarize the key aspects of platelet biorheology and mechanobiology, focusing on the molecular and cellular scale events and how they build up to thrombosis through platelet adhesion and aggregation in the presence or absence of platelet activation. In particular, we highlight recent advancements in multiscale modeling of platelet biorheology and mechanobiology and how they can lead to the better prediction and quantification of thrombus formation, exemplifying the exciting paradigm of digital medicine.


Assuntos
Plaquetas , Hemostasia , Trombose , Humanos , Trombose/metabolismo , Plaquetas/metabolismo , Hemostasia/fisiologia , Ativação Plaquetária , Animais , Adesividade Plaquetária , Agregação Plaquetária
4.
Colloids Surf B Biointerfaces ; 239: 113927, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38714078

RESUMO

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.


Assuntos
Coagulação Sanguínea , Nanocompostos , Dióxido de Silício , Zinco , Nanocompostos/química , Coagulação Sanguínea/efeitos dos fármacos , Zinco/química , Zinco/farmacologia , Dióxido de Silício/química , Humanos , Tempo de Tromboplastina Parcial , Tempo de Protrombina , Adesividade Plaquetária/efeitos dos fármacos , Animais , Tamanho da Partícula , Produtos de Degradação da Fibrina e do Fibrinogênio/metabolismo , Hemostáticos/química , Hemostáticos/farmacologia
5.
Int J Biol Macromol ; 271(Pt 2): 132487, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38768910

RESUMO

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.


Assuntos
Ligas , Quitosana , Materiais Revestidos Biocompatíveis , Grafite , Magnésio , Grafite/química , Quitosana/química , Quitosana/análogos & derivados , Quitosana/farmacologia , Magnésio/química , Magnésio/farmacologia , Ligas/química , Ligas/farmacologia , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia , Humanos , Monóxido de Carbono/química , Monóxido de Carbono/farmacologia , Stents , Hemólise/efeitos dos fármacos , Adesividade Plaquetária/efeitos dos fármacos , Corrosão , Adesão Celular/efeitos dos fármacos , Teste de Materiais , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Animais , Células Endoteliais/efeitos dos fármacos
6.
Int J Biol Macromol ; 272(Pt 2): 132747, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38821301

RESUMO

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.


Assuntos
Ligas , Adesão Celular , Magnésio , Teste de Materiais , Peptídeos , Polissacarídeos , Ligas/química , Ligas/farmacologia , Polissacarídeos/química , Polissacarídeos/farmacologia , Humanos , Peptídeos/química , Peptídeos/farmacologia , Magnésio/química , Adesão Celular/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Corrosão , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia , Adesividade Plaquetária/efeitos dos fármacos , Camundongos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Propriedades de Superfície , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Organismos Aquáticos/química , Indóis , Polímeros
7.
Int J Biol Macromol ; 270(Pt 2): 132181, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38740155

RESUMO

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.


Assuntos
Prótese Vascular , Celulose , Heparina , Células Endoteliais da Veia Umbilical Humana , Celulose/química , Heparina/química , Heparina/farmacologia , Humanos , Adesividade Plaquetária/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Polímeros/química , Polímeros/farmacologia , Indóis/química , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia
8.
Int J Biol Macromol ; 269(Pt 1): 132040, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38702003

RESUMO

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.


Assuntos
Coagulação Sanguínea , Colágeno , Adesividade Plaquetária , Animais , Adesividade Plaquetária/efeitos dos fármacos , Coagulação Sanguínea/efeitos dos fármacos , Colágeno/química , Engenharia Tecidual/métodos , Teste de Materiais , Liofilização , Prótese Vascular , Alicerces Teciduais/química , Plaquetas/metabolismo , Plaquetas/química , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Artérias Carótidas/efeitos dos fármacos , Humanos , Etanol/química
9.
Cancer Lett ; 591: 216902, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38641310

RESUMO

Platelets have received growing attention for their roles in hematogenous tumor metastasis. However, the tumor-platelet interaction in osteosarcoma (OS) remains poorly understood. Here, using platelet-specific focal adhesion kinase (FAK)-deficient mice, we uncover a FAK-dependent F3/TGF-ß positive feedback loop in OS. Disruption of the feedback loop by inhibition of F3, TGF-ß, or FAK significantly suppresses OS progression. We demonstrate that OS F3 initiated the feedback loop by increasing platelet TGF-ß secretion, and platelet-derived TGF-ß promoted OS F3 expression in turn and modulated OS EMT process. Immunofluorescence results indicate platelet infiltration in OS niche and we verified it was mediated by platelet FAK. In addition, platelet FAK was proved to mediate platelet adhesion to OS cells, which was vital for the initiation of F3/TGF-ß feedback loop. Collectively, these findings provide a rationale for novel therapeutic strategies targeting tumor-platelet interplay in metastatic OS.


Assuntos
Plaquetas , Neoplasias Ósseas , Transição Epitelial-Mesenquimal , Osteossarcoma , Fator de Crescimento Transformador beta , Osteossarcoma/patologia , Osteossarcoma/metabolismo , Osteossarcoma/genética , Animais , Plaquetas/metabolismo , Plaquetas/patologia , Fator de Crescimento Transformador beta/metabolismo , Neoplasias Ósseas/patologia , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/genética , Humanos , Linhagem Celular Tumoral , Quinase 1 de Adesão Focal/metabolismo , Quinase 1 de Adesão Focal/genética , Retroalimentação Fisiológica , Camundongos , Camundongos Knockout , Progressão da Doença , Transdução de Sinais , Adesividade Plaquetária
10.
Acta Biomater ; 180: 372-382, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38614415

RESUMO

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.


Assuntos
Catéteres , Óxido Nítrico , S-Nitroso-N-Acetilpenicilamina , Animais , Coelhos , Óxido Nítrico/metabolismo , Humanos , S-Nitroso-N-Acetilpenicilamina/farmacologia , S-Nitroso-N-Acetilpenicilamina/química , Trombose/patologia , Teste de Materiais , Linhagem Celular , Adesividade Plaquetária/efeitos dos fármacos , Modelos Animais de Doenças
11.
Colloids Surf B Biointerfaces ; 238: 113908, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38677153

RESUMO

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.


Assuntos
Materiais Revestidos Biocompatíveis , Stents , Humanos , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia , Vasos Coronários/efeitos dos fármacos , Adesividade Plaquetária/efeitos dos fármacos , Anticoagulantes/farmacologia , Anticoagulantes/química , Propriedades de Superfície , Proliferação de Células/efeitos dos fármacos , Aço Inoxidável/química , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/citologia , Animais , Levodopa/química , Levodopa/farmacologia
12.
Langmuir ; 40(18): 9661-9668, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38664943

RESUMO

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.


Assuntos
Colágeno , Peptídeos , Colágeno/química , Peptídeos/química , Peptídeos/farmacologia , Fibrinolíticos/química , Fibrinolíticos/farmacologia , Sequência de Aminoácidos , Desenho de Fármacos , Humanos , Redes Neurais de Computação , Simulação de Dinâmica Molecular , Adesividade Plaquetária/efeitos dos fármacos
13.
Phytomedicine ; 128: 155420, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38547619

RESUMO

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.


Assuntos
Neoplasias do Colo , Camundongos Endogâmicos C57BL , Microambiente Tumoral , Animais , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Microambiente Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral , Plaquetas/efeitos dos fármacos , Camundongos , Agregação Plaquetária/efeitos dos fármacos , Adesividade Plaquetária/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Humanos , Extratos Vegetais/farmacologia , Metástase Neoplásica
14.
Biorheology ; 59(3-4): 63-80, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38461497

RESUMO

Leukocytes and platelets must adhere to the wall of blood vessels to carry out their protective functions in inflammation and haemostasis. Recruitment is critically dependent on rheological variables (wall shear rate and stress, red cell aggregation and haematocrit) which affect delivery to the vessel wall as well as velocities and forces experienced there. Leukocyte recruitment is efficient only up to wall shear rates of about 300 s-1 and usually restricted to low-shear post-capillary venules in inflammation. Being smaller, platelets experience lower velocities and shear forces adjacent to the wall and can adhere at much higher shear rates for haemostasis in arteries. In addition, we found quite different effects of variations in haematocrit or red cell aggregation on attachment of neutrophils or platelets, which also assist their separate recruitment in venules or arteries. However, it has become increasingly evident that inflammatory and thrombotic responses may occur together, with platelets promoting the adhesion and activation of neutrophils and monocytes. Indeed, it is 30 years since we demonstrated that platelets could cause neutrophils to aggregate in suspension and, when attached to a surface, could support selectin-mediated rolling of all leukocytes. Thrombin-activated platelets could further induce neutrophil activation and immobilisation. In some conditions, platelets could bind to intact endothelial monolayers and capture neutrophils or monocytes. Subsequently, we found that extracellular vesicles released by activated platelets (PEV) fulfilled similar functions when deposited on surfaces or bound to endothelial cells. In murine models, platelets or PEV could act as bridges for monocytes in inflamed vessels. Thus, leukocytes and platelets are rheologically adapted for their separate functions, while novel thrombo-inflammatory pathways using platelets or PEV may underlie pathogenic leukocyte recruitment.


Assuntos
Agregação Eritrocítica , Adesividade Plaquetária , Humanos , Animais , Camundongos , Adesividade Plaquetária/fisiologia , Células Endoteliais , Plaquetas/fisiologia , Leucócitos/fisiologia , Neutrófilos , Reologia , Inflamação/metabolismo , Adesão Celular , Selectina-P/metabolismo
15.
Colloids Surf B Biointerfaces ; 237: 113866, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38520952

RESUMO

The inhibition of platelet adhesion to collagen in exposed vessels represents an innovative approach to the treatment of atherosclerosis and thrombosis. This study aimed to engineer peptide-based nanoparticles that prevent platelet binding to subendothelial collagen by engaging with collagen with high affinity. We examined the interactions between integrin α2/ glycoprotein VI/ von Willebrand factor A3 domain and collagen, as well as between the synthesized peptide nanoparticles and collagen, utilizing molecular dynamics simulations and empirical assays. Our findings indicated that the bond between von Willebrand factor and collagen was more robust. Specifically, the sequences SITTIDV, VDVMQRE, and YLTSEMH in von Willebrand factor were identified as essential for its attachment to collagen. Based on these sequences, three peptide nanoparticles were synthesized (BPa: Capric-GNNQQNYK-SITTIDV, BPb: Capric-GNNQQNYK-VDVMQRE, BPc: Capric-GNNQQNYK-YLTSEMH), each displaying significant affinity towards collagen. Of these, the BPa nanoparticles exhibited the most potent interaction with collagen, leading to a 75% reduction in platelet adhesion.


Assuntos
Adesividade Plaquetária , Fator de von Willebrand , Fator de von Willebrand/metabolismo , Colágeno/química , Peptídeos/farmacologia , Peptídeos/metabolismo , Plaquetas/metabolismo
16.
ACS Biomater Sci Eng ; 10(3): 1403-1417, 2024 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-38308598

RESUMO

Assessing blood compatibility is crucial before in vivo procedures and is considered more reliable than many in vitro tests. This study examines the physiochemical properties and blood compatibility of bioactive powders ((0.5-2 wt % carbon nanotube (CNT)/alumina)-20 wt %)) produced through a heterocoagulation colloidal technique followed by ball milling with hydroxyapatite (HAp). The 1 wt % CNT composite demonstrated a surface charge ∼5 times higher than HAp at pH 7.4, with a value of -11 mV compared to -2 mV. This increase in electrostatic charge is desirable for achieving hemocompatibility, as evidenced by a range of blood compatibility assessments, including hemolysis, blood clotting, platelet adhesion, platelet activation, and coagulation assays (prothrombin time (PT) and activated partial thrombin time (aPTT)). The 1 wt % CNT composite exhibited hemolysis ranging from 2 to 7%, indicating its hemocompatibility. In the blood clot investigation, the absorbance values for 1-2 wt % CNT samples were 0.927 ± 0.038 and 1.184 ± 0.128, respectively, indicating their nonthrombogenicity. Additionally, the percentage of platelet adhered on the 1 wt % CNT sample (∼5.67%) showed a ∼2.5-fold decrement compared to the clinically used negative control, polypropylene (∼13.73%). The PT and aPTT experiments showed no difference in the coagulation time for CNT samples even at higher concentrations, unlike HAC2 (80 mg). In conclusion, the 1 wt % CNT sample was nontoxic to human blood, making it more hemocompatible, nonhemolytic, and nonthrombogenic than other samples. This reliable study reduces the need for additional in vitro and in vivo studies before clinical trials, saving time and cost.


Assuntos
Durapatita , Nanotubos de Carbono , Humanos , Durapatita/química , Durapatita/farmacologia , Nanotubos de Carbono/toxicidade , Nanotubos de Carbono/química , Hemólise , Plaquetas , Adesividade Plaquetária
17.
Platelets ; 35(1): 2316743, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38390892

RESUMO

Microfluidic technology has emerged as a powerful tool in studying arterial thrombosis, allowing researchers to construct artificial blood vessels and replicate the hemodynamics of blood flow. This technology has led to significant advancements in understanding thrombosis and platelet adhesion and aggregation. Microfluidic models have various types and functions, and by studying the fabrication methods and working principles of microfluidic chips, applicable methods can be selected according to specific needs. The rapid development of microfluidic integrated system and modular microfluidic system makes arterial thrombosis research more diversified and automated, but its standardization still needs to be solved urgently. One key advantage of microfluidic technology is the ability to precisely control fluid flow in microchannels and to analyze platelet behavior under different shear forces and flow rates. This allows researchers to study the physiological and pathological processes of blood flow, shedding light on the underlying mechanisms of arterial thrombosis. In conclusion, microfluidic technology has revolutionized the study of arterial thrombosis by enabling the construction of artificial blood vessels and accurately reproducing hemodynamics. In the future, microfluidics will place greater emphasis on versatility and automation, holding great promise for advancing antithrombotic therapeutic and prophylactic measures.


What is the context? To study the mechanism of arterial thrombosis, including the platelet adhesion and aggregation behavior and the coagulation process.Microfluidic technology is commonly used to study thrombosis. Microfluidic technology can simulate the real physiological environment on the microscopic scale in vitro, with high throughput, low cost, and fast speed.As an innovative experimental platform, microfluidic technology has made remarkable progress and has found applications in the fields of biology and medicine.What is new? This review summarizes the different fabrication methods of microfluidics and compares the advantages and disadvantages of these methods. Recent developments in microfluidic integrated systems and modular microfluidic systems have led to more diversified and automated microfluidic chips in the future.The different types and functions of microfluidic models are summarized. Platelet adhesion aggregation and coagulation processes, as well as arterial thrombus-related shear force changes and mechanical behaviors, were investigated by constructing artificial blood vessels and reproducing hemodynamics.Microfluidics can provide a basis for the development of personalized thrombosis treatment strategies. By analyzing the mechanism of action of existing drugs, using microfluidic technology for high-throughput screening of drugs and evaluating drug efficacy, more drug therapy possibilities can be developed.What is the impact?This review utilizes microfluidics to further advance the study of arterial thrombosis, and microfluidics is also expected to play a greater role in the biomedical field in the future.


Assuntos
Substitutos Sanguíneos , Trombose , Humanos , Microfluídica/métodos , Plaquetas/patologia , Trombose/patologia , Adesividade Plaquetária
18.
Blood ; 143(19): 1992-2004, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38290109

RESUMO

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.


Assuntos
Anticorpos de Domínio Único , Fator de von Willebrand , Fator de von Willebrand/metabolismo , Fator de von Willebrand/química , Humanos , Anticorpos de Domínio Único/farmacologia , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/metabolismo , Agregação Plaquetária/efeitos dos fármacos , Conformação Proteica , Domínios Proteicos , Ligação Proteica , Adesividade Plaquetária/efeitos dos fármacos , Cristalografia por Raios X , Animais , Plaquetas/metabolismo
19.
Biomech Model Mechanobiol ; 23(3): 737-756, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38217745

RESUMO

An excessive von Willebrand factor (VWF) secretion, coupled with a moderate to severe deficiency of ADAMTS13 activity, serves as a linking mechanism between inflammation to thrombosis. The former facilitates platelet adhesion to the vessel wall and the latter is required to cleave VWF multimers. As a result, the ultra-large VWF (UL-VWF) multimers released by Weibel-Palade bodies remain uncleaved. In this study, using a computational model based on first principles, we quantitatively show how the uncleaved UL-VWF multimers interact with the blood cells to initiate microthrombosis. We observed that platelets first adhere to unfolded and stretched uncleaved UL-VWF multimers anchored to the microvessel wall. By the end of this initial adhesion phase, the UL-VWF multimers and platelets make a mesh-like trap in which the red blood cells increasingly accumulate to initiate a gradually growing microthrombosis. Although high-shear rate and blood flow velocity are required to activate platelets and unfold the UL-VWFs, during the initial adhesion phase, the blood velocity drastically drops after thrombosis, and as a result, the wall shear stress is elevated near UL-VWF roots, and the pressure drops up to 6 times of the healthy condition. As the time passes, these trends progressively continue until the microthrombosis fully develops and the effective size of the microthrombosis and these flow quantities remain almost constant. Our findings quantitatively demonstrate the potential role of UL-VWF in coagulopathy.


Assuntos
Transtornos da Coagulação Sanguínea , Fator de von Willebrand , Humanos , Transtornos da Coagulação Sanguínea/sangue , Transtornos da Coagulação Sanguínea/metabolismo , Plaquetas/metabolismo , Simulação por Computador , Modelos Biológicos , Análise Numérica Assistida por Computador , Adesividade Plaquetária , Multimerização Proteica , Estresse Mecânico , Trombose/metabolismo , Fator de von Willebrand/metabolismo
20.
Microvasc Res ; 151: 104613, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37793562

RESUMO

Thrombosis can lead to significant mortality and morbidity. Both platelets and vascular endothelial cells play significant roles in thrombosis. Platelets' response to blood flow-induced shear stress can vary greatly depending on shear stress magnitude, pattern and shear exposure time. Endothelial cells are also sensitive to the biomechanical environment. Endothelial cell activation and dysfunction can occur under low oscillatory shear stress and low tensile strain. Platelet and endothelial cell interaction can also be affected by mechanical conditions. The goal of this study was to investigate how blood flow-induced shear stress, vascular wall tensile strain, platelet-endothelial cell stress history, and platelet-endothelial cell interaction affect platelet thrombogenicity. Platelets and human coronary artery endothelial cells were pretreated with physiological and pathological shear stress and/or tensile strain separately. The pretreated cells were then put together and exposed to pulsatile shear stress and cyclic tensile strain simultaneously in a shearing-stretching device. Following treatment, platelet thrombin generation rate, platelet and endothelial cell activation, and platelet adhesion to endothelial cells was measured. The results demonstrated that shear stress pretreatment of endothelial cells and platelets caused a significant increase in platelet thrombin generation rate, cell surface phosphatidylserine expression, and adhesion to endothelial cells. Shear stress pretreatment of platelets and endothelial cells attenuated endothelial cell ICAM-1 expression under stenosis conditions, as well as vWF expression under recirculation conditions. These results indicate that platelets are sensitized by prior shearing, while in comparison, the interaction with shear stress-pretreated platelets may reduce endothelial cell sensitivity to pathological shear stress and tensile strain.


Assuntos
Células Endoteliais , Trombose , Humanos , Células Endoteliais/metabolismo , Trombina/metabolismo , Trombina/farmacologia , Plaquetas/metabolismo , Adesividade Plaquetária , Trombose/etiologia , Estresse Mecânico , Ativação Plaquetária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA