Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.143
Filter
1.
Int J Mol Sci ; 25(9)2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38732019

ABSTRACT

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.


Subject(s)
Blood Platelets , Hemostasis , Thrombosis , Humans , Thrombosis/metabolism , Blood Platelets/metabolism , Hemostasis/physiology , Platelet Activation , Animals , Platelet Adhesiveness , Platelet Aggregation
2.
Cancer Lett ; 591: 216902, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38641310

ABSTRACT

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.


Subject(s)
Blood Platelets , Bone Neoplasms , Epithelial-Mesenchymal Transition , Osteosarcoma , Transforming Growth Factor beta , Osteosarcoma/pathology , Osteosarcoma/metabolism , Osteosarcoma/genetics , Animals , Blood Platelets/metabolism , Blood Platelets/pathology , Transforming Growth Factor beta/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Bone Neoplasms/genetics , Humans , Cell Line, Tumor , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Feedback, Physiological , Mice , Mice, Knockout , Disease Progression , Signal Transduction , Platelet Adhesiveness
3.
Colloids Surf B Biointerfaces ; 238: 113908, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677153

ABSTRACT

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


Subject(s)
Coated Materials, Biocompatible , Stents , Humans , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Coronary Vessels/drug effects , Platelet Adhesiveness/drug effects , Anticoagulants/pharmacology , Anticoagulants/chemistry , Surface Properties , Cell Proliferation/drug effects , Stainless Steel/chemistry , Blood Platelets/drug effects , Blood Platelets/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/cytology , Animals , Levodopa/chemistry , Levodopa/pharmacology
4.
Acta Biomater ; 180: 372-382, 2024 May.
Article in English | MEDLINE | ID: mdl-38614415

ABSTRACT

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


Subject(s)
Catheters , Nitric Oxide , S-Nitroso-N-Acetylpenicillamine , Animals , Rabbits , Nitric Oxide/metabolism , Humans , S-Nitroso-N-Acetylpenicillamine/pharmacology , S-Nitroso-N-Acetylpenicillamine/chemistry , Thrombosis/pathology , Materials Testing , Cell Line , Platelet Adhesiveness/drug effects , Disease Models, Animal
5.
Langmuir ; 40(18): 9661-9668, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38664943

ABSTRACT

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


Subject(s)
Collagen , Peptides , Collagen/chemistry , Peptides/chemistry , Peptides/pharmacology , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/pharmacology , Amino Acid Sequence , Drug Design , Humans , Neural Networks, Computer , Molecular Dynamics Simulation , Platelet Adhesiveness/drug effects
6.
Colloids Surf B Biointerfaces ; 237: 113866, 2024 May.
Article in English | MEDLINE | ID: mdl-38520952

ABSTRACT

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.


Subject(s)
Platelet Adhesiveness , von Willebrand Factor , von Willebrand Factor/metabolism , Collagen/chemistry , Peptides/pharmacology , Peptides/metabolism , Blood Platelets/metabolism
7.
Phytomedicine ; 128: 155420, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38547619

ABSTRACT

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


Subject(s)
Colonic Neoplasms , Mice, Inbred C57BL , Tumor Microenvironment , Animals , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Tumor Microenvironment/drug effects , Cell Line, Tumor , Blood Platelets/drug effects , Mice , Platelet Aggregation/drug effects , Platelet Adhesiveness/drug effects , Epithelial-Mesenchymal Transition/drug effects , Humans , Plant Extracts/pharmacology , Neoplasm Metastasis
8.
Biorheology ; 59(3-4): 63-80, 2024.
Article in English | MEDLINE | ID: mdl-38461497

ABSTRACT

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.


Subject(s)
Erythrocyte Aggregation , Platelet Adhesiveness , Humans , Animals , Mice , Platelet Adhesiveness/physiology , Endothelial Cells , Blood Platelets/physiology , Leukocytes/physiology , Neutrophils , Rheology , Inflammation/metabolism , Cell Adhesion , P-Selectin/metabolism
9.
Platelets ; 35(1): 2316743, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38390892

ABSTRACT

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.


Subject(s)
Blood Substitutes , Thrombosis , Humans , Microfluidics/methods , Blood Platelets/pathology , Thrombosis/pathology , Platelet Adhesiveness
10.
ACS Biomater Sci Eng ; 10(3): 1403-1417, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38308598

ABSTRACT

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.


Subject(s)
Durapatite , Nanotubes, Carbon , Humans , Durapatite/chemistry , Durapatite/pharmacology , Nanotubes, Carbon/toxicity , Nanotubes, Carbon/chemistry , Hemolysis , Blood Platelets , Platelet Adhesiveness
11.
Biomech Model Mechanobiol ; 23(3): 737-756, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38217745

ABSTRACT

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.


Subject(s)
von Willebrand Factor , von Willebrand Factor/metabolism , Humans , Protein Multimerization , Blood Platelets/metabolism , Platelet Adhesiveness , Thrombosis/metabolism , Stress, Mechanical , Blood Coagulation Disorders/blood , Blood Coagulation Disorders/metabolism , Computer Simulation , Numerical Analysis, Computer-Assisted , Models, Biological
12.
Blood ; 143(19): 1992-2004, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38290109

ABSTRACT

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


Subject(s)
Single-Domain Antibodies , von Willebrand Factor , von Willebrand Factor/metabolism , von Willebrand Factor/chemistry , Humans , Single-Domain Antibodies/pharmacology , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/metabolism , Platelet Aggregation/drug effects , Protein Conformation , Protein Domains , Protein Binding , Platelet Adhesiveness/drug effects , Crystallography, X-Ray , Animals , Blood Platelets/metabolism
13.
Microvasc Res ; 151: 104613, 2024 01.
Article in English | MEDLINE | ID: mdl-37793562

ABSTRACT

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.


Subject(s)
Endothelial Cells , Thrombosis , Humans , Endothelial Cells/metabolism , Thrombin/metabolism , Thrombin/pharmacology , Blood Platelets/metabolism , Platelet Adhesiveness , Thrombosis/etiology , Stress, Mechanical , Platelet Activation
14.
Biomater Sci ; 11(23): 7655-7662, 2023 Nov 21.
Article in English | MEDLINE | ID: mdl-37850341

ABSTRACT

Thromboembolism, arising from the utilization of cardiovascular medical devices, remains a prevalent issue entailing substantial morbidity and mortality. Despite the proposal of various surface modification strategies, each approach possesses inherent limitations and drawbacks. Herein, we propose a novel approach for the in situ growth of nanocoatings on various material surfaces through the cooperative assembly of silk fibroin (SF) and lysozyme. The intrinsic in situ growth characteristic enables the nanocoatings to achieve stable and uniform adherence to diverse substrate surfaces, including the inner surface of intravascular catheters, to redefine the surface properties of the material. The features of the hydrophilic and negatively charged nanocoating contribute to its antithrombotic properties, as evidenced by the reduced likelihood of platelet adhesion upon modification of the ultrathin and mechanically robust coating. In vitro assessment confirms a significant reduction in blood clot formation along with the promotion of anticoagulation. Such a SF/Ly nanocoating holds substantial promise as a surface modification strategy to enhance the hemocompatibility of medical devices and other materials that come into contact with blood, particularly in situations where medical-grade materials are temporarily unavailable, thus providing a feasible alternative.


Subject(s)
Thromboembolism , Thrombosis , Humans , Coated Materials, Biocompatible/chemistry , Platelet Adhesiveness , Surface Properties
15.
Int J Mol Sci ; 24(17)2023 Aug 28.
Article in English | MEDLINE | ID: mdl-37686158

ABSTRACT

Proteoglycans form a heterogeneous family of proteins with covalently bound sulfated glycosaminoglycans. The extracellular matrix proteoglycan perlecan has been proposed to bind to the platelet- and megakaryocyte-specific receptor G6bB, co-regulating platelet glycoprotein VI (GPVI) signaling. The derived non-sulfate proteoglycan endorepellin was previously shown to enhance platelet adhesion via the collagen receptor, integrin α2ß1. Here, we compared the roles of perlecan and other matrix proteoglycans in platelet responses and thrombus formation. We used multi-color flow cytometry to measure the degranulation and integrin αIIbß3 activation of washed platelets in response to various proteoglycans and collagen-related peptide (CRP), the GPVI agonist. Perlecan, but not endorepellin, enhanced the CRP-induced activation of platelets in a time- and concentration-dependent manner. Similar to collagen, immobilized perlecan, but not other proteoglycans, supported static platelet adhesion and spreading. In-flowed whole-blood perlecan diminished shear-dependent platelet adhesion, while it enforced GPVI-dependent thrombus formation-to a larger extent than endorepellin-to induce more contracted aggregates of activated platelets. We concluded that the sulfated proteoglycan perlecan enhances GPVI-dependent platelet responses extending to thrombus formation, but it does so at the expense of reduced adhesion of platelets under flow.


Subject(s)
Heparan Sulfate Proteoglycans , Thrombosis , Humans , Extracellular Matrix Proteins , Platelet Adhesiveness
16.
J Thromb Haemost ; 21(12): 3619-3632, 2023 12.
Article in English | MEDLINE | ID: mdl-37678551

ABSTRACT

BACKGROUND: Upon vessel injury, platelets adhere to exposed matrix constituents via specific membrane receptors, including the von Willebrand factor receptor glycoprotein (GP)Ib-IX-V complex and integrins ß1 and ß3. In platelets, the Fes/CIP4-homology Bin-Amphiphysin-Rvs protein PACSIN2 associates with the cytoskeletal and scaffolding protein filamin A (FlnA), linking GPIbα and integrins to the cytoskeleton. OBJECTIVES: Here we investigated the role of PACSIN2 in platelet function. METHODS: Platelet parameters were evaluated in mice lacking PACSIN2 and platelet integrin ß1. RESULTS: Pacsin2-/- mice displayed mild thrombocytopenia, prolonged bleeding time, and delayed thrombus formation in a ferric chloride-mediated carotid artery injury model, which was normalized by injection of control platelets. Pacsin2-/- platelets formed unstable thrombi that embolized abruptly in a laser-induced cremaster muscle injury model. Pacsin2-/- platelets had hyperactive integrin ß1, as evidenced by increased spreading onto surfaces coated with the collagen receptor α2ß1-specific peptide GFOGER and increased binding of the antibody 9EG7 directed against active integrin ß1. By contrast, Pacsin2-/- platelets had normal integrin αIIbß3 function and expressed P-selectin normally following stimulation through the collagen receptor GPVI or with thrombin. Deletion of platelet integrin ß1 in Pacsin2-/- mice normalized platelet count, hemostasis, and thrombus formation. A PACSIN2 peptide mimicking the FlnA-binding site mediated the pull-down of a FlnA rod 2 construct by integrin ß7, a model for integrin ß-subunits. CONCLUSIONS: Pacsin2-/- mice displayed severe thrombus formation defects due to hyperactive platelet integrin ß1. The data suggest that PACSIN2 binding to FlnA negatively regulates platelet integrin ß1 hemostatic function.


Subject(s)
Integrin beta1 , Platelet Activation , Thrombosis , Animals , Mice , Blood Platelets/metabolism , Hemostasis , Hemostatics/metabolism , Integrin beta1/metabolism , Peptides/pharmacology , Platelet Adhesiveness , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoproteins/metabolism , Receptors, Collagen/metabolism , Thrombosis/metabolism
17.
Hamostaseologie ; 43(4): 238-240, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37611603

ABSTRACT

Platelets are key drivers of hemostasis. Low platelet counts, dysfunction in platelet adhesion, and aggregation lead to increased bleeding tendency. Inherited platelet disorders (IPDs) form a highly heterogeneous group of rare diseases with variable bleeding tendency. IPDs may be associated with other signs and symptoms often referred to as "syndromic." The underlying genetic defect may prone patients to develop hematopoietic diseases such as leukemia. Over the last decade, accumulating knowledge in genetics has led to the detection of many "new" platelet disorders. However, still many patients with a well-described platelet dysfunction remain undetected until severe bleeding occurs.


Subject(s)
Blood Platelet Disorders , Illusions , Leukemia , Humans , Blood Platelet Disorders/diagnosis , Blood Platelet Disorders/genetics , Blood Platelet Disorders/therapy , Blood Platelets , Platelet Adhesiveness
18.
Arterioscler Thromb Vasc Biol ; 43(10): 1775-1783, 2023 10.
Article in English | MEDLINE | ID: mdl-37615110

ABSTRACT

Platelet adhesion and activation is fundamental to the formation of a hemostatic response to limit loss of blood and instigate wound repair to seal a site of vascular injury. The process of platelet aggregate formation is supported by the coagulation system driving injury-proximal formation of thrombin, which converts fibrinogen to insoluble fibrin. This highly coordinated series of molecular and membranous events must be routinely achieved in flowing blood, at vascular fluid shear rates that place significant strain on molecular and cellular interactions. Platelets have long been recognized to be able to slow down and adhere to sites of vascular injury and then activate and recruit more platelets that forge and strengthen adhesive ties with the vascular wall under these conditions. It has been a major challenge for the Platelet Research Community to construct experimental conditions that allow precise definition of the molecular steps occurring under flow. This brief review will discuss work to date from our group, as well as others that has furthered our understanding of platelet function in flowing blood.


Subject(s)
Hemostatics , Vascular System Injuries , Humans , Blood Platelets/physiology , Hemostasis , Blood Coagulation , Platelet Adhesiveness
19.
Blood ; 142(13): 1156-1166, 2023 09 28.
Article in English | MEDLINE | ID: mdl-37506337

ABSTRACT

von Willebrand factor (VWF) mediates primary hemostasis and thrombosis in response to hydrodynamic forces. We previously showed that high shear promoted self-association of VWF into hyperadhesive strands, which can be attenuated by high-density lipoprotein (HDL) and apolipoprotein A-I. In this study, we show that low-density lipoprotein (LDL) binds VWF under shear and enhances self-association. Vortexing VWF in tubes resulted in its loss from the solution and deposition onto tube surfaces, which was prevented by HDL. At a stabilizing HDL concentration of 1.2 mg/mL, increasing concentrations of LDL progressively increased VWF loss, the effect correlating with the LDL-to-HDL ratio and not the absolute concentration of the lipoproteins. Similarly, HDL diminished deposition of VWF in a post-in-channel microfluidic device, whereas LDL increased both the rate and extent of strand deposition, with both purified VWF and plasma. Hypercholesterolemic human plasma also displayed accelerated VWF accumulation in the microfluidic device. The initial rate of accumulation correlated linearly with the LDL-to-HDL ratio. In Adamts13-/- and Adamts13-/-LDLR-/- mice, high LDL levels enhanced VWF and platelet adhesion to the myocardial microvasculature, reducing cardiac perfusion, impairing systolic function, and producing early signs of cardiomyopathy. In wild-type mice, high plasma LDL concentrations also increased the size and persistence of VWF-platelet thrombi in ionophore-treated mesenteric microvessels, exceeding the accumulation seen in similarly treated ADAMTS13-deficient mice that did not receive LDL infusion. We propose that targeting the interaction of VWF with itself and with LDL may improve the course of thrombotic microangiopathies, atherosclerosis, and other disorders with defective microvascular circulation.


Subject(s)
Thrombosis , von Willebrand Factor , Mice , Humans , Animals , von Willebrand Factor/metabolism , Lipoproteins, LDL , Thrombosis/metabolism , Hemostasis , Platelet Adhesiveness , ADAMTS13 Protein
20.
Int J Numer Method Biomed Eng ; 39(11): e3747, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37366014

ABSTRACT

A multimeric glycoprotein of blood plasma-Von Willebrand factor (VWF)-mediates platelet adhesion to the fibrillar collagen of the subendothelial matrix if the blood vessel walls are damaged. The adsorption of VWF to collagen is thus essential for the initial stages of platelet hemostasis and thrombosis, as it plays a role of a molecular bridge between the injury and platelet adhesion receptors. Biomechanical complexity and sensitivity to the hydrodynamics are inherent in this system, therefore, modern computational methods supplement experimental studies of biophysical and molecular mechanisms that underlie platelet adhesion and aggregation in the blood flow. In the present paper, we propose a simulation framework for the VWF-mediated platelet adhesion to a plane wall with immobilized binding sites for VWF under the action of shear flow. VWF multimers and platelets are represented in the model by particles connected by elastic bonds and immersed in a viscous continuum fluid. This work complements the scientific field by taking into account the shape of a flattened platelet, but keeping a compromise between the detail of the description and the computational complexity of the model.


Subject(s)
Blood Platelets , von Willebrand Factor , von Willebrand Factor/metabolism , Adsorption , Blood Platelets/metabolism , Platelet Adhesiveness/physiology , Collagen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...