Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.536
Filter
1.
J Extra Corpor Technol ; 56(2): 37-44, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38888546

ABSTRACT

INTRODUCTION: Cardiopulmonary bypass is an essential component of cardiothoracic surgeries. However, significant complications such as systemic inflammatory response syndrome (SIRS) resulting from cardiopulmonary bypass (CPB) are a common occurrence due to contact between circulating blood and foreign surfaces that leads to platelet activation. It is suggested that different available CPB circuit coatings can potentially reduce platelet activation. However, there have been no published evidence-based reports confirming these claims. In addition, there is no well-established protocol for studying platelet activation biomarkers during CPB in vitro in a laboratory setting. METHODS: CPB was simulated in the laboratory using bovine blood in two different types of coated CPB circuits: Trillium® Biosurface by Medtronic, and XcoatingTM Surface by Terumo. Fresh bovine blood samples were collected and circulated through the CPB circuit following the standard protocol used in the operation rooms. Blood samples were then collected at 5 min, 30 min, and 55 min during the circulation. Blood plasmas were separated and subjected to enzyme-linked immunosorbent assay to measure most established platelet activation markers P-selectin, Platelet Factor 4 (PF4), Glycoprotein IIb/IIIa (GPIIb/IIIa), and ß-thromboglobulin (ß-TG) at different time points. RESULTS: The biomarker values at 30 min and 55 min were compared to the base values at 5 min for each type of CPB circuit. The results of the means from all measured biomarkers showed data measurements that indicated no significant variability within each coating. All collected data points fell within ±2 SD of the means, which was considered acceptable variations across technical replicates.  Conclusion: In this study, we were able to establish an in vitro protocol in the laboratory setting that is precise and reliable with minimum intra-variability. This established protocol will allow for future studies in which different coated CPB circuits can be compared for their effectiveness in blocking platelet activation during the CPB.


Subject(s)
Biomarkers , Cardiopulmonary Bypass , Coated Materials, Biocompatible , Platelet Activation , Cardiopulmonary Bypass/instrumentation , Cardiopulmonary Bypass/adverse effects , Platelet Activation/physiology , Animals , Biomarkers/blood , Cattle , Materials Testing/methods
2.
Life Sci ; 351: 122846, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38880165

ABSTRACT

Understanding the mechanisms controlling platelet function is crucial for exploring potential therapeutic targets related to atherothrombotic pathologies and primary hemostasis disorders. Our research, which focuses on the role of platelet mitochondria and Ca2+ fluxes in platelet activation, the formation of the procoagulant phenotype, and thrombosis, has significant implications for the development of new therapeutic strategies. Traditionally, Ca2+-dependent cellular signaling has been recognized as a determinant process throughout the platelet activation, controlled primarily by store-operated Ca2+ entry and the PLC-PKC signaling pathway. However, despite the accumulated knowledge of these regulatory mechanisms, the effectiveness of therapy based on various commonly used antiplatelet drugs (such as acetylsalicylic acid and clopidogrel, among others) has faced challenges due to bleeding risks and reduced efficacy associated with the phenomenon of high platelet reactivity. Recent evidence suggests that platelet mitochondria could play a fundamental role in these aspects through Ca2+-dependent mechanisms linked to apoptosis and forming a procoagulant phenotype. In this context, the present review describes the latest advances regarding the role of platelet mitochondria and Ca2+ fluxes in platelet activation, the formation of the procoagulant phenotype, and thrombosis.


Subject(s)
Aging , Blood Platelets , Calcium , Mitochondria , Platelet Activation , Humans , Mitochondria/metabolism , Platelet Activation/physiology , Calcium/metabolism , Blood Platelets/metabolism , Aging/metabolism , Animals , Thrombosis/metabolism , Calcium Signaling/physiology
3.
Life Sci ; 351: 122848, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38885879

ABSTRACT

AIMS: This research aimed to study the changes in platelet function and their underlying mechanisms in iron deficiency anemia. MAIN METHODS: Initially, we evaluated platelet function in an IDA mice model. Due to the inability to accurately reduce intracellular Fe2+ concentrations, we investigated the impact of Fe2+ on platelet function by introducing varying concentrations of Fe2+. To probe the underlying mechanism, we simultaneously examined the dynamics of calcium in the cytosol, and integrin αIIbß3 activation in Fe2+-treated platelets. Ferroptosis inhibitors Lip-1 and Fer-1 were applied to determine whether ferroptosis was involved in this process. KEY FINDINGS: Our study revealed that platelet function was suppressed in IDA mice. Fe2+ concentration-dependently facilitated platelet activation and function in vitro. Mechanistically, Fe2+ promoted calcium mobilization, integrin αIIbß3 activation, and its downstream outside-in signaling. Additionally, we also demonstrated that ferroptosis might play a role in this process. SIGNIFICANCE: Our data suggest an association between iron and platelet activation, with iron deficiency resulting in impaired platelet function, while high concentrations of Fe2+ contribute to platelet activation and function by promoting calcium mobilization, αIIbß3 activation, and ferroptosis.


Subject(s)
Anemia, Iron-Deficiency , Blood Platelets , Calcium , Ferroptosis , Mice, Inbred C57BL , Platelet Activation , Animals , Mice , Blood Platelets/metabolism , Anemia, Iron-Deficiency/metabolism , Anemia, Iron-Deficiency/blood , Ferroptosis/physiology , Calcium/metabolism , Platelet Activation/physiology , Male , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Iron/metabolism , Disease Models, Animal
4.
J Intern Med ; 296(1): 39-52, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38704820

ABSTRACT

Platelet hyperreactivity and hyperlipidaemia contribute significantly to atherosclerosis. Thus, it is desirable to review the platelet-hyperlipidaemia interplay and its impact on atherogenesis. Native low-density lipoprotein (nLDL) and oxidized LDL (oxLDL) are the key proatherosclerotic components of hyperlipidaemia. nLDL binds to the platelet-specific LDL receptor (LDLR) ApoE-R2', whereas oxLDL binds to the platelet-expressed scavenger receptor CD36, lectin-type oxidized LDLR 1 and scavenger receptor class A 1. Ligation of nLDL/oxLDL induces mild platelet activation and may prime platelets for other platelet agonists. Platelets, in turn, can modulate lipoprotein metabolisms. Platelets contribute to LDL oxidation by enhancing the production of reactive oxygen species and LDLR degradation via proprotein convertase subtilisin/kexin type 9 release. Platelet-released platelet factor 4 and transforming growth factor ß modulate LDL uptake and foam cell formation. Thus, platelet dysfunction and hyperlipidaemia work in concert to aggravate atherogenesis. Hypolipidemic drugs modulate platelet function, whereas antiplatelet drugs influence lipid metabolism. The research prospects of the platelet-hyperlipidaemia interplay in atherosclerosis are also discussed.


Subject(s)
Atherosclerosis , Blood Platelets , Hyperlipidemias , Lipoproteins, LDL , Humans , Atherosclerosis/etiology , Blood Platelets/metabolism , Lipoproteins, LDL/metabolism , Platelet Activation/physiology , Receptors, LDL/metabolism , Hypolipidemic Agents/therapeutic use
5.
Thromb Res ; 237: 64-70, 2024 May.
Article in English | MEDLINE | ID: mdl-38552496

ABSTRACT

Cirrhosis presents with thrombocytopenia and possibly thrombocytopathy. Previous studies exploring platelet function gave conflicting results and most controversies are explained by the variety of methods employed for investigation. We sought to assess in-vitro the overall platelet function in cirrhosis. We investigated 34 patients by using the following tests. (i)Aggregometry. (ii)Measurement of the content of platelet granules. (iii)Cytometric platelet activation. (iv)Plasmatic markers of in-vivo platelet activation. (v)Platelet procoagulant activity by thrombin generation (TG) in platelet-rich plasma (PRP). TG measured in PRP for patients and controls was similar. Platelets from patients with cirrhosis showed reduction of aggregation and secretion of ATP. Similar results were observed for platelet activation parameters such as P-selectin expression and PAC-1 platelet binding. Plasma levels of ßeta-thromboglobulin and soluble P-selectin, were increased in patients-vs-controls. In contrast, there were no patients-vs-controls differences for plasmatic platelet-factor-4. Results are consistent with a state of in-vivo platelet activation and decreased in-vitro aggregation. Since bleeding events following invasive procedures are uncommon in cirrhosis, we speculate that in-vitro aggregometry testing does not reflect the situation occurring in-vivo. Results of the study and pathophysiological considerations support the conclusion that platelet function in cirrhosis as determined by aggregometry, although somewhat impaired, may support the overall hemostatic potential, which is needed for most invasive interventions. These conclusions are in line with the recommendations of international guidelines, warning against indiscriminate use of prophylactic preprocedural administration of platelets before invasive procedures. Decision on platelet support should not be made based on in-vitro laboratory testing for platelet function.


Subject(s)
Blood Platelets , Liver Cirrhosis , Platelet Activation , Platelet Aggregation , Platelet Function Tests , Humans , Male , Female , Middle Aged , Blood Platelets/metabolism , Liver Cirrhosis/blood , Platelet Function Tests/methods , Platelet Activation/physiology , Aged , P-Selectin/blood , Adult , Thrombin/metabolism , Thrombin/analysis
6.
FASEB J ; 38(4): e23468, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38334433

ABSTRACT

The endothelial regulation of platelet activity is incompletely understood. Here we describe novel approaches to find molecular pathways implicated on the platelet-endothelium interaction. Using high-shear whole-blood microfluidics, employing coagulant or non-coagulant conditions at physiological temperature, we observed that the presence of human umbilical vein endothelial cells (HUVEC) strongly suppressed platelet adhesion and activation, via the collagen receptor glycoprotein VI (GPVI) and the PAR receptors for thrombin. Real-time monitoring of the cytosolic Ca2+ rises in the platelets indicated no major improvement of inhibition by prostacyclin or nitric oxide. Similarly under stasis, exposure of isolated platelets to HUVEC reduced the Ca2+ responses by collagen-related peptide (CRP-XL, GPVI agonist) and thrombin (PAR agonist). We then analyzed the label-free phosphoproteome of platelets (three donors), exposed to HUVEC, CRP-XL, and/or thrombin. High-resolution mass spectrometry gave 5463 phosphopeptides, corresponding to 1472 proteins, with good correlation between biological and technical replicates (R > .86). Stringent filtering steps revealed 26 regulatory pathways (Reactome) and 143 regulated kinase substrates (PhosphoSitePlus), giving a set of protein phosphorylation sites that was differentially (44) or similarly (110) regulated by HUVEC or agonist exposure. The differential regulation was confirmed by stable-isotope analysis of platelets from two additional donors. Substrate analysis indicated major roles of poorly studied protein kinase classes (MAPK, CDK, DYRK, STK, PKC members). Collectively, these results reveal a resetting of the protein phosphorylation profile in platelets exposed to endothelium or to conventional agonists and to endothelium-promoted activity of a multi-kinase network, beyond classical prostacyclin and nitric oxide actors, that may contribute to platelet inhibition.


Subject(s)
Platelet Membrane Glycoproteins , Thrombin , Humans , Platelet Membrane Glycoproteins/metabolism , Thrombin/metabolism , Protein Kinases/metabolism , Nitric Oxide/metabolism , Endothelial Cells/metabolism , Platelet Activation/physiology , Blood Platelets/metabolism , Endothelium/metabolism , Prostaglandins I
7.
Semin Thromb Hemost ; 50(2): 224-235, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37192651

ABSTRACT

CD36 (also known as platelet glycoprotein IV) is expressed by a variety of different cell entities, where it possesses functions as a signaling receptor, but additionally acts as a transporter for long-chain fatty acids. This dual function of CD36 has been investigated for its relevance in immune and nonimmune cells. Although CD36 was first identified on platelets, the understanding of the role of CD36 in platelet biology remained scarce for decades. In the past few years, several discoveries have shed a new light on the CD36 signaling activity in platelets. Notably, CD36 has been recognized as a sensor for oxidized low-density lipoproteins in the circulation that mitigates the threshold for platelet activation under conditions of dyslipidemia. Thus, platelet CD36 transduces atherogenic lipid stress into an increased risk for thrombosis, myocardial infarction, and stroke. The underlying pathways that are affected by CD36 are the inhibition of cyclic nucleotide signaling pathways and simultaneously the induction of activatory signaling events. Furthermore, thrombospondin-1 secreted by activated platelets binds to CD36 and furthers paracrine platelet activation. CD36 also serves as a binding hub for different coagulation factors and, thus, contributes to the plasmatic coagulation cascade. This review provides a comprehensive overview of the recent findings on platelet CD36 and presents CD36 as a relevant target for the prevention of thrombotic events for dyslipidemic individuals with an elevated risk for thrombosis.


Subject(s)
CD36 Antigens , Thrombosis , Humans , CD36 Antigens/metabolism , Blood Platelets/metabolism , Platelet Activation/physiology , Thrombosis/etiology , Biology
8.
Artif Organs ; 48(1): 28-36, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37792630

ABSTRACT

BACKGROUND: When nonphysiological stenosis occurs, the transient high shear stress formed in vessels increases the risk of thrombosis and is a potential factor for cardiovascular diseases. But the platelet adhesion and aggregation behavior at nonphysiological post-stenosis and its affecting factors are not fully understood yet. METHODS: In this experiment, platelet aggregation on collagen and fibrinogen at different shear stresses and different hematocrits were observed by microfluidic technology. Platelet activation (P-selectin, glycoprotein IIb/IIIa) and monocyte-platelet aggregate (MPA) levels under different shear stresses were analyzed by flow cytometry. RESULTS: On fibrinogen, platelets aggregate more at higher shear stress conditions. While on collagen, it becomes more difficult for platelets to form stable aggregation at higher shear stress conditions. If platelets adhere initially at low shear stress, stable platelet aggregation can be formed at subsequent high shear stress. Moreover, when the shear stress increases, platelet activity markers (P-selectin, glycoprotein IIb/IIIa and MPAs) increase significantly. Hematocrit affects the degree of platelet aggregation, and the influence of hematocrit is obvious at high shear stress. CONCLUSION: Transient high shear stress (46 ms) can effectively activate platelets. Platelet aggregation behavior was different for coated fibrinogen and collagen protein. Stable platelet adhesion at post-stenosis is more dependent on fibrinogen and platelet aggregation is stable on both fibrinogen and collagen. Hematocrit can significantly affect the formation of platelet aggregation.


Subject(s)
Microfluidics , P-Selectin , Humans , Constriction, Pathologic/metabolism , Platelet Activation/physiology , Platelet Aggregation/physiology , Blood Platelets/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Fibrinogen/metabolism , Collagen/metabolism
9.
FEBS J ; 291(2): 376-391, 2024 01.
Article in English | MEDLINE | ID: mdl-37845743

ABSTRACT

Platelet activation and related cardiovascular complications are the hallmarks of type 2 diabetes (T2D). We investigated the mechanism of platelet activation in T2D using MS-based identification of differentially expressed platelet proteins with a focus on glycosylated forms. Glycosylation is considered one of the common post-translational modifications in T2D, and N/O-linked glycosylation of glycoproteins (GPs)/integrins is known to play crucial roles in platelet activation. Our platelet proteome data revealed elevated levels of GPs GPIbα, GPIIbIIIa, GPIV (CD36), GPV and integrins in T2D patients. T2D platelets had elevated N-linked glycosylation of CD36 at asparagine (Asn)408,417 . Enrichment analysis revealed a close association of glycosylated CD36 with thrombospondin-1, fibrinogen and SERPINA1 in T2D platelets. The glycosylation of CD36 has previously been reported to increase cellular uptake of long-chain fatty acids. Our in silico molecular docking data also showed a favorable binding of cholesterol with glycosylated Asn417 CD36 compared to the non-glycosylated form. We further investigated the CD36:LDL cholesterol axis in T2D. Elevated levels of oxidized-low density lipoprotein (oxLDL) were found to cause significant platelet activation via CD36-mediated stimulation of Lyn-JNK signaling. Sulfo-N-succinimidyl oleate, an inhibitor of CD36, effectively inhibited oxLDL-mediated platelet activation and adhesion in vitro. Our study suggests increased glycosylation of CD36 in T2D platelets as a potential route for oxLDL-mediated platelet activation. The oxLDL:CD36 axis may thus be exploited as a prospective target to develop therapeutics against thrombosis in T2D.


Subject(s)
Diabetes Mellitus, Type 2 , Humans , Diabetes Mellitus, Type 2/genetics , Glycosylation , Molecular Docking Simulation , Platelet Activation/physiology , Lipoproteins, LDL/pharmacology , Risk Factors , Integrins/metabolism
10.
Sci Total Environ ; 912: 169235, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38097078

ABSTRACT

BACKGROUND: Accumulating epidemiological evidence suggests the association between low ambient temperature exposure and the risk of ischemic stroke, but the underlying mechanisms remain unclear. OBJECTIVE: Given the crucial role of platelet activation and thrombosis in ischemic stroke, this study aims to investigate the effect of ambient temperature on platelet activation through multi-center clinical data in Tianjin as well as animal experiments. METHODS: From 2018 to 2020, nearly 3000 ischemic stroke patients from three stroke centers in Tianjin were included in the analysis, among them the ADP induced platelet aggregation rate was available. Meteorological data from the same period had also been collected. After controlling for confounding factors, the generalized additive mixed model (GAMM) was used to evaluate the correlation between environmental temperature and platelet aggregation rate. In further animal experiments, platelet function assessments were conducted on mice from the cold exposure group and the normal temperature group, including platelet aggregation, spreading, and clot retraction. Additionally, tail bleeding and mesentery thrombosis were also tested to monitor hemostasis and thrombosis in vivo. RESULT: A nonlinear "S" shaped relationship between outdoor temperature and platelet aggregation was found. Each 1 °C decrease of mean temperature was associated with an increase of 7.77 % (95 % CI: 2.06 % - 13.48 %) in platelet aggregation. The ambient temperature is not related to other platelet parameters. Subgroup analysis found that males, people aged ≥65 years, and hypertensive individuals are more susceptible to temperature changes. Furthermore, animal experiments demonstrated that the increased CIRBP levels and subsequent activation of p-AKT/p-ERK may be one of the reasons for cold exposure induced platelets activation. CONCLUSION: Both clinical data and basic research support that low ambient temperature exposure has the potential to increase platelet activation. These results provide a basis for understanding the potential mechanism of temperature variations on the pathogenesis of cerebrovascular diseases.


Subject(s)
Ischemic Stroke , Stroke , Thrombosis , Male , Humans , Mice , Animals , Temperature , Platelet Activation/physiology , Platelet Aggregation , Stroke/epidemiology , RNA-Binding Proteins
11.
J Thromb Haemost ; 21(12): 3633-3639, 2023 12.
Article in English | MEDLINE | ID: mdl-37657560

ABSTRACT

BACKGROUND: Most platelet agonists work through G protein-coupled receptors, activating pathways that involve members of the Gq, Gi, and G12/G13 families of heterotrimeric G proteins. Gq signaling has been shown to be critical for efficient platelet activation. Growing evidence suggests that regulatory mechanisms converge on G protein-coupled receptors and Gq to prevent overly robust platelet reactivity. OBJECTIVES: To identify and characterize mechanisms by which Gq signaling is regulated in platelets. METHODS: Based on our prior experience with a Gαi2 variant that escapes regulation by regulator of G protein signaling (RGS) proteins, a Gαq variant was designed with glycine 188 replaced with serine (G188S) and then incorporated into a mouse line so that its effects on platelet activation and thrombus formation could be studied in vitro and in vivo. RESULTS AND CONCLUSIONS: As predicted, the G188S substitution in Gαq disrupted its interaction with RGS18. Unexpectedly, it also uncoupled PLCß-3 from activation by platelet agonists as evidenced by a loss rather than a gain of platelet function in vitro and in vivo. Binding studies showed that in addition to preventing the binding of RGS18 to Gαq, the G188S substitution also prevented the binding of PLCß-3 to Gαq. Structural analysis revealed that G188 resides in the region that is also important for Gαq binding to PLCß-3 in platelets. We conclude that the Gαq signaling node is more complex than that has been previously understood, suggesting that there is cross-talk between RGS proteins and PLCß-3 in the context of Gαq signaling.


Subject(s)
Blood Platelets , GTP-Binding Protein alpha Subunits, Gq-G11 , RGS Proteins , Animals , Mice , Blood Platelets/metabolism , Platelet Activation/physiology , Receptors, G-Protein-Coupled/metabolism , RGS Proteins/genetics , RGS Proteins/metabolism , Signal Transduction , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism
12.
Cell Rep Methods ; 3(7): 100513, 2023 07 24.
Article in English | MEDLINE | ID: mdl-37533637

ABSTRACT

A characteristic clinical complication in cancer patients is the frequent incidence of thrombotic events. Numerous studies have shown hyperactive/activated platelets to be a critical earlier trigger for cancer-associated thrombus formation. However, there currently is no viable approach to monitor specific changes in tumor-associated platelet activity. Here, we describe a chromatograph-like microfluidic device that is highly sensitive to the activity status of peripheral circulating platelets in both tumor-bearing mice and clinical cancer patients. Our results show a strongly positive correlation between platelet activation status and tumor progression. Six-month follow-up data from advanced cancer patients reveal positive links between platelet activity level and thrombus occurrence rate, with a high predictive capacity of thrombotic events (AUC = 0.842). Our findings suggest that circulating platelet activity status determined by this microfluidic device exhibits sensitive, predictive potential for thrombotic events in cancer patients for directing well-timed antithrombosis treatment.


Subject(s)
Neoplasms , Thrombosis , Mice , Animals , Blood Platelets/pathology , Platelet Activation/physiology , Thrombosis/etiology , Neoplasms/complications
13.
Arterioscler Thromb Vasc Biol ; 43(10): 1747-1754, 2023 10.
Article in English | MEDLINE | ID: mdl-37534465

ABSTRACT

Lymphatic vessels form a systemic network that maintains interstitial fluid homeostasis and regulates immune responses and is strictly separated from the circulatory system. During embryonic development, lymphatic endothelial cells originate from blood vascular endothelial cells in the cardinal veins and form lymph sacs. Platelets are critical for separating lymph sacs from the cardinal veins through interactions between CLEC-2 (C-type lectin-like receptor-2) and PDPN (podoplanin) in lymphatic endothelial cells. Therefore, deficiencies of these genes cause blood-filled lymphatic vessels, leading to abnormal lymphatic vessel maturation. The junction between the thoracic duct and the subclavian vein has valves and forms physiological thrombi dependent on CLEC-2/PDPN signaling to prevent blood backflow into the thoracic duct. In addition, platelets regulate lymphangiogenesis and maintain blood/lymphatic separation in pathological conditions, such as wound healing and inflammatory diseases. More recently, it was reported that the entire hemostatic system is involved in lymphangiogenesis. Thus, the hemostatic system plays a crucial role in the establishment, maintenance, and rearrangement of lymphatic networks and contributes to body fluid homeostasis, which suggests that the hemostatic system is a potential target for treating lymphatic disorders. This review comprehensively summarizes the role of the hemostatic system in lymphangiogenesis and lymphatic vessel function and discusses challenges and future perspectives.


Subject(s)
Hemostatics , Lymphatic Vessels , Female , Pregnancy , Humans , Endothelial Cells , Lymphangiogenesis , Platelet Activation/physiology , Lectins, C-Type
14.
J Thromb Haemost ; 21(9): 2339-2353, 2023 09.
Article in English | MEDLINE | ID: mdl-37331517

ABSTRACT

Mechanotransduction is the ability of cells to "feel" or sense their mechanical microenvironment and integrate and convert these physical stimuli into adaptive biochemical cellular responses. This phenomenon is vital for the physiology of numerous nucleated cell types to affect their various cellular processes. As the main drivers of hemostasis and clot retraction, platelets also possess this ability to sense the dynamic mechanical microenvironments of circulation and convert those signals into biological responses integral to clot formation. Like other cell types, platelets leverage their "hands" or receptors/integrins to mechanotransduce important signals in responding to vascular injury to achieve hemostasis. The clinical relevance of cellular mechanics and mechanotransduction is imperative as pathologic alterations or aberrant mechanotransduction in platelets has been shown to lead to bleeding and thrombosis. As such, the aim of this review is to provide an overview of the most recent research related to platelet mechanotransduction, from platelet generation to platelet activation, within the hemodynamic environment and clot contraction at the site of vascular injury, thereby covering the entire "life cycle" of platelets. Additionally, we describe the key mechanoreceptors in platelets and discuss the new biophysical techniques that have enabled the field to understand how platelets sense and respond to their mechanical microenvironment via those receptors. Finally, the clinical significance and importance of continued exploration of platelet mechanotransduction have been discussed as the key to better understanding of both thrombotic and bleeding disorders lies in a more complete mechanistic understanding of platelet function by way of mechanotransduction.


Subject(s)
Thrombosis , Vascular System Injuries , Humans , Blood Platelets/metabolism , Mechanotransduction, Cellular/physiology , Vascular System Injuries/pathology , Hemostasis , Platelet Activation/physiology , Thrombosis/metabolism
15.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 31(2): 495-502, 2023 Apr.
Article in Chinese | MEDLINE | ID: mdl-37096525

ABSTRACT

OBJECTIVE: To study the effect of gradient shear stress on platelet aggregation by microfluidic chip Technology. METHODS: Microfluidic chip was used to simulate 80% fixed stenotic microchannel, and the hydrodynamic behavior of the stenotic microchannel model was analyzed by the finite element analysis module of sollidwork software. Microfluidic chip was used to analyze the adhesion and aggregation behavior of platelets in patients with different diseases, and flow cytometry was used to detect expression of the platelet activation marker CD62p. Aspirin, Tirofiban and protocatechuic acid were used to treat the blood, and the adhesion and aggregation of platelets were observed by fluorescence microscope. RESULTS: The gradient fluid shear rate produced by the stenosis model of microfluidic chip could induce platelet aggregation, and the degree of platelet adhesion and aggregation increased with the increase of shear rate within a certain range of shear rate. The effect of platelet aggregation in patients with arterial thrombotic diseases were significantly higher than normal group (P<0.05), and the effect of platelet aggregation in patients with myelodysplastic disease was lower than normal group (P<0.05). CONCLUSION: The microfluidic chip analysis technology can accurately analyze and evaluate the platelet adhesion and aggregation effects of various thrombotic diseases unde the environment of the shear rate, and is helpful for auxiliary diagnosis of clinical thrombotic diseases.


Subject(s)
Microfluidics , Thrombosis , Humans , Platelet Adhesiveness , Platelet Aggregation , Blood Platelets/metabolism , Platelet Aggregation Inhibitors/metabolism , Platelet Aggregation Inhibitors/pharmacology , Platelet Activation/physiology
16.
Curr Protoc ; 3(3): e711, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36921209

ABSTRACT

Hemostasis is a multifactorial process that involves vasoconstriction of blood vessels, activation of the coagulation cascade, and platelet aggregation. Inappropriate activation of hemostatic processes can result in thrombosis and tissue ischemia. In patients at risk for thrombotic events, antiplatelet therapeutic agents inhibit platelet activation, thereby reducing the incidence of pathologic clot formation. Platelets are activated by several endogenous chemical mediators, including adenosine diphosphate, thrombin, and thromboxane. These activation pathways serve as attractive drug targets. The protocols described in this article are designed to evaluate the preclinical efficacy and safety of novel antiplatelet therapeutics in rabbits. Here, we provide two protocols for blood collection, two for determining platelet activation, and one for assessing bleeding safety. Together, these protocols can be used to characterize the efficacy and safety of antiplatelet agents for hemostasis. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Blood collection via the central ear artery Alternative Protocol 1: Blood collection via the jugular vein Basic Protocol 2: Platelet aggregation assessment via light transmission aggregometry Alternative Protocol 2: Platelet activation assessment via flow cytometry Basic Protocol 3: Determination of tongue bleeding time.


Subject(s)
Blood Coagulation , Thrombosis , Animals , Rabbits , Blood Coagulation/physiology , Platelet Aggregation Inhibitors/adverse effects , Blood Platelets/metabolism , Hemostasis , Platelet Activation/physiology , Thrombosis/drug therapy , Thrombosis/metabolism
17.
Clin Hemorheol Microcirc ; 83(2): 171-180, 2023.
Article in English | MEDLINE | ID: mdl-36463438

ABSTRACT

OBJECTIVE: To investigate the clinical value of platelet and inflammatory factor activation in vascular endothelial injury in hypertension. METHODS: A total of 120 hypertension patients diagnosed in our hospital from December 2019 to June 2021 were enrolled as study objects (Hypertension group); besides, another cohort of 60 healthy people undergoing physical examination at the same period were recruited as the controls (Control group). Next, the baseline clinical characteristics of subjects in the two groups were recorded and compared. Specifically, a hematology analyzer was adopt for detecting the mean platelet volume (MPV), platelet distribution width (PDW) and platelet hematocrit (PCT); ELISA for the level of IL-6, IL-8 and TNF-α; PHILIPS EPIQ 7 C (a device assessing endothelial vasodilator function in a non-invasive fashion) for reactive hyperemia index (RHI); univariate and multivariate regression analysis for risk factors triggering endothelial dysfunction; and Spearman correlation analysis for the correlation of platelet activation indicators and inflammatory factor level with vascular endothelial function. RESULTS: Compared with the Control group, the patients in the Hypertension group exhibited higher levels of MPV, PDW, PCT, inflammatory factors (IL-6, IL-8 and TNF-α) and lower RHI. Moreover, Spearman correlation analysis showed a significant negative correlation of MPV, PDW, PCT, IL-6, IL-8 and TNF-α level with RHI level. In addition, univariate and multivariate regression analysis presented that MPV, PCT, IL-8 and TNF-α were risk factors for vascular endothelial dysfunction. CONCLUSION: The activation of platelet and inflammatory factor is closely related to vascular endothelial function injury in patients with hypertension. To be specifically, platelet and inflammatory factor activation can effectively reflect the vascular endothelial function injury in patients with hypertension and has high clinical value.


Subject(s)
Hypertension , Interleukin-6 , Humans , Interleukin-8 , Tumor Necrosis Factor-alpha , Platelet Count , Blood Platelets , Mean Platelet Volume , Platelet Activation/physiology , Essential Hypertension
18.
Clin Hemorheol Microcirc ; 83(3): 293-303, 2023.
Article in English | MEDLINE | ID: mdl-36565105

ABSTRACT

BACKGROUND: Resistance exercise induces thrombocytosis and increases platelet activation and function. These changes might be related to exercise variables including exercise intensity and type. OBJECTIVE: We compared the effects of traditional resistance exercise (TRE) and circuit resistance exercise (CRE) on cellular markers of platelet activation and function. METHODS: In this crossover study ten healthy male (mean±SD: age, 25.6±2.4 years) subjects performed TRE encompassed 3 sets of 10 repetitions at 100% of 10-RM (10 repetition maximum) for 6 exercises, and CRE protocols included 3 sets of 10 repetitions at 100% of 10-RM for all 6 exercises consecutively, in two separate weeks. To measure platelet indices, PAC1, CD41a, CD42b and CD62P three blood samples were taken before, immediately after exercise, and after 30 min recovery. RESULTS: Lactate concentration, blood pressure, platelet count (PLT), and mean platelet volume (MPV) were significantly (p < 0.05) increased following both resistance exercise trials. Significant increases in PAC1, and CD62P; and significant reductions for CD42b and CD41a were detected following both REs (p < 0.05). However, changes in PAC1 and CD62P were significantly different between the two protocols (p < 0.05), with higher increases detected following CRE. CONCLUSIONS: Acute RE increases platelet indices and platelet activation; and that CRE results in higher platelet activation than TRE, probably due to exercise-induced increases in shear stress.


Subject(s)
Resistance Training , Humans , Male , Young Adult , Adult , Cross-Over Studies , Platelet Activation/physiology , Blood Platelets/physiology , Lactic Acid
19.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-982086

ABSTRACT

OBJECTIVE@#To study the effect of gradient shear stress on platelet aggregation by microfluidic chip Technology.@*METHODS@#Microfluidic chip was used to simulate 80% fixed stenotic microchannel, and the hydrodynamic behavior of the stenotic microchannel model was analyzed by the finite element analysis module of sollidwork software. Microfluidic chip was used to analyze the adhesion and aggregation behavior of platelets in patients with different diseases, and flow cytometry was used to detect expression of the platelet activation marker CD62p. Aspirin, Tirofiban and protocatechuic acid were used to treat the blood, and the adhesion and aggregation of platelets were observed by fluorescence microscope.@*RESULTS@#The gradient fluid shear rate produced by the stenosis model of microfluidic chip could induce platelet aggregation, and the degree of platelet adhesion and aggregation increased with the increase of shear rate within a certain range of shear rate. The effect of platelet aggregation in patients with arterial thrombotic diseases were significantly higher than normal group (P<0.05), and the effect of platelet aggregation in patients with myelodysplastic disease was lower than normal group (P<0.05).@*CONCLUSION@#The microfluidic chip analysis technology can accurately analyze and evaluate the platelet adhesion and aggregation effects of various thrombotic diseases unde the environment of the shear rate, and is helpful for auxiliary diagnosis of clinical thrombotic diseases.


Subject(s)
Humans , Microfluidics , Platelet Adhesiveness , Platelet Aggregation , Blood Platelets/metabolism , Platelet Aggregation Inhibitors/pharmacology , Platelet Activation/physiology , Thrombosis
20.
Int J Mol Sci ; 23(22)2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36430576

ABSTRACT

Type 2 DM is a risk factor for dementia, including Alzheimer's disease (AD), and is associated with brain atrophy. Amyloid ß protein (Aß) deposition in the brain parenchyma is implicated in the neurodegeneration that occurs in AD. Platelets, known as abundant storage of Aß, are recognized to play important roles in the onset and progression of AD. We recently showed that Aß negatively regulates platelet activation induced by thrombin receptor-activating protein (TRAP) in healthy people. In the present study, we investigated the effects of Aß on the TRAP-stimulated platelet activation in DM patients, and the relationship between the individual responsiveness to Aß and quantitative findings of MRI, the volume of white matter hyperintensity (WMH)/intracranial volume (IC) and the volume of parenchyma (PAR)/IC. In some DM patients, Aß reduced platelet aggregation induced by TRAP, while in others it was unchanged or rather enhanced. The TRAP-induced levels of phosphorylated-Akt and phosphorylated-HSP27, the levels of PDGF-AB and the released phosphorylated-HSP27 correlated with the degree of platelet aggregability. The individual levels of not WMH/IC but PAR/IC was correlated with those of TRAP-stimulated PDGF-AB release. Collectively, our results suggest that the reactivity of TRAP-stimulated platelet activation to Aß differs in DM patients from healthy people. The anti-suppressive feature of platelet activation to Aß might be protective for brain atrophy in DM patients.


Subject(s)
Amyloid beta-Peptides , Diabetes Complications , Platelet Activation , Humans , Alzheimer Disease , Amyloid beta-Peptides/metabolism , Atrophy/metabolism , Brain/metabolism , Brain/pathology , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , HSP27 Heat-Shock Proteins/metabolism , Platelet Activation/physiology , Receptor, PAR-1/metabolism , Receptors, Thrombin/metabolism , Diabetes Complications/metabolism , Diabetes Complications/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...