[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (808)

Search Parameters:
Keywords = thrombin

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 4932 KiB  
Article
Parvovirus B19 Infection Is Associated with the Formation of Neutrophil Extracellular Traps and Thrombosis: A Possible Linkage of the VP1 Unique Region
by Bor-Show Tzang, Hao-Yang Chin, Chih-Chen Tzang, Pei-Hua Chuang, Der-Yuan Chen and Tsai-Ching Hsu
Int. J. Mol. Sci. 2024, 25(18), 9917; https://doi.org/10.3390/ijms25189917 (registering DOI) - 13 Sep 2024
Viewed by 300
Abstract
Neutrophil extracellular traps (NETs) formation, namely NETosis, is implicated in antiphospholipid syndrome (APS)-related thrombosis in various autoimmune disorders such as systemic lupus erythematosus (SLE) and APS. Human parvovirus B19 (B19V) infection is closely associated with SLE and APS and causes various clinical manifestations [...] Read more.
Neutrophil extracellular traps (NETs) formation, namely NETosis, is implicated in antiphospholipid syndrome (APS)-related thrombosis in various autoimmune disorders such as systemic lupus erythematosus (SLE) and APS. Human parvovirus B19 (B19V) infection is closely associated with SLE and APS and causes various clinical manifestations such as blood disorders, joint pain, fever, pregnancy complications, and thrombosis. Additionally, B19V may trigger the production of autoantibodies, including those against nuclear and phospholipid components. Thus, exploring the connection between B19V, NETosis, and thrombosis is highly relevant. An in vitro NETosis model using differentiated HL-60 neutrophil-like cells (dHL-60) was employed to investigate the effect of B19V-VP1u IgG on NETs formation. A venous stenosis mouse model was used to test how B19V-VP1u IgG-mediated NETs affect thrombosis in vivo. The NETosis was observed in the dHL-60 cells treated with rabbit anti-B19V-VP1u IgG and was inhibited in the presence of either 8-Br-cAMP or CGS216800 but not GSK484. Significantly elevated reactive oxygen species (ROS), myeloperoxidase (MPO), and citrullinated histone (Cit-H3) levels were detected in the dHL60 treated with phorbol myristate acetate (PMA), human aPLs IgG and rabbit anti-B19V-VP1u IgG, respectively. Accordingly, a significantly larger thrombus was observed in a venous stenosis-induced thrombosis mouse model treated with PMA, human aPLs IgG, rabbit anti-B19V-VP1u IgG, and human anti-B19V-VP1u IgG, respectively, along with significantly increased amounts of Cit-H3-, MPO- and CRAMP-positive infiltrated neutrophils in the thrombin sections. This research highlights that anti-B19V-VP1u antibodies may enhance the formation of NETosis and thrombosis and implies that managing and treating B19V infection could lower the risk of thrombosis. Full article
Show Figures

Figure 1

Figure 1
<p>Rabbit anti-B19 VP1u IgG induces NETs release. The dHL-60 cells treated with PMA, rabbit IgG, and rabbit anti-B19V-VP1u IgG (rabbit VP1u IgG), respectively, were stained with (<b>A</b>) SYTOX Green (left panel) and Hoechst 33342 (middle panel). The merged images were shown in the right panel, and arrows indicated the NETs. (<b>B</b>) The quantified results of SYTOX Green-positive signal. Three independent experiments were performed. The symbols * and # indicate significance (<span class="html-italic">p</span> &lt; 0.05) as compared to the control and rabbit IgG, respectively. <span class="html-italic">p</span>-value is calculated by one-way ANOVA followed by Tukey’s multiple comparison test.</p>
Full article ">Figure 2
<p>Inhibitory effect of NETs inhibitors (8-Br-cAMP, CGS21680, GSK484) on PMA and rabbit anti-B19V-VP1u IgG induced NETs. (<b>A</b>) The PMA and (<b>B</b>) rabbit anti-B19V-VP1u IgG (rabbit VP1u IgG) treated-dHL-60 cells in the presence of 8-Br-cAMP, CGS21680 or GSK484 were stained with SYTOX Green (left panel) and Hoechst 33342 (middle panel). The merged images were shown in the right panel, and arrows indicated the NETs. The quantified results of SYTOX Green-positive signals were shown in the lower panel. Three independent experiments were performed. The symbols *, #, and <span>$</span> indicate significance (<span class="html-italic">p</span> &lt; 0.05) as compared to the control, PMA, and rabbit anti-B19V-VP1u IgG, respectively. <span class="html-italic">p</span>-value is calculated by one-way ANOVA followed by Tukey’s multiple comparison test.</p>
Full article ">Figure 2 Cont.
<p>Inhibitory effect of NETs inhibitors (8-Br-cAMP, CGS21680, GSK484) on PMA and rabbit anti-B19V-VP1u IgG induced NETs. (<b>A</b>) The PMA and (<b>B</b>) rabbit anti-B19V-VP1u IgG (rabbit VP1u IgG) treated-dHL-60 cells in the presence of 8-Br-cAMP, CGS21680 or GSK484 were stained with SYTOX Green (left panel) and Hoechst 33342 (middle panel). The merged images were shown in the right panel, and arrows indicated the NETs. The quantified results of SYTOX Green-positive signals were shown in the lower panel. Three independent experiments were performed. The symbols *, #, and <span>$</span> indicate significance (<span class="html-italic">p</span> &lt; 0.05) as compared to the control, PMA, and rabbit anti-B19V-VP1u IgG, respectively. <span class="html-italic">p</span>-value is calculated by one-way ANOVA followed by Tukey’s multiple comparison test.</p>
Full article ">Figure 3
<p>Rabbit B19V-VP1u IgG increases citH3 and MPO expressions. (<b>A</b>) The representative results of the dHL-60 cells were treated with PMA, normal human IgG (NH IgG), human aPLs IgG, rabbit IgG, and rabbit anti-B19V-VP1u IgG (rabbit VP1u IgG), and the presence of NETs was measured by detecting the expressions of citH3 and MPO with flow cytometry (<b>B</b>) The quantified results of NETs. Three independent experiments were performed. The symbols *, <span>$</span>, and # indicate significance (<span class="html-italic">p</span> &lt; 0.05) as compared to the control, human IgG, and rabbit anti-B19V-VP1u IgG, respectively. <span class="html-italic">p</span>-value is calculated by one-way ANOVA followed by Tukey’s multiple comparison test.</p>
Full article ">Figure 4
<p>Rabbit anti-B19V-VP1u IgG increases citrullinated histone H3 (Cit-H3) expression. (<b>A</b>) Western blot analysis was used to detect the presence of Cit-H3 in dHL-60 cells treated with PMA, normal human IgG (NH IgG), human aPLs IgG, rabbit IgG, and rabbit anti-B19V-VP1u IgG (rabbit VP1u IgG). (<b>B</b>) The ratio of Cit-H3 amount relative to total H3. (<b>C</b>) The ratio of Cit-H3 amount relative to β-actin. Three independent experiments were performed. The symbols *, <span>$</span>, and # indicate significance (<span class="html-italic">p</span> &lt; 0.05) compared to the Control, NH IgG, and rabbit VP1u IgG, respectively. <span class="html-italic">p</span>-value is calculated by one-way ANOVA followed by Tukey’s multiple comparison test.</p>
Full article ">Figure 5
<p>Human aPLs IgG and rabbit anti-B19V-VP1u IgG increase ROS production. (<b>A</b>) The dHL-60 cells were treated with PMA, normal human IgG (NH IgG), human aPLs IgG, rabbit IgG, and rabbit anti-B19V-VP1u IgG (rabbit VP1u IgG), and the ROS level was measured in the presence of Dichloro-dihydro-fluorescein diacetate (DCFH-DA) with flow cytometry. The arrow indicates the proportion of DCF-positive cells, defined as cells exhibiting fluorescence intensity greater than the established threshold value. (<b>B</b>) The quantified results of mean DCF. Three independent experiments were performed. The symbols *, <span>$</span>, and # indicate significance (<span class="html-italic">p</span> &lt; 0.05) compared to the control, NH IgG, and rabbit VP1u IgG, respectively. <span class="html-italic">p</span>-value is calculated by one-way ANOVA followed by Tukey’s multiple comparison test.</p>
Full article ">Figure 6
<p>Rabbit anti-B19V-VP1u IgG promotes venous thrombosis in C57BL/6 mice with inferior vena cava ligation. (<b>A</b>) The representative images of thrombus from the mice treated with PBS, rabbit IgG, and rabbit anti-B19V-VP1u IgG (rabbit VP1u IgG). Sections of the thrombus stained with (<b>B</b>) H&amp;E, (<b>C</b>) anti-citrullinated histone H3 (Cit-H3), (<b>D</b>) MPO, and (<b>E</b>) CRAMP. Scale bar = 1 mm. The right panel showed the quantified results of thrombus weight, thrombus length, and positive cells of Cit-H3, MPO, and CRAMP signal. The symbol * and # indicate significance (<span class="html-italic">p</span> &lt; 0.05) as compared to the control and rabbit IgG, respectively. <span class="html-italic">p</span>-value is calculated by one-way ANOVA followed by Tukey’s multiple comparison test.</p>
Full article ">Figure 7
<p>Human aPLs and anti-B19V-VP1u IgG promote venous thrombosis in C57BL/6 mice with inferior vena cava ligation. (<b>A</b>) The representative images of thrombus from the mice treated with normal human IgG (NH IgG), human aPLs IgG, and human anti-B19V-VP1u IgG. Sections of the thrombus stained with (<b>B</b>) H&amp;E, (<b>C</b>) anti-citrullinated histone H3 (Cit-H3), (<b>D</b>) MPO, and (<b>E</b>) CRAMP. Scale bar = 1 mm. The right panel showed the quantified results of thrombus weight, length, and positive cells of Cit-H3, MPO, and CRAMP signal. The symbol * indicate a significance (<span class="html-italic">p</span> &lt; 0.05) as compared to the NH IgG. <span class="html-italic">p</span>-value is calculated by one-way ANOVA followed by Tukey’s multiple comparison test.</p>
Full article ">
16 pages, 9164 KiB  
Article
Exploring the Role of Fibrin Gels in Enhancing Cell Migration for Vasculature Formation
by Joana A. Moura, Hugh J. Barlow, Shareen H. Doak, Karl Hawkins, Iris Muller and Martin J. D. Clift
J. Funct. Biomater. 2024, 15(9), 265; https://doi.org/10.3390/jfb15090265 - 12 Sep 2024
Viewed by 306
Abstract
A hallmark of angiogenesis is the sprouting of endothelial cells. To replicate this event in vitro, biomaterial approaches can play an essential role in promoting cell migration. To study the capacity of a scaffold of fibrin (fibrinogen:thrombin mix) to support the movement of [...] Read more.
A hallmark of angiogenesis is the sprouting of endothelial cells. To replicate this event in vitro, biomaterial approaches can play an essential role in promoting cell migration. To study the capacity of a scaffold of fibrin (fibrinogen:thrombin mix) to support the movement of the endothelial cells, the migration area of spheroids formed with the HULEC cell line was measured. The cells were first allowed to form a spheroid using the hanging drop technique before being encapsulated in the fibrin gel. The cells’ migration area was then measured after two days of embedding in the fibrin gel. Various conditions affecting fibrin gel polymerization, such as different concentrations of fibrinogen and thrombin, were evaluated alongside rheology, porosity, and fiber thickness analysis to understand how these factors influenced cell behavior within the composite biomaterial. Data point toward thrombin’s role in governing fibrin gel polymerization; higher concentrations result in less rigid gels (loss tangent between 0.07 and 0.034) and increased cell migration (maximum concentration tested: 5 U/mL). The herein presented method allows for a more precise determination of the crosslinking conditions of fibrin gel that can be used to stimulate angiogenic sprouting. Full article
(This article belongs to the Topic Advanced Functional Materials for Regenerative Medicine)
Show Figures

Figure 1

Figure 1
<p>Endothelial cell 2D characterization. (<b>A</b>) Live cell quantification with trypan blue over 7 days. The cell growth fits the following exponential curve: y = 5381e0.2741x. (<b>B</b>) Live and dead cell ratio using trypan blue assay. (<b>C</b>) Picogreen dsDNA quantification over 7 days on the left y-axis. The increase in dsDNA is statistically significant from day 1 to 7 with a <span class="html-italic">p</span>-value of 0.0070. The right y-axis corresponds to the MTS absorbance over the number of live cells. The cell’s metabolism increases from day 1 to 7 and is also statistically significant, with a <span class="html-italic">p</span>-value of 0.0040. (<b>D</b>) Expression of the CD31 marker on days 5 and 7. The increase in CD31 surface marker from day 5 to 7 is statistically significant, with a <span class="html-italic">p</span>-value of 0.044. This is associated also with a decrease in CD31, with a <span class="html-italic">p</span>-value of 0.044. (<b>E</b>) Endothelial cells on day 7 of the cell culture stained with DAPI, Phalloidin, and CD31 (scale bar represents 50 µm). (<b>F</b>) VE-Cadherin (scale bar represents 50 µm). The significance values were taken when <span class="html-italic">p</span>-No value &lt; 0.05, graphically denoted as * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01. The error bars correspond to the Standard Deviation. <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 2
<p>Fibrin gel rheological characterization. (<b>A</b>) Storage (G′) and (<b>B</b>) Loss modulus (G″) of fibrin gel in Pascal (Pa). The data are grouped by the fibrinogen concentration. (<b>C</b>) Storage (G′) and (<b>D</b>) Loss modulus (G″) of fibrin gel in Pascal (Pa). The data are grouped by the thrombin concentration. (<b>E</b>) The table below shows the loss tangent values. The color scheme highlights values in the same range. The fibrinogen concentrations tested were 5, 2.5, and 1.25 mg/mL, and for thrombin, they were 5, 1, and 0.1 U/mL. The error bars correspond to the Standard Deviation. The data are between <span class="html-italic">n</span> = 3 and 6. The significance values were taken when <span class="html-italic">p</span> &lt; 0.05, graphically denoted as * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Confocal microscope images of fibrin gel polymerized with 5, 2.5, and 1.25 mg/mL of fibrinogen and 5, 1, and 0.1 U/mL of thrombin. The fibrinogen solution was mixed with 4% of fibrinogen from human plasma Alexa Fluor 488 conjugated to allow for fiber visualization when excited with a 488 laser. The scale represents 20 µm. <span class="html-italic">n</span> = 6 to 9.</p>
Full article ">Figure 4
<p>Scanning electron microscope images of fibrin gel polymerized with 5, 2.5, and 1.25 mg/mL of fibrinogen and 5, 1, and 0.1 U/mL of thrombin. The scale bar represents 5 µm, except for condition 1.25 mg/mL with 0.1 U/mL, which represents 4 µm. <span class="html-italic">n</span> = 3.</p>
Full article ">Figure 5
<p>Fibrin gel porosity and fiber diameter. (<b>A</b>) Percentage of the area corresponding to pores in the fibrin gel in function of fibrinogen and thrombin concentration (<span class="html-italic">n</span> = 6 to 9). (<b>B</b>) Fiber diameter in micrometers (µm) in function of fibrinogen and thrombin concentration (<span class="html-italic">n</span> = 3). The significance values were taken when <span class="html-italic">p</span> &lt; 0.05, graphically denoted as * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001. The error bars represent the Standard Deviation.</p>
Full article ">Figure 6
<p>Endothelial cell migration area in function of the fibrinogen and thrombin polymerization conditions. The asterisk symbol in the plot represents * <span class="html-italic">p</span> &lt; 0.05. The error bars represent the Standard Deviation. <span class="html-italic">n</span> = 5.</p>
Full article ">Figure 7
<p>Spheroid bright and fluorescence staining. Spheroids on day 2 after fibrin embedding. The column on the left represents hematoxylin stain, and on the right, Hoechst and PI. The scale bar represents 200 µm. <span class="html-italic">n</span> = 5.</p>
Full article ">
14 pages, 1176 KiB  
Article
Platelet Aggregation Alterations in Patients with Severe Viral Infection Treated at the Intensive Care Unit: Implications for Mortality Risk
by Wojciech Bakowski, Jakub Smiechowicz, Barbara Dragan, Waldemar Goździk and Barbara Adamik
Pathogens 2024, 13(9), 778; https://doi.org/10.3390/pathogens13090778 - 10 Sep 2024
Viewed by 274
Abstract
Severe viral infections often result in abnormal platelet function, affecting various stages of hemostasis. Activated platelets are often considered prothrombotic and more susceptible to further stimulation. However, emerging evidence suggests that initial hyperactivation is followed by platelet exhaustion and hypo-responsiveness, affecting platelet degranulation, [...] Read more.
Severe viral infections often result in abnormal platelet function, affecting various stages of hemostasis. Activated platelets are often considered prothrombotic and more susceptible to further stimulation. However, emerging evidence suggests that initial hyperactivation is followed by platelet exhaustion and hypo-responsiveness, affecting platelet degranulation, activation, and aggregation. We examined early alterations in platelet aggregation among patients (N = 28) with acute respiratory distress syndrome and SARS-CoV-2 infection who were receiving mechanical ventilation and venovenous extracorporeal membrane oxygenation support. Blood samples were stimulated with four different activators: arachidonic acid, adenosine diphosphate, thrombin receptor-activating protein 6, and ristocetin. Our observations revealed that platelet aggregation was reduced in most patients upon admission (ranging from 61 to 89%, depending on the agonist used), and this trend intensified during the 5-day observation period. Concurrently, other coagulation parameters remained within normal ranges, except for elevated d-dimer and fibrinogen levels. Importantly, we found a significant association between platelet aggregation and patient mortality. Impaired platelet aggregation was more severe in patients who ultimately died, and reduced aggregation was associated with a significantly lower probability of survival, as confirmed by Kaplan–Meier analysis (p = 0.028). These findings underscore the potential of aggregometry as an early detection tool for identifying patients at higher risk of mortality within this specific cohort. Full article
(This article belongs to the Section Emerging Pathogens)
Show Figures

Figure 1

Figure 1
<p>Graphs comparing platelet receptor activity between survivors and nonsurvivors on days 1, 3, and 5 of ECMO treatment. Dotted lines represent the lower and upper reference ranges for each test: ASPI 745–1361 AU/min (<b>A</b>), ADP 534–1220 AU/min (<b>B</b>), TRAP 941–1536 AU/min (<b>C</b>), RISTO 896–2013 AU/min (<b>D</b>). The <span class="html-italic">p</span>-values represent differences between the study groups at each time point. The box plots represent the median values (middle line) with upper and lower quartiles (box); the whiskers represent the minimum and maximum values.</p>
Full article ">Figure 1 Cont.
<p>Graphs comparing platelet receptor activity between survivors and nonsurvivors on days 1, 3, and 5 of ECMO treatment. Dotted lines represent the lower and upper reference ranges for each test: ASPI 745–1361 AU/min (<b>A</b>), ADP 534–1220 AU/min (<b>B</b>), TRAP 941–1536 AU/min (<b>C</b>), RISTO 896–2013 AU/min (<b>D</b>). The <span class="html-italic">p</span>-values represent differences between the study groups at each time point. The box plots represent the median values (middle line) with upper and lower quartiles (box); the whiskers represent the minimum and maximum values.</p>
Full article ">Figure 2
<p>Kaplan–Meier curves stratified by the presence of platelet aggregation results recorded on day 1 in all tests (ASPI, ADP, TRAP, and RISTO) below vs. within the reference range (log-rank test).</p>
Full article ">Figure 3
<p>ROC curve statistics for aggregometry results measured on day 1 as predictors of ICU mortality. AUC, area under the curve.</p>
Full article ">
15 pages, 2335 KiB  
Article
Looping Flexible Fluoropolymer Microcapillary Film Extends Analysis Times for Vertical Microfluidic Blood Testing
by Rüya Meltem Sarıyer, Kirandeep K. Gill, Sarah H. Needs, Nuno M. Reis, Chris I. Jones and Alexander Daniel Edwards
Sensors 2024, 24(18), 5870; https://doi.org/10.3390/s24185870 - 10 Sep 2024
Viewed by 275
Abstract
The microfluidic measurement of capillary flow can be used to evaluate the response of biological samples to stimulation, where distance and velocity are altered. Melt-extruded multi-bored microfluidic capillaries allow for high-throughput testing with low device cost, but simple devices may limit control over [...] Read more.
The microfluidic measurement of capillary flow can be used to evaluate the response of biological samples to stimulation, where distance and velocity are altered. Melt-extruded multi-bored microfluidic capillaries allow for high-throughput testing with low device cost, but simple devices may limit control over sample flow when compared to the more complex “lab-on-a-chip” devices produced using advanced microfluidic fabrication methods. Previously, we measured the dynamics of global haemostasis stimulated by thrombin by dipping straight vertical microcapillaries into blood, but only the most rapid response could be monitored, as flow slowed significantly within 30 s. Here, we show an innovative method to extend both the stimulation process and flow measurement time without increasing the cost of the device by adding simple loops to the flexible extruded device. The loops enable longer time-scale measurements by increasing resistance to flow, thereby reducing the dependence on high stimulus concentrations for rapid reactions. The instantaneous velocity and equilibrium heights of straight and looped vertical microcapillary films were assessed with water, plasma and whole blood, showing that the loops create additional frictional resistances, reduce flow velocity and prolong residence times for increased time scales of the stimulation process. A modified pressure balance model was used to capture flow dynamics with the added loop. Looped devices loaded with thrombin and collagen showed an improved detection of blood stimulation responses even with lower stimulus concentrations, compared to straight vertical capillaries. Thrombin-activated blood samples in straight capillaries provided a maximum measurement zone of only 4 mm, while the looped design significantly increased this to 11 mm for much longer time scale measurements. Our results suggest that extending stimulation times can be achieved without complex microfluidic fabrication methods, potentially improving concentration–response blood stimulation assays, and may enhance the accuracy and reliability. We conclude adding a loop to low-cost extruded microfluidic devices may bring microfluidic devices closer to delivering on their promise of widespread, decentralized low-cost evaluation of blood response to stimulation in both research and clinical settings. Full article
(This article belongs to the Special Issue Biomedical Sensors Based on Microfluidics)
Show Figures

Figure 1

Figure 1
<p>The loop design provides a way of increasing the measurement range by controlling the stimulation time. (<b>a</b>) Concept of capillary rise in “dip-and-test” microfluidic straight vertical dipsticks to measure blood function. (<b>b</b>) Vertical capillary rise with added loop concept with front and side view. (<b>c</b>) The dip-stick tests work by allowing blood samples to flow up vertically into ~200-micron straight channels where the blood mixes with stimuli. When a simple loop is added to the vertical dipstick, blood flows vertically upwards, but then around a loop, and while in that loop it begins to be stimulated. (<b>d</b>) The vertical flow in straight channels takes only a few seconds before slowing down. With the addition of a loop, this rapid flow continues for &gt;30 s or so. (<b>e</b>) A representation of capillary rise in height and length over time for straight, loop- and double-loop-added dipsticks. Data indicates the mean of 10 replicate capillaries where error bars indicate ± SD from a single donor; similar results were observed in 2 replicate experiments with different donors. (<b>f</b>) In loop dipsticks, the blood rises over a greater distance due to the added length of the loop. Straight dipsticks increase by about 10 mm after 30 s, while looped dipsticks achieve around 30 mm within the same time.</p>
Full article ">Figure 2
<p>Adding loops to the straight vertical strips creates additional frictional resistances (<math display="inline"><semantics> <mrow> <mo>∆</mo> <mi>P</mi> <mi>F</mi> </mrow> </semantics></math>). (<b>a</b>) <math display="inline"><semantics> <mrow> <msub> <mrow> <mo>∆</mo> <mi>P</mi> </mrow> <mrow> <mi>F</mi> <mn>1</mn> </mrow> </msub> </mrow> </semantics></math> represents the pressure difference driving the flow, while <math display="inline"><semantics> <mrow> <msub> <mrow> <mo>∆</mo> <mi>P</mi> </mrow> <mrow> <mi>F</mi> <mn>2</mn> </mrow> </msub> </mrow> </semantics></math> and <math display="inline"><semantics> <mrow> <msub> <mrow> <mo>∆</mo> <mi>P</mi> </mrow> <mrow> <mi>F</mi> <mn>2</mn> </mrow> </msub> <mo>+</mo> <msub> <mrow> <mo>∆</mo> <mi>P</mi> </mrow> <mrow> <mi>F</mi> <mn>3</mn> </mrow> </msub> </mrow> </semantics></math> represent the additional pressure differences due to the loops. (<b>b</b>) The graph shows how the instantaneous superficial fluid velocity <math display="inline"><semantics> <mrow> <mi>d</mi> <mi>H</mi> <mo>/</mo> <mi>d</mi> <mi>t</mi> </mrow> </semantics></math> changes with the reciprocal <math display="inline"><semantics> <mrow> <mi>H</mi> <mo>(</mo> <mi>t</mi> <mo>)</mo> </mrow> </semantics></math> for each fluid. The experimental data points are plotted along with the model predictions calculated from the transient pressure balance. The dynamic flows observed for pure water, PRP and WB in straight strips fit our model. Data were collected from 10 replicate capillaries, with error bars representing ± SD from a single donor; consistent results were observed across 2 replicate experiments with different donors. (<b>c</b>) Graphs show experimental data points and corresponding model predictions for straight, single-loop and double-loop configurations. Adding loops reduces the flow rate, which leads to longer residence times. The differences in flow rate become more pronounced with different fluid viscosities. Mean of 10 replicate capillaries are shown in the chart, with similar results across 2 independent experiments (N = 2).</p>
Full article ">Figure 3
<p>The capillary rise is unaffected by the loop, and impact of stimulation with thrombin and collagen can be measured. (<b>a</b>) Demonstration of the capillary rise of water, buffer and blood components in straight, loop- and double-loop-added dipsticks after 15 s. (<b>b</b>) An image of the strips before reaching equilibrium height. Strips of different sizes were used. (<b>c</b>) After 10 min, the equilibrium heights reached by water, plasma, and blood in the dipsticks with straight, loop, and double-loop configurations showed nearly the same values. Data indicates 10 replicate capillaries where error bars indicate ± SD from a single donor. (<b>d</b>) At the end of the 2 min experiment, stimulation was observed in straight and loop-added dipsticks loaded with thrombin (loaded with 0, 5, 15, 50, 150 and 300 U mL<sup>−1</sup>). Since blood at concentrations of 50 U mL<sup>−1</sup> and above could not pass through the loop within the 2 min time frame, these concentrations are not included in the calculations. The images provided are representative examples, with similar results observed across four different donors (N = 4). (<b>e</b>) Stimulation was observed in straight and loop-added dipsticks loaded with collagen (loaded with 0, 50, 158 and 500 mg mL<sup>−1</sup>). The images are representative examples of three replicate collagen stimulation experiments (N = 3).</p>
Full article ">
9 pages, 806 KiB  
Article
Assessment of Platelet Aggregation and Thrombin Generation in Patients with Familial Chylomicronemia Syndrome Treated with Volanesorsen: A Cross-Sectional Study
by Ilenia Lorenza Calcaterra, Renata Santoro, Nicoletta Vitelli, Ferdinando Cirillo, Guido D’Errico, Cornelia Guerrino, Giovanna Cardiero, Maria Donata Di Taranto, Giuliana Fortunato, Gabriella Iannuzzo and Matteo Nicola Dario Di Minno
Biomedicines 2024, 12(9), 2017; https://doi.org/10.3390/biomedicines12092017 - 4 Sep 2024
Viewed by 295
Abstract
Background: The antisense oligonucleotide against APOC3 mRNA volanesorsen was recently introduced to treat Familial Chylomicronemia Syndrome (FCS). Cases of decreased platelet count are reported among patients treated with volanesorsen. The aim of the study was to evaluate platelet function and thrombin generation (TG) [...] Read more.
Background: The antisense oligonucleotide against APOC3 mRNA volanesorsen was recently introduced to treat Familial Chylomicronemia Syndrome (FCS). Cases of decreased platelet count are reported among patients treated with volanesorsen. The aim of the study was to evaluate platelet function and thrombin generation (TG) assessment in FCS patients receiving volanesorsen. We performed a cross-sectional study on FCS patients treated with volanesorsen. Methods: Changes in platelet count PLC were assessed from baseline to Tw12 and Tw36. To assess TG, samples were processed by CAT (with PPP-reagent LOW). The results were expressed by the thrombogram graphic (thrombin variation over time); LagTime; endogenous thrombin potential (ETP); peak; time to reach peak (ttpeak), StartTail and Velocity Index. Platelet aggregation was assessed by testing different agonists using the turbidimetry method. Results: Four FCS patients and four matched healthy controls were included in the present study. Changes in PLC were 30% at Tw12 and 34% at Tw36. Thrombin generation results showed values in the normal range (for patients and controls, respectively, LagTime:10.42 ± 4.40 and 9.25 ± 0.99; ttPeak:14.33 ± 4.01 and 13.10 ± 0.67; StartTail: 32.13 ± 3.54 and 29.46 ± 1.69; Velocity Index: 20.21 ± 3.63 and 33.05 ± 13.21; ETP: 599.80 ± 73.47 and 900.2 ± 210.99; peak value: 76.84 ± 1.07 and 123.30 ± 39.45) and no significant difference between cases and controls. Platelet aggregation test showed values in range, with no significant difference compared to healthy controls. Conclusions: Our study showed for the first time that no significant changes in general hemostasis assessed by TG and in platelet function were observed in FCS patients receiving volanesorsen. Full article
Show Figures

Figure 1

Figure 1
<p>Individual percent changes in PLC from baseline to T12w and T36w (<b>a</b>). Cumulative percent changes in PLC from baseline to T12w and T36w (<b>b</b>).</p>
Full article ">Figure 2
<p>Outcome of thrombin generation (CAT) shown for each patient.</p>
Full article ">
12 pages, 2755 KiB  
Article
Decreased Protein C Pathway Activity in COVID-19 Compared to Non-COVID Sepsis: An Observational and Comparative Cohort Study
by Heiko Rühl, Christian Bode, Tobias Becher, Sebastian Eckert, Ghaith Mohsen, Hannah L. McRae, Jens Müller, Sara Reda, Dirk Loßnitzer, Johannes Oldenburg, Christian Putensen and Bernd Pötzsch
Biomedicines 2024, 12(9), 1982; https://doi.org/10.3390/biomedicines12091982 - 2 Sep 2024
Viewed by 304
Abstract
Sepsis-associated coagulopathy increases risk of mortality. Impairment of the anticoagulant protein C (PC) pathway may contribute to the thrombotic phenotype in coronavirus disease 2019 (COVID-19) sepsis. This study assessed the functionality of this pathway in COVID-19 and non-COVID sepsis by measuring its key [...] Read more.
Sepsis-associated coagulopathy increases risk of mortality. Impairment of the anticoagulant protein C (PC) pathway may contribute to the thrombotic phenotype in coronavirus disease 2019 (COVID-19) sepsis. This study assessed the functionality of this pathway in COVID-19 and non-COVID sepsis by measuring its key enzymes, thrombin and activated PC (APC). The study population included 30 patients with COVID-19, 47 patients with non-COVID sepsis, and 40 healthy controls. In healthy controls, coagulation activation and subsequent APC formation was induced by 15 µg/kg recombinant activated factor VII one hour before blood sampling. APC and thrombin in plasma were measured using oligonucleotide-based enzyme capture assays. The indirect thrombin markers prothrombin-fragment 1+2 (F1+2) and thrombin-antithrombin complex (TAT) were also measured. Compared with stimulated healthy controls, median thrombin, F1+2, and TAT levels were higher in patients with COVID-19 (up to 6-fold, p < 2 × 10−6) and non-COVID sepsis (up to 4.7-fold, p < 0.010). APC levels were 2.4-fold higher in patients with COVID-19 (7.44 pmol/L, p = 0.011) and 3.4-fold higher in non-COVID sepsis patients (10.45 pmol/L, p = 2 × 10−4) than in controls (3.08 pmol/L). Thrombin markers and APC showed correlation in both COVID-19 (r = 0.364–0.661) and non-COVID sepsis patients (r = 0.535–0.711). After adjustment for PC levels, median APC/thrombin, APC/F1+2, and APC/TAT ratios were 2-fold (p = 0.036), 6-fold (p = 3 × 10−7) and 3-fold (p = 8 × 10−4) lower in the COVID-19 group than in the non-COVID sepsis group, and the latter two were also lower in the COVID-19 group than in stimulated healthy controls. In conclusion, it was found that a comparatively lower anticoagulant APC response in COVID-19 patients as compared to non-COVID sepsis patients, potentially linked to endothelial dysfunction, contributes to the prothrombotic phenotype of COVID-19 sepsis. Full article
(This article belongs to the Special Issue Sepsis: Pathophysiology and Early Diagnostics)
Show Figures

Figure 1

Figure 1
<p>Study flow chart. Hepatic and renal function tests included transaminases, γ-glutamyl transferase, urea, and creatinine in serum. BMI, body mass index.</p>
Full article ">Figure 2
<p>Hemostasis parameters. (<b>A</b>) PT in patients with COVID-19 induced sepsis (red, n = 30), non-COVID sepsis (blue, n = 37), and healthy controls (grey, n = 40). (<b>B</b>) aPTT in COVID-19 (n = 30) and non-COVID induced sepsis (n = 43), and healthy controls (n = 40). (<b>C</b>) Plasma levels of PC and (<b>D</b>) D-dimer in COVID-19 (n = 30) and non-COVID induced sepsis (n = 47), and in healthy controls one hour after IV administration of 15 µg/kg rFVIIa (n = 40). Data are presented as box plots indicating quartiles and median of the data, the whiskers extending up to 1.5 times the IQR from the box, and circles showing outlying values. <span class="html-italic">p</span> values were calculated using the Kruskal–Wallis test followed by pairwise comparison using the Dunn procedure. The Bonferroni method was used to correct for multiple comparisons (n = 3). Values of <span class="html-italic">p</span> ≤ 0.05 are shown in bold font.</p>
Full article ">Figure 3
<p>Thrombin markers and APC. Plasma levels of (<b>A</b>) thrombin, (<b>B</b>) F1+2, (<b>C</b>) TAT, and (<b>D</b>) APC were compared in patients with COVID-19 induced sepsis (red, n = 30), non-COVID sepsis (blue, n = 47), and in healthy controls one hour after IV administration of 15 µg/kg rFVIIa (grey, n = 40). Data are presented as box plots indicating quartiles and median of the data, the whiskers extending up to 1.5 times the IQR from the box, and circles showing outlying values. <span class="html-italic">p</span> values were calculated using the Kruskal–Wallis test followed by pairwise comparison using the Dunn procedure. The Bonferroni method was used to correct for multiple comparisons (n = 3). Values of <span class="html-italic">p</span> ≤ 0.05 are shown in bold font. The correlation between plasma levels of (<b>E</b>) thrombin, as well as plasma levels of the thrombin formation markers (<b>F</b>) F1+2 and (<b>G</b>) TAT, and plasma levels of APC. The Pearson correlation coefficient (r) is shown in cases of <span class="html-italic">p</span> ≤ 0.05.</p>
Full article ">Figure 4
<p>APC in plasma in relation to thrombin formation. Plasma levels of APC, and the ratio of APC/PC in relation to plasma levels of (<b>A</b>) thrombin, (<b>B</b>) F1+2, and (<b>C</b>) TAT were compared in patients with COVID-19 induced sepsis (red, n = 30), non-COVID sepsis (blue, n = 47), and in healthy controls one hour after IV administration of 15 µg/kg recombinant activated factor VII (grey, n = 40). Data are presented as box plots indicating quartiles and median of the data, the whiskers extending up to 1.5 times the IQR from the box, and circles showing outlying values. <span class="html-italic">p</span> values were calculated using the Kruskal–Wallis test followed by pairwise comparison using the Dunn procedure. The Bonferroni method was used to correct for multiple comparisons (n = 3).</p>
Full article ">
16 pages, 309 KiB  
Review
Thrombosis and Thrombotic Risk in Athletes
by Ciro Miele, Cristina Mennitti, Alessandro Gentile, Iolanda Veneruso, Carmela Scarano, Aniello Vastola, Ilaria La Monica, Fabiana Uomo, Fernanda Iafusco, Filomena Capasso, Raffaela Pero, Valeria D’Argenio, Barbara Lombardo, Nadia Tinto, Pierpaolo Di Micco, Olga Scudiero, Giulia Frisso and Cristina Mazzaccara
J. Clin. Med. 2024, 13(16), 4881; https://doi.org/10.3390/jcm13164881 - 19 Aug 2024
Viewed by 563
Abstract
The hemostatic system is characterized by a delicate balance between pro- and anticoagulant forces, and the smallest alteration can cause serious events such as hemorrhages or thrombosis. Although exercise has been shown to play a protective role in athletes, several factors may increase [...] Read more.
The hemostatic system is characterized by a delicate balance between pro- and anticoagulant forces, and the smallest alteration can cause serious events such as hemorrhages or thrombosis. Although exercise has been shown to play a protective role in athletes, several factors may increase the risk of developing venous thromboembolism (VTE), including hemoconcentration induced by exertion, immobilization following sports injuries, frequent long-distance flights, dehydration, and the use of oral contraceptives in female athletes. Biomarkers such as D-dimer, Factor VIII, thrombin generation, inflammatory cytokines, and leukocyte count are involved in the diagnosis of deep vein thrombosis (DVT), although their interpretation is complex and may indicate the presence of other conditions such as infections, inflammation, and heart disease. Therefore, the identification of biomarkers with high sensitivity and specificity is needed for the screening and early diagnosis of thromboembolism. Recent evidence about the correlation between the intensity of physical activity and VTE is divergent, whereas the repeated gestures in sports such as baseball, hockey, volleyball, swimming, wrestling, or, on the other hand, soccer players, runners, and martial art training represent a risk factor predisposing to the onset of upper and lower DVT. Anticoagulant therapy is the gold standard, reducing the risk of serious complications such as pulmonary embolism. The aim of this review is to provide a general overview about the interplay between physical exercise and the risk of thromboembolism in athletes, focusing on the main causes of thrombosis in professional athletes and underlying the need to identify new markers and therapies that can represent a valid tool for safeguarding the athlete’s health. Full article
(This article belongs to the Special Issue Advances in Thrombotic Disorders and Antithrombotic Treatments)
14 pages, 3003 KiB  
Article
Stable and Thin-Polymer-Based Modification of Neurovascular Stents with 2-Methacryloyloxyethyl Phosphorylcholine Polymer for Antithrombogenicity
by Naoki Inuzuka, Yasuhiro Shobayashi, Satoshi Tateshima, Yuya Sato, Yoshio Ohba, Kazuhiko Ishihara and Yuji Teramura
Bioengineering 2024, 11(8), 833; https://doi.org/10.3390/bioengineering11080833 - 15 Aug 2024
Viewed by 703
Abstract
The advent of intracranial stents has revolutionized the endovascular treatment of cerebral aneurysms. The utilization of stents has rendered numerous cerebral aneurysm amenable to endovascular treatment, thereby obviating the need for otherwise invasive open surgical options. Stent placement has become a mainstream approach [...] Read more.
The advent of intracranial stents has revolutionized the endovascular treatment of cerebral aneurysms. The utilization of stents has rendered numerous cerebral aneurysm amenable to endovascular treatment, thereby obviating the need for otherwise invasive open surgical options. Stent placement has become a mainstream approach because of its safety and efficacy. However, further improvements are required for clinically approved devices to avoid the frequent occurrence of thrombotic complications. Therefore, controlling the thrombotic complications associated with the use of devices is of significant importance. Our group has developed a unique stent coated with a 2-methacryloyloxyethyl phosphorylcholine (MPC)-based polymer. In this study, the surface characteristics of the polymer coating were verified using X-ray photoelectron spectroscopy and atomic force microscopy. Subsequently, the antithrombotic properties of the coating were evaluated by measuring platelet count and thrombin–antithrombin complex levels of whole human blood after 3 h of incubation in a Chandler loop model. Scanning electron microscopy was utilized to examine thrombus formation on the stent surface. We observed that MPC polymer-coated stents significantly reduced thrombus formation as compared to bare stents and several clinically approved devices. Finally, the coated stents were further analyzed by implanting them in the internal thoracic arteries of pigs. Angiographic imaging and histopathological examinations that were performed one week after implantation revealed that the vascular lumen was well maintained and coated stents were integrated within the vascular endothelium without inducing adverse effects. Thus, we demonstrated the efficacy of MPC polymer coating as a viable strategy for avoiding the thrombotic risks associated with neurovascular stents. Full article
Show Figures

Figure 1

Figure 1
<p>Representative atomic force microscopic image of an Ni-Ti flat substrate coated with 2-methacryloyloxyethyl phosphorylcholine (MPC) polymer. (<b>a</b>) Bare Ni-Ti flat substrate; (<b>b</b>) MPC polymer-coated Ni-Ti flat substrate. Swelled MPC polymer was detected over the entire surface in the measurements conducted in phosphate-buffered saline.</p>
Full article ">Figure 2
<p>MPC polymer deposition between narrow struts after the coating with Lipidure<sup>®</sup> CM5206 via hydrophobic interaction. Visible MPC polymer can be observed between the narrow struts, in which the deposition forms bridges after air drying of the polymer solution. Such deposition can lead to the potential risk of distal embolism in neurovascular applications.</p>
Full article ">Figure 3
<p>Blood compatibility test of stents using the Chandler loop model with whole human blood. (<b>a</b>) Schematic illustration of the Chandler loop model. In this model, the stent is placed opposite the connector in the tube, and fresh whole blood (2 mL) is poured into the tube, while the remaining volume is filled with air. Here, a bare stent, MPC polymer-coated stent, Neuroform Atlas<sup>®</sup>, and Enterprise<sup>®</sup> were tested. (<b>b</b>) Relative platelet counts in the whole blood following the blood experiment using the Chandler loop model. The initial platelet count was set as the baseline (100%) when blood sampling was performed immediately after the commencement of the test. The figure shows the percentage of platelets remaining in the blood after 3 h of contact with the stent. Each stent showed a statistically significant difference compared to the bare stent (*** <span class="html-italic">p</span> &lt; 0.005, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05). (<b>c</b>) Relative thrombin–antithrombin complex (TAT) concentration in the blood after 3 h of incubation. The initial TAT concentration was set as the baseline (100%), which was measured on blood samples collected immediately after the commencement of the test. Blood samples from three different experiments (<span class="html-italic">n</span> = 3) were analyzed, and the data are expressed as the mean percentage ± standard deviation. The stent coated with MPC polymer significantly suppressed the increase in TAT compared to the bare stent (** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3 Cont.
<p>Blood compatibility test of stents using the Chandler loop model with whole human blood. (<b>a</b>) Schematic illustration of the Chandler loop model. In this model, the stent is placed opposite the connector in the tube, and fresh whole blood (2 mL) is poured into the tube, while the remaining volume is filled with air. Here, a bare stent, MPC polymer-coated stent, Neuroform Atlas<sup>®</sup>, and Enterprise<sup>®</sup> were tested. (<b>b</b>) Relative platelet counts in the whole blood following the blood experiment using the Chandler loop model. The initial platelet count was set as the baseline (100%) when blood sampling was performed immediately after the commencement of the test. The figure shows the percentage of platelets remaining in the blood after 3 h of contact with the stent. Each stent showed a statistically significant difference compared to the bare stent (*** <span class="html-italic">p</span> &lt; 0.005, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05). (<b>c</b>) Relative thrombin–antithrombin complex (TAT) concentration in the blood after 3 h of incubation. The initial TAT concentration was set as the baseline (100%), which was measured on blood samples collected immediately after the commencement of the test. Blood samples from three different experiments (<span class="html-italic">n</span> = 3) were analyzed, and the data are expressed as the mean percentage ± standard deviation. The stent coated with MPC polymer significantly suppressed the increase in TAT compared to the bare stent (** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>Representative scanning electron microscopic (SEM) images of four different stent samples at 3 h in the Chandler loop model. (<b>a</b>) Bare stent; (<b>b</b>) MPC polymer-coated stent; (<b>c</b>) Neuroform Atlas<sup>®</sup>; (<b>d</b>) Enterprise<sup>®</sup>. The yellow box is an image that shows a 3x magnification of the central part of the image.</p>
Full article ">Figure 5
<p>Thrombogenicity scoring of stents based on analysis. For each stent, SEM images were captured at 20 predefined locations, ensuring comprehensive coverage. Scores were assigned to each stent at these 20 distinct locations based on the criteria established in <a href="#bioengineering-11-00833-t001" class="html-table">Table 1</a>. MPC polymer-coated surfaces had significantly reduced thrombogenicity scores compared to other stents, indicating improved antithrombotic performance (**** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 6
<p>In vivo evaluation of an MPC polymer-coated stent that was implanted in the internal thoracic artery of a pig. (<b>a</b>) Representative angiographic image 1 week after stent implantation. The numbers in the figure represent the respective measurement points: 1 indicates the proximal stent diameter, 2 indicates the distal stent diameter, and 3 indicates the stent length. The corresponding measurements are as follows: 1 = 1.8 mm, 2 = 1.8 mm, 3 = 19 mm.; (<b>b</b>) representative image of hematoxylin and eosin staining of a sliced section from the stent-implanted vessels. Black dots represent struts of stents.</p>
Full article ">
9 pages, 2475 KiB  
Article
Novel Flowable Hemostatic Agent ActiClot: Efficacy and Safety Assessment in Rat and Porcine Models
by Hee-Jung Kim, Su-Kyoung Lee, Yun-Jeh Ko, Soo-Hyeon Jeon, Eun-Jin Kim, Oh-Hyeong Kwon and Yang-Hyun Cho
J. Clin. Med. 2024, 13(16), 4770; https://doi.org/10.3390/jcm13164770 - 14 Aug 2024
Viewed by 443
Abstract
Background/Objectives: This study evaluated the hemostatic performance and safety of ActiClot (ATC), a new flowable hemostatic agent, through in vivo tests. Methods: ATC was compared with the commercially available FLOSEAL®. ATC consists of carboxymethyl starch, thrombin, and sorbitol powders [...] Read more.
Background/Objectives: This study evaluated the hemostatic performance and safety of ActiClot (ATC), a new flowable hemostatic agent, through in vivo tests. Methods: ATC was compared with the commercially available FLOSEAL®. ATC consists of carboxymethyl starch, thrombin, and sorbitol powders in Syringe I, and a calcium chloride solution in Syringe II. In vivo evaluation used rat liver bleeding and porcine heart bleeding models. Safety was assessed using a rat subcutaneous implantation model. Results: ATC significantly reduced hemostasis time (70.00 ± 7.35 s) compared to gauze control (240.63 ± 32.31 s) in the rat liver model, showing a 70% reduction. There was no significant difference between ATC and FLOSEAL® (58.75 ± 13.42 s). In the porcine heart model, both agents achieved 100% hemostasis within 3 min, with no significant difference in success rates within 2 min (ATC 87.5%, FLOSEAL® 75%). The gauze control group failed in all tests. The rat subcutaneous implantation model showed no visual ATC observation after 48 h, indicating biocompatibility, with no inflammation observed. Conclusions: ATC demonstrated effective hemostatic performance similar to FLOSEAL® in two in vivo models, with faster hemostasis in the rat liver model. It also showed excellent safety and biocompatibility, indicating its potential for surgical and emergency bleeding control. Full article
(This article belongs to the Section General Surgery)
Show Figures

Figure 1

Figure 1
<p>The ActiClot consists of two syringes (Syringe I and II, 15 mL) (<b>a</b>) and a connection hub. Syringe I contains carboxymethyl starch, thrombin, and sorbitol powders. Syringe II contains a calcium chloride solution. The ActiClot can be used simply by connecting and mixing both syringes using a hub (<b>b</b>) and a nozzle (<b>c</b>) without the process of reconstitution of thrombin or refilling it into another syringe.</p>
Full article ">Figure 2
<p>In vivo hemostatic efficacy on rat liver injury model. (<b>a</b>) Hemostasis time measurement of the gauze, ActiClot, and FLOSEAL<sup>®</sup> groups and additional bleeding check results after hemostatic agent removal; (<b>b</b>) gauze, (<b>c</b>) ActiClot, and (<b>d</b>) FLOSEAL<sup>®</sup> groups. (n = 8, ** <span class="html-italic">p</span> &lt; 0.05, NS means not significant)<b>.</b></p>
Full article ">Figure 3
<p>In vivo hemostasis confirmation after 2 min of compression in the porcine heart muscle incision model. The left (<b>a</b>–<b>c</b>) and right (<b>d</b>–<b>f</b>) ventricles of the porcine heart treated with (<b>a</b>,<b>d</b>) cotton gauze, (<b>b</b>,<b>e</b>) ActiClot, and (<b>c</b>,<b>f</b>) FLOSEAL<sup>®</sup>. The yellow arrow indicates the hemostatic condition after applying hemostatic methods.</p>
Full article ">Figure 4
<p>In vivo safety evaluation of the ActiClot matrix in the rat model. (<b>a</b>) Photographs of the site after subcutaneous implantation and (<b>b</b>) the photomicrographs H&amp;E, ((<b>b</b>), pink; cytoplasm, dark blue; nuclei), Masson’s trichrome ((<b>c</b>), dark red; keratin and muscle fibers, pink; cytoplasm, light blue; collagen, dark blue; nuclei) staining of the ActiClot implanted tissue.</p>
Full article ">
12 pages, 2220 KiB  
Article
Lung EC-SOD Overexpression Prevents Hypoxia-Induced Platelet Activation and Lung Platelet Accumulation
by Daniel Colon Hidalgo, Mariah Jordan, Janelle N. Posey, Samuel D. Burciaga, Thi-Tina N. Nguyen, Christina Sul, Caitlin V. Lewis, Cassidy Delaney and Eva S. Nozik
Antioxidants 2024, 13(8), 975; https://doi.org/10.3390/antiox13080975 - 10 Aug 2024
Viewed by 751
Abstract
Pulmonary hypertension (PH) is a progressive disease marked by pulmonary vascular remodeling and right ventricular failure. Inflammation and oxidative stress are critical in PH pathogenesis, with early pulmonary vascular inflammation preceding vascular remodeling. Extracellular superoxide dismutase (EC-SOD), a key vascular antioxidant enzyme, mitigates [...] Read more.
Pulmonary hypertension (PH) is a progressive disease marked by pulmonary vascular remodeling and right ventricular failure. Inflammation and oxidative stress are critical in PH pathogenesis, with early pulmonary vascular inflammation preceding vascular remodeling. Extracellular superoxide dismutase (EC-SOD), a key vascular antioxidant enzyme, mitigates oxidative stress and protects against inflammation and fibrosis in diverse lung and vascular disease models. This study utilizes a murine hypobaric hypoxia model to investigate the role of lung EC-SOD on hypoxia-induced platelet activation and platelet lung accumulation, a critical factor in PH-related inflammation. We found that lung EC-SOD overexpression blocked hypoxia-induced platelet activation and platelet accumulation in the lung. Though lung EC-SOD overexpression increased lung EC-SOD content, it did not impact plasma extracellular SOD activity. However, ex vivo, exogenous extracellular SOD treatment specifically blunted convulxin-induced platelet activation but did not blunt platelet activation with thrombin or ADP. Our data identify platelets as a novel target of EC-SOD in response to hypoxia, providing a foundation to advance the understanding of dysregulated redox signaling and platelet activation in PH and other chronic hypoxic lung diseases. Full article
(This article belongs to the Special Issue Role of Redox in Pulmonary Vascular Diseases)
Show Figures

Figure 1

Figure 1
<p>Platelet counts are similar across wildtype and EC-SOD overexpressing mice in normoxia and hypoxia, but platelet activation is attenuated by EC-SOD overexpression. (<b>A</b>) Using the Heska HT5 hematologic analyzer, total platelet counts were obtained from blood samples acquired from WT and lung EC-SOD overexpressing mice in both normoxia and hypoxia. N = 8–13. (<b>B</b>) P-selectin (Thermofisher) and (<b>C</b>) αIIBβ3 (Emfret) expression were assessed by flow cytometry in freshly isolated platelets from wildtype and lung EC-SOD overexpressing mice. N = 8, * <span class="html-italic">p</span> &lt; 0.05 by two-way ANOVA. (<b>D</b>) Platelet-leukocyte aggregation was measured by co-expression of CD41-BV421 (BioLegend) and CD45-APC (BioLegend) antibodies. N = 4–9, * <span class="html-italic">p</span> &lt; 0.05 by two-way ANOVA. (<b>E</b>) ELISA measuring PF4 (Abcam), a marker of platelet chemokine release, in platelet-poor plasma. N = 6–9, * <span class="html-italic">p</span> &lt; 0.05 by two-way ANOVA. WT = Wildtype, Tg+ = mice overexpressing lung EC-SOD, NMX = normoxia, HPX = hypoxia, PF4 = platelet factor 4.</p>
Full article ">Figure 2
<p>Hypoxia leads to increases in platelet infiltration and lung PF4 levels, which is prevented by EC-SOD overexpression. (<b>A</b>) Percentage of positive platelet marker CD41 (BioLegend) pixels in the lungs of wildtype mice and mice overexpressing lung EC-SOD in normoxia and hypoxia. N = 6–8, * <span class="html-italic">p</span> &lt; 0.05 by two-way ANOVA. (<b>B</b>) Lung PF4 ELISA showing the concentration of PF4 in the lungs of wildtype mice and mice overexpressing lung EC-SOD exposed to normoxia or hypoxia. N = 5–10, * <span class="html-italic">p</span> &lt; 0.05 by two-way ANOVA. (<b>C</b>) Representative histology (20x) images of CD41 (BioLegend) lung immunohistochemistry of the lungs of wildtype mice and mice overexpressing lung EC-SOD in normoxia and hypoxia. WT = Wildtype, Tg+ = mice overexpressing lung EC-SOD, NMX = normoxia, HPX = hypoxia, PF4 = platelet factor 4.</p>
Full article ">Figure 3
<p>Plasma SOD activity was similar between mice overexpressing lung EC-SOD (Tg) mice and wildtype mice. (<b>A</b>) Plasma SOD activity in normoxic and 3-day hypoxic WT and Tg mice using a colorimetric assay (Dojindo). N = 6. (<b>B</b>) Representative western blot of lung EC-SOD expression along with housekeeping protein, Vinculin in WT mice and mice with lung overexpression of EC-SOD. (<b>C</b>) Densitometry of lung EC-SOD expression. N = 6, * <span class="html-italic">p</span> &lt; 0.05 by two-way ANOVA. WT = Wildtype, Tg = mice overexpressing lung EC-SOD, NMX = normoxia, HPX = hypoxia.</p>
Full article ">Figure 4
<p>Ex vivo superoxide dismutase administration prevents convulxin-induced platelet activation. Freshly isolated platelets from wildtype mice were exposed to platelet agonists thrombin (0.1 U/mL, Chrono-log Corporation), adenosine diphosphate (ADP) (5 uM, Chrono-log Corporation) and Convulxin (350 ng/mL, Cayman Chemical), in the presence or absence of superoxide dismutase (SOD, 300 U/mL, Sigma Aldrich). Platelet activation was measured by expression (<b>A</b>) P-selectin and (<b>B</b>) activated αIIbβ3 via flow cytometry. N = 8, * <span class="html-italic">p</span> &lt; 0.05. SOD = superoxide dismutase, BL = baseline, THR = thrombin, ADP = adenosine diphosphate, CONV = Convulxin.</p>
Full article ">
0 pages, 3047 KiB  
Article
Investigation of a Serine Protease Inhibitor Active in the Infectious Stage of the Human Liver Fluke Opisthorchis viverrini
by Rosnanee Salang, Wansika Phadungsil, Amornrat Geadkaew-Krenc and Rudi Grams
Pathogens 2024, 13(8), 678; https://doi.org/10.3390/pathogens13080678 - 10 Aug 2024
Viewed by 627
Abstract
Serine protease inhibitors (serpins) participate in the regulation of inflammation, blood coagulation, and complement activation in humans. This research aimed to identify and characterize such inhibitors of the human liver fluke Opisthorchis viverrini. Parasite proteins that might contribute to the modulation of [...] Read more.
Serine protease inhibitors (serpins) participate in the regulation of inflammation, blood coagulation, and complement activation in humans. This research aimed to identify and characterize such inhibitors of the human liver fluke Opisthorchis viverrini. Parasite proteins that might contribute to the modulation of host physiology are of particular interest, especially as chronic opisthorchiasis increases the risk of developing biliary cancer. BLAST was used to find hypothetical serpins predicted from the parasite genome data. RNA extraction and reverse transcriptase PCR were used to isolate a serpin cDNA and to determine developmental transcript abundance. The evolutionary relation to other trematode serpins was revealed by phylogenetic analysis. Recombinant serpin was expressed in Escherichia coli and used to test the immunoreactivity of human opisthorchiasis sera and the inhibition of human serine proteases. A substantial serpin family with high sequence divergence among the members was found in the genus Opisthorchis. A serpin, different from previously analyzed trematode serpins, was cloned. The transcript was only detected in metacercariae and newly excysted juveniles. Human opisthorchiasis sera showed statistically significant reactivity to recombinant serpin. The serpin caused moderate inhibition of thrombin and low inhibition of kallikrein and chymotrypsin. This parasite serpin could be further evaluated as a diagnostic tool for early infection. Kallikrein and thrombin are involved in fibrinolysis; therefore, further research should explore the effects of the parasite serpin on this process. Full article
(This article belongs to the Section Parasitic Pathogens)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Multiple sequence alignment of <span class="html-italic">S. haematobium</span> SPI (UniProt: Q26502), <span class="html-italic">O. viverrini</span> SIS (UniProt: A0A075AEP2), <span class="html-italic">C. sinensis</span> SIS (UniProt: A0A419PW56), and <span class="html-italic">O. felineus</span> SIS (UniProt: A0A4S2MGE0). The α-helical and β-strand regions of <span class="html-italic">Sh</span>SPI and <span class="html-italic">Ov</span>SIS based on their AlphaFold native state models [<a href="#B23-pathogens-13-00678" class="html-bibr">23</a>] are indicated at the top and bottom, respectively. The numbering of the secondary structures follows α<sub>1</sub>-antitrypsin [<a href="#B24-pathogens-13-00678" class="html-bibr">24</a>]. The region of the reactive center loop (RCL) with the P1 residue is indicated by a gray bar. Strand 4A in the RCL was manually added as it forms and integrates into β-sheet A after cleavage at P1. Sequence conservation using the BLOSUM 62 similarity matrix and <span class="html-italic">Sh</span>SPI as the reference sequence is indicated by color shading (fully conserved: purple background, ≥50% conserved: blue background, and similar: red background). The graphical representation of the aligned sequences was created using TEXshade v1.28 [<a href="#B25-pathogens-13-00678" class="html-bibr">25</a>].</p>
Full article ">Figure 2
<p><span class="html-italic">Ov</span>SIS structure model predicted in AlphaFold [<a href="#B23-pathogens-13-00678" class="html-bibr">23</a>] and obtained from the AlphaFold Protein Structure Database [<a href="#B26-pathogens-13-00678" class="html-bibr">26</a>]. The <span class="html-italic">Ov</span>SIS model with the three β-sheets A (red), B (green), and C (yellow), RCL (dark blue), and threonine 365 in the P1 position is shown on the left. The <span class="html-italic">Ov</span>SIS model prediction confidence from high to low is indicated by red to blue shading, respectively. Overlays with <span class="html-italic">Sh</span>SPI (PDB: 3STO) and <span class="html-italic">Sm</span>SPI (PDB: 6SSV) demonstrate structural conservation between the three serpins. Graphical representations were created using ChimeraX [<a href="#B27-pathogens-13-00678" class="html-bibr">27</a>].</p>
Full article ">Figure 3
<p>(<b>a</b>) Phylogenetic tree of trematode serpins specified by their UniProt accession numbers. The three previously described serpins of <span class="html-italic">C. sinensis</span> are indicated with a trailing •. Schistosome serpins are shown in blue, with the SPI in bold lettering. <span class="html-italic">Ov</span>SIS and its orthologs are shown in red. <span class="html-italic">Opisthorchis</span>/<span class="html-italic">Clonorchis</span> SIS is not orthologous to schistosome SIP. (<b>b</b>) Sequence conservation of the reactive center loop in evolutionarily close serpins (specified by their UniProt accession numbers) in the species <span class="html-italic">O. viverrini</span> (Ov), <span class="html-italic">O. felineus</span> (Of), <span class="html-italic">C. sinensis</span> (Cs), <span class="html-italic">S. haematobium</span> (Sh), <span class="html-italic">S. japonicum</span> (Sj), and <span class="html-italic">S. mansoni</span> (Sm). The P1 position in the protease interaction is indicated. The color shading indicates fully conserved: purple background, ≥50% conserved: blue background, and similar: red background.</p>
Full article ">Figure 4
<p>(<b>a</b>) RT-PCR products resolved in 0.8% agarose gels. M: marker, 1: newly excysted juveniles, 2: 2-week-old juveniles, and 3: mature parasites. <span class="html-italic">O. viverrini</span> β-actin (292 bp) was used as a positive control. A 640 bp <span class="html-italic">Ov</span>SIS cDNA was only produced with RNA of newly excysted juveniles. (<b>b</b>) Micrograph of a newly excysted <span class="html-italic">O. viverrini</span> juvenile (top) and a metacercaria (bottom), the developmental stages in which <span class="html-italic">Ov</span>SIS is present. The dark granule-filled excretory bladder, which can also be seen through the cyst wall, is the most prominent. Other outstanding morphological features are the oral and ventral suckers. It is thought that parasite-secreted serine proteases participate in the release of the encysted juveniles and that cognate serpins regulate their excess activity.</p>
Full article ">Figure 5
<p>(<b>a</b>) SDS-PAGE showing pre-induced (1) and IPTG-induced bacterial proteins (2) and affinity-purified recombinant <span class="html-italic">Ov</span>SIS (3). The protein migrates at the expected 46 kDa molecular weight. (<b>b</b>) Absorption values of two groups of human sera against recombinant <span class="html-italic">Ov</span>SIS obtained by indirect ELISA. n: normal sera (<span class="html-italic">n</span> = 10). i: sera of <span class="html-italic">O. viverrini</span>-infected individuals (<span class="html-italic">n</span> = 30). The minimum, 25th percentile, median, 75th percentile, and maximum are indicated in the box plots. The star symbols indicate that all infected sera were statistically significantly different (*** <span class="html-italic">p</span> &lt; 0.05) from the normal sera. (<b>c</b>) Human infection with <span class="html-italic">O. viverrini</span>. The metacercariae are released from infested undercooked fish in the stomach. The acidic environment activates the excystment of the juveniles. The newly excysted juveniles are released with the stomach content into the duodenum and migrate via the sphincter of Oddi into the biliary system (green).</p>
Full article ">Figure 6
<p>Inhibition kinetics of <span class="html-italic">Ov</span>SIS against human serine proteases. Proteases without inhibitors are indicated in black, proteases incubated with recombinant <span class="html-italic">Ov</span>SIS are shown in red, and proteases incubated with 20 mM PMSF are shown in teal. After 30 min of preincubation with 0.5 µM recombinant <span class="html-italic">Ov</span>SIS and 30 min of incubation with the substrate, the activities of 1 nM chymotrypsin, 1 nM kallikrein, and 0.1 U thrombin were lowered to 69.21, 66.76, and 48.84%, respectively. The enzyme substrates are listed in <a href="#pathogens-13-00678-t001" class="html-table">Table 1</a>.</p>
Full article ">
87 pages, 3719 KiB  
Review
Beyond Anticoagulation: A Comprehensive Review of Non-Vitamin K Oral Anticoagulants (NOACs) in Inflammation and Protease-Activated Receptor Signaling
by Shirin Jannati, Rajashree Patnaik and Yajnavalka Banerjee
Int. J. Mol. Sci. 2024, 25(16), 8727; https://doi.org/10.3390/ijms25168727 - 10 Aug 2024
Viewed by 739
Abstract
Non-vitamin K oral anticoagulants (NOACs) have revolutionized anticoagulant therapy, offering improved safety and efficacy over traditional agents like warfarin. This review comprehensively examines the dual roles of NOACs—apixaban, rivaroxaban, edoxaban, and dabigatran—not only as anticoagulants, but also as modulators of inflammation via protease-activated [...] Read more.
Non-vitamin K oral anticoagulants (NOACs) have revolutionized anticoagulant therapy, offering improved safety and efficacy over traditional agents like warfarin. This review comprehensively examines the dual roles of NOACs—apixaban, rivaroxaban, edoxaban, and dabigatran—not only as anticoagulants, but also as modulators of inflammation via protease-activated receptor (PAR) signaling. We highlight the unique pharmacotherapeutic properties of each NOAC, supported by key clinical trials demonstrating their effectiveness in preventing thromboembolic events. Beyond their established anticoagulant roles, emerging research suggests that NOACs influence inflammation through PAR signaling pathways, implicating factors such as factor Xa (FXa) and thrombin in the modulation of inflammatory responses. This review synthesizes current evidence on the anti-inflammatory potential of NOACs, exploring their impact on inflammatory markers and conditions like atherosclerosis and diabetes. By delineating the mechanisms by which NOACs mediate anti-inflammatory effects, this work aims to expand their therapeutic utility, offering new perspectives for managing inflammatory diseases. Our findings underscore the broader clinical implications of NOACs, advocating for their consideration in therapeutic strategies aimed at addressing inflammation-related pathologies. This comprehensive synthesis not only enhances understanding of NOACs’ multifaceted roles, but also paves the way for future research and clinical applications in inflammation and cardiovascular health. Full article
(This article belongs to the Special Issue New Trends in Diabetes, Hypertension and Cardiovascular Diseases 2.0)
Show Figures

Figure 1

Figure 1
<p>The current model of the blood coagulation cascade, depicting NOACs’ mechanism of action. There are two pathways, the intrinsic pathway and the extrinsic pathway. These multicomponent processes are illustrated as enzymes, inhibitors, zymogens, or complexes. On injury to the vessel wall, tissue factor, the cofactor for the extrinsic tenase complex, is exposed to circulating FVIIa and forms the extrinsic tenase. FIX and FX are converted to their serine proteases FIXa and FXa, which then form the intrinsic tenase and the prothrombinase complexes, respectively. The combined actions of the intrinsic and extrinsic tenase and the prothrombinase complexes lead to an explosive burst of the enzyme thrombin (IIa). In addition to its multiple procoagulant roles, thrombin also acts in an anticoagulant capacity when combined with the cofactor thrombomodulin in the protein Case complex. The product of the protein Case reaction, activated protein C (APC), inactivates the cofactors FVa and FVIIIa. The cleaved species, FVai and FVIIIai, no longer support the respective procoagulant activities. Once thrombin is generated through procoagulant mechanisms, thrombin cleaves fibrinogen (releasing fibrinopeptide A and B [FPA and FPB]), as well as activating FXIII to form a cross-linked fibrin clot. Thrombin–thrombomodulin also activates thrombin activate-able fibrinolysis inhibitor, which slows fibrin degradation by plasmin. The procoagulant response is downregulated by the stoichiometric inhibitor tissue factor pathway inhibitor (TFPI) and antithrombin III (AT-III). TFPI serves to attenuate the activity of the extrinsic tenase trigger of coagulation. AT-III directly inhibits thrombin, FIXa, and FXa. The accessory pathway provides an alternate route for the generation of FIXa. Thrombin has also been shown to activate FXI. The fibrin clot is eventually degraded by plasmin, yielding soluble fibrin peptides. Factor Xa inhibitors (apixaban, edoxaban, and rivaroxaban) act by binding to the active site of factor Xa, inhibiting the conversion of prothrombin to thrombin, the final enzyme in the coagulation cascade. Dabigatran, conversely, functions as a direct thrombin inhibitor. It binds with high affinity to the active site of thrombin, inhibiting its ability to convert fibrinogen to fibrin, thereby preventing clot formation.</p>
Full article ">Figure 2
<p>Schematic representation of structural domains of FXa and binding sites of FXa inhibitors. A schematic representation of FXa protein structural domains and the location of the binding sites of FXa inhibitors (apixaban, edoxaban, and rivaroxaban) are depicted. (<b>A</b>) illustrates the domain organization of FXa, highlighting the serine protease domain in the heavy chain (indicated by green) where the binding site is located. (<b>B</b>–<b>D</b>) depict the structures of apixaban, edoxaban, and rivaroxaban, respectively, as obtained from the Protein Data Bank, indicating their binding sites with FXa, which exhibit enzyme kinetics similar to competitive inhibitors.</p>
Full article ">Figure 3
<p>Schematic representation of PAR1- and PAR2-mediated signal transduction. PAR1 and PAR2 are G protein-coupled receptors that can be activated by thrombin and FXa, initiating a cascade of cellular responses. Upon cleavage, PAR1/2 interact with different G proteins like Gα<sub>i</sub>, Gα<sub>12/13</sub>, Gα<sub>s</sub>, and Gα<sub>q</sub>. Gα<sub>12/13</sub> leads to Ras homolog family member A (RhoA) activation, via Rho guanine nucleotide exchange factors (RhoGEFs) influencing cell hypertrophy. Gα<sub>q</sub> activates phospholipase C-β, generating second messengers that trigger calcium release and Protein Kinase C (PKC) activation. PKC can further activate the nuclear factor kappa B (NF-κB) signaling pathway to upregulate production of SRY-box transcription factor 4 (SOX4) and A disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5). Gα<sub>i</sub> can inhibit adenylate cyclase (AC) to regulate downstream cAMP, whereas Gα<sub>s</sub> can increase cAMP. β-arrestin can activate the ERK1/2 signaling pathway but exhibits inhibitory effects on PKC and calcium release. Anticoagulants like dabigatran (thrombin inhibitor) and apixaban, edoxaban, and rivaroxaban (FXa inhibitors) can potentially disrupt this signaling by preventing PAR activation. Ultimately, these signal transduction pathways can trigger physiological changes like inflammatory and immune responses, cell hypertrophy, and cell migration.</p>
Full article ">Figure 4
<p>Schematic representation of PAR4-mediated signal transduction. PAR4 can activate signaling pathways involving Gα<sub>12/13</sub> and Gα<sub>q</sub>. Gα<sub>12/13</sub> prompts RhoGEFs to activate RhoA, while Gα<sub>q</sub> -Phospholipase C- β (PLC- β) leads to downstream effects such as upregulation of inositol triphosphate (IP3) and diacylglycerol (DAG), resulting in calcium alterations and PKC upregulation, which ultimately leads to activation of the NF-κB signaling pathway. Additionally, β-arrestin can facilitate ERK1/2 phosphorylation but has inhibitory effects on PKC. Thrombin, known for its ability to cleave PAR4, can influence these pathways, therefore dabigatran (thrombin inhibitor) can modulate signal transduction by attenuating thrombin’s effects. As seen, PAR4 activation can cause physiological alterations such as inflammatory and immune response, endothelial barrier dysfunction, and platelet activation.</p>
Full article ">Figure 5
<p>Proposed mechanism of apixaban’s modulatory effects on factor Xa and associated inflammatory signaling pathways in an osteoarthritic chondrocyte model. This illustration delineates the pathways through which apixaban may exert anti-inflammation in the context of osteoarthritis. Apixaban targets FXa, inhibiting its ability to bind and activate PAR2, which is represented by the red inhibitory line. This intervention most likely attenuates the downstream signaling cascades involved in OA pathophysiology: 1. PAR2 inhibition: The blockage of PAR2 activation by apixaban may ameliorate the downstream signaling events mediated by ERK1/2 that lead to the production of pro-inflammatory cytokines, such as TNF-α and IL-1β, potentially alleviating chronic pain associated with OA. 2. Cytokine modulation: The expected reduction in TNF-α and IL-1β due to apixaban’s action on FXa mitigates the upregulation of molecules like MCP-1, which are involved in monocyte recruitment and the NF-κB signaling pathway, both key contributors to inflammation and osteoclastogenesis. 3. Protein expression: The illustration also indicates the potential effects of apixaban on the expression of regulatory proteins, including SOX4 and ADAMTS5, and their impact on critical components like aggrecan, which is essential for cartilage integrity. 4. Chondrocyte integrity and bone health: By modulating these inflammatory and catabolic pathways, apixaban may help preserve chondrocyte integrity, mitigate the generation of reactive oxygen species (ROS), and contribute to maintaining joint health by potentially impacting the RANK/RANKL pathway, which is crucial for osteoclast activity and bone resorption.</p>
Full article ">
12 pages, 1428 KiB  
Communication
β-Adrenoceptor Agonists Attenuate Thrombin-Induced Impairment of Human Lung Endothelial Cell Barrier Function and Protect the Lung Vascular Barrier during Resuscitation from Hemorrhagic Shock
by Michelle Y. McGee, Ololade Ogunsina, Sadia N. Boshra, Xianlong Gao and Matthias Majetschak
Biomedicines 2024, 12(8), 1813; https://doi.org/10.3390/biomedicines12081813 - 9 Aug 2024
Viewed by 573
Abstract
β-adrenoceptor (β-AR) agonists are known to antagonize thrombin-induced impairment (TII) of bovine and ovine lung endothelial barrier function. The effects of adrenoceptor agonists and other vasoactive agents on human lung microvascular endothelial cell (HULEC-5a) barrier function upon thrombin exposure have not been studied. [...] Read more.
β-adrenoceptor (β-AR) agonists are known to antagonize thrombin-induced impairment (TII) of bovine and ovine lung endothelial barrier function. The effects of adrenoceptor agonists and other vasoactive agents on human lung microvascular endothelial cell (HULEC-5a) barrier function upon thrombin exposure have not been studied. Furthermore, it is unknown whether the in vitro effects of adrenoceptor agonists translate to lung protective effects in vivo. We observed that epinephrine, norepinephrine, and phenylephrine enhanced normal and prevented TII of HULEC-5a barrier function. Arginine vasopressin and angiotensin II were ineffective. α1B-, α2A/B-, and β1/2-ARs were detectable in HULEC-5a by RT-PCR. Propranolol but not doxazosin blocked the effects of all adrenoceptor agonists. Phenylephrine stimulated β2-AR-mediated Gαs activation with 13-fold lower potency than epinephrine. The EC50 to inhibit TII of HULEC-5a barrier function was 1.8 ± 1.9 nM for epinephrine and >100 nM for phenylephrine. After hemorrhagic shock and fluid resuscitation in rats, Evans blue extravasation into the lung increased threefold (p < 0.01 vs. sham). Single low-dose (1.8 μg/kg) epinephrine administration at the beginning of resuscitation had no effects on blood pressure and reduced Evans blue extravasation by 60% (p < 0.05 vs. vehicle). Our findings confirm the effects of β-adrenoceptor agonists in HULEC-5a and suggest that low-dose β-adrenoceptor agonist treatment protects lung vascular barrier function after traumatic hemorrhagic shock. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Therapeutics in Hemorrhagic Shock)
Show Figures

Figure 1

Figure 1
<p>Effects of adrenoceptor agonists, arginine vasopressin (AVP), and angiotensin II (ATII) on normal (<b>A</b>/<b>B</b>) and thrombin-stimulated (<b>C</b>/<b>D</b>) HULEC-5a barrier function. RFU (%): Relative fluorescence units (RFU) in % of the RFU measured in untreated cell monolayers after 1 h. Bars and error bars indicate mean ± SE. Open circles show the RFU (%) from duplicate measurements from each experiment. (<b>A</b>) HULEC-5a cell monolayers were treated with vehicle (ctrl.) or 5 μM of epinephrine (EPI), norepinephrine (NE), or phenylephrine (PE), and FITC-dextran permeability measured after 1 h and 4 h. <span class="html-italic">n</span> = 3 independent experiments. *: <span class="html-italic">p</span> &lt; 0.05 vs. vehicle-treated cells after 4 h. (<b>B</b>) HULEC-5a cell monolayers were treated with vehicle (ctrl.) or 5 μM of AVP or ATII and FITC-dextran permeability was measured after 1 h and 4 h. <span class="html-italic">n</span> = 3 independent experiments. (<b>C</b>) HULEC-5a cell monolayers were treated with 25 nM thrombin (+) or vehicle (−) plus vehicle drug (−) or 5 μM of EPI, NE or PE, and FITC-dextran permeability measured after 1 h and 4 h. <span class="html-italic">n</span> = 3 independent experiments. *: <span class="html-italic">p</span> &lt; 0.05 vs. cells treated with thrombin plus vehicle at the corresponding time point. #: <span class="html-italic">p</span> &lt; 0.05 vs. EPI and NE at the corresponding time point. (<b>D</b>) HULEC-5a cell monolayers were treated with 25 nM thrombin (+) or vehicle (−) plus vehicle drug (−) or 5 μM AVP or ATII, and FITC-dextran permeability was measured after 1 h and 4 h. <span class="html-italic">n</span> = 3 independent experiments.</p>
Full article ">Figure 2
<p>mRNA expression of adrenergic receptors in HULEC-5a cells detected by reverse transcription (RT)-PCR. Images show the agarose gel electrophoresis of cDNAs amplified by PCR and represent <span class="html-italic">n</span> = 3 independent experiments. M: molecular marker; RT: reverse transcription.</p>
Full article ">Figure 3
<p>Propranolol but not doxazosin antagonizes the effects of EPI and PE on thrombin-induced impairment of HULEC-5a barrier function. RFU (%): Relative fluorescence units (RFU) in % of the RFU measured in untreated cell monolayers after 1 h. Bars and error bars indicate mean ± SE. Open circles show the RFU (%) from duplicate measurements from each experiment. (<b>A</b>/<b>D</b>) HULEC-5a cell monolayers were treated with 25 nM thrombin (+) or vehicle (−) plus vehicle drug (−) or 5 μM of doxazosin (Dox, <b>A</b>) or propranolol (Prop, <b>D</b>) and FITC-dextran permeability measured after 1 h and 4 h. <span class="html-italic">n</span> = 3–4 independent experiments in duplicate. (<b>B</b>) HULEC-5a cell monolayers were treated with 25 nM thrombin (+) or vehicle (−) plus vehicle drug (−) or 5 μM of doxazosin and 5 μM of EPI, and FITC-dextran permeability measured after 1 h and 4 h. <span class="html-italic">n</span> = 3 independent experiments in duplicate. *: <span class="html-italic">p</span> &lt; 0.05 vs. cells treated with thrombin plus vehicle. (<b>C</b>) HULEC-5a cell monolayers were treated with 25 nM thrombin (+) or vehicle (−) plus vehicle drug (−) or 5 μM of doxazosin and 5 μM of PE, and FITC-dextran permeability measured after 1 h and 4 h. <span class="html-italic">n</span> = 3 independent experiments in duplicate. *: <span class="html-italic">p</span> &lt; 0.05 vs. cells treated with thrombin plus vehicle. (<b>E</b>) HULEC-5a cell monolayers were treated with 25 nM thrombin (+) or vehicle (−) plus vehicle drug (−) or 5 μM of propranolol and 5 μM of EPI, and FITC-dextran permeability measured after 1 h and 4 h. <span class="html-italic">n</span> = 3 independent experiments in duplicate. (<b>F</b>) HULEC-5a cell monolayers were treated with 25 nM thrombin (+) or vehicle (−) plus vehicle drug (−) or 5 μM of propranolol and 5 μM of PE, and FITC-dextran permeability measured after 1 h and 4 h. <span class="html-italic">n</span> = 3 independent experiments in duplicate.</p>
Full article ">Figure 4
<p>(<b>A</b>) β<sub>2</sub>-AR mediated Gαs activation monitored by BRET. HEK293T cells were transfected to express β<sub>2</sub>-AR together with GαsS-Rluc8, Gβ3, and Gγ9-GFP2. Activation of β<sub>2</sub>-AR by epinephrine (EPI) or phenylephrine (PE) leads to dissociation of GαsS from Gγ9 and thereby to the reduction of BRET. Data are the mean ± SE from <span class="html-italic">n</span> = 3 independent experiments in duplicate. (<b>B</b>/<b>C</b>) RFU (%): Relative fluorescence units (RFU) in % of the RFU measured in untreated cell monolayers after 1 h. Bars and error bars indicate mean ± SE. Open circles show the RFU (%) from duplicate measurements from each experiment. (<b>B</b>) HULEC-5a cell monolayers were treated with 25 nM thrombin (+) or vehicle (−) in the absence or presence of various concentrations of PE and FITC-dextran permeability measured after 1 h and 4 h. <span class="html-italic">n</span> = 3 independent experiments in duplicate. (<b>C</b>) HULEC-5a cell monolayers were treated with 25 nM thrombin (+) or vehicle (−) in the absence or presence of various concentrations of EPI and FITC-dextran permeability measured after 1 h and 4 h. <span class="html-italic">n</span> = 3 independent experiments in duplicate. *: <span class="html-italic">p</span> &lt; 0.05 vs. cells incubated with thrombin plus vehicle.</p>
Full article ">Figure 5
<p>A single low-dose EPI treatment protects the lung vascular barrier during resuscitation from hemorrhagic shock. Animals were hemorrhaged to a MAP of 30 mmHg for 60 min, followed by crystalloid fluid resuscitation with 1.5 times the shed blood volume. At t = 60 min, animals were treated with EPI (grey symbols, <span class="html-italic">n</span> = 4) or vehicle (open symbols, <span class="html-italic">n</span> = 4). Evans blue was injected at t = 90 min. Animals were euthanized at t = 120 min, and lungs were harvested. Data are the mean ± SE. (<b>A</b>) MAP, mmHg. (<b>B</b>) Hemorrhage volumes (mL/kg) and fluid resuscitation volumes (mL/kg). (<b>C</b>) Typical appearance of lungs from sham-treated animals (left) and animals after hemorrhage and fluid resuscitation with vehicle (hem vehicle, center) or EPI (hem EPI, right) treatment. (<b>D</b>/<b>E</b>) Quantification of Evans blue extravasation into the right (<b>D</b>) and left (<b>E</b>) lungs (μg/mg). Sham, <span class="html-italic">n</span> = 3. Hem vehicle, <span class="html-italic">n</span> = 4. Hem EPI, <span class="html-italic">n</span> = 4. Bars and error bars indicate mean ± SE. Open circles show the individual measurements. The levels of statistical significance are shown in the graphs.</p>
Full article ">
39 pages, 10887 KiB  
Article
Molecular Pro-Apoptotic Activities of Flavanone Derivatives in Cyclodextrin Complexes: New Implications for Anticancer Therapy
by Angelika A. Adamus-Grabicka, Pawel Hikisz, Artur Stepniak, Magdalena Malecka, Piotr Paneth, Joanna Sikora and Elzbieta Budzisz
Int. J. Mol. Sci. 2024, 25(15), 8488; https://doi.org/10.3390/ijms25158488 - 3 Aug 2024
Viewed by 549
Abstract
This study evaluates the antiproliferative potential of flavanones, chromanones and their spiro-1-pyrazoline derivatives as well as their inclusion complexes. The main goal was to determine the biological basis of molecular pro-apoptotic activities and the participation of reactive oxygen species (ROS) in shaping the [...] Read more.
This study evaluates the antiproliferative potential of flavanones, chromanones and their spiro-1-pyrazoline derivatives as well as their inclusion complexes. The main goal was to determine the biological basis of molecular pro-apoptotic activities and the participation of reactive oxygen species (ROS) in shaping the cytotoxic properties of the tested conjugates. For this purpose, changes in mitochondrial potential and the necrotic/apoptotic cell fraction were analyzed. Testing with specific fluorescent probes found that ROS generation had a significant contribution to the biological anticancer activity of complexes of flavanone analogues. TT (thrombin time), PT (prothrombin time) and APTT (activated partial tromboplastin time) were used to evaluate the influence of the compounds on the extrinsic and intrinsic coagulation pathway. Hemolysis assays and microscopy studies were conducted to determine the effect of the compounds on RBCs. Full article
(This article belongs to the Special Issue Advances in Drug Discovery and Synthesis)
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>). Changes in the mitochondrial potential (fluorescence level of JC-1 aggregates/JC-1 monomers) of MCF-7, MDA-MB-231, HCC38, Hec-1-A and Ishikawa treated with investigated chromanone analogues condensed with pyrazolines at IC<sub>50</sub>. The results are presented as percentages of controls; the values were calculated by comparing the fluorescence intensity ratio of the test sample and the control (100%). Results represent mean ± SEM of the data from three individual experiments, * <span class="html-italic">p</span> &lt; 0.05 vs. control (untreated cells). Designation of compounds on the chart: <b>1</b>; <b>3</b>; <b>5</b>. (<b>b</b>). Changes in the mitochondrial potential (fluorescence level of JC-1 aggregates/JC-1 monomers) of MCF-7, MDA-MB-231, HCC38, Hec-1-A and Ishikawa treated with investigated chromanone analogues at IC<sub>50</sub>. The microscopic images were captured at a magnification of 20× with a Nikon Eclipse Te200 microscope with a ZEISS Axiocam 208 color microscope camera using a Nikon LWD Ph1 DL 20 × 0.40 lens.</p>
Full article ">Figure 1 Cont.
<p>(<b>a</b>). Changes in the mitochondrial potential (fluorescence level of JC-1 aggregates/JC-1 monomers) of MCF-7, MDA-MB-231, HCC38, Hec-1-A and Ishikawa treated with investigated chromanone analogues condensed with pyrazolines at IC<sub>50</sub>. The results are presented as percentages of controls; the values were calculated by comparing the fluorescence intensity ratio of the test sample and the control (100%). Results represent mean ± SEM of the data from three individual experiments, * <span class="html-italic">p</span> &lt; 0.05 vs. control (untreated cells). Designation of compounds on the chart: <b>1</b>; <b>3</b>; <b>5</b>. (<b>b</b>). Changes in the mitochondrial potential (fluorescence level of JC-1 aggregates/JC-1 monomers) of MCF-7, MDA-MB-231, HCC38, Hec-1-A and Ishikawa treated with investigated chromanone analogues at IC<sub>50</sub>. The microscopic images were captured at a magnification of 20× with a Nikon Eclipse Te200 microscope with a ZEISS Axiocam 208 color microscope camera using a Nikon LWD Ph1 DL 20 × 0.40 lens.</p>
Full article ">Figure 2
<p>Fluorescence anisotropy of TMA-DPH probe in MCF-7, MDA-MB-231, HCC38, Ishikawa and Hec-1-A lines treated with investigated chromanone analogues condensed with pyrazolines at a concentration of IC<sub>50</sub>. The results are presented as percentages of controls; the results were calculated by comparing the fluorescence anisotropy intensity ratio of the test sample and the control (100%). Results represent mean ± SEM of the data from three individual experiments, * <span class="html-italic">p</span> &lt; 0.05 vs. control (untreated cells). Designation of compounds on the chart: <b>1</b>; <b>3</b>; <b>5</b>.</p>
Full article ">Figure 3
<p>Fluorescence anisotropy of DAUDA probe in MCF-7, MDA-MB-231, HCC38, Ishikawa and Hec-1-A lines treated with investigated chromanone analogues condensed with pyrazolines at a concentration of IC<sub>50</sub>. The results are presented as percentages of controls; the data were calculated by comparing the fluorescence anisotropy intensity ratio of the test sample and the control (100%). Results represent mean ± SEM of the data from three individual experiments, * <span class="html-italic">p</span> &lt; 0.05 vs. control (untreated cells). Designation of compounds on the chart: <b>1</b>; <b>3</b>; <b>5</b>.</p>
Full article ">Figure 4
<p>Induction of apoptotic and necrotic death of cancer cells of the MCF-7, MDA-MB-231, HCC38, Ishikawa and Hec-1-A lines treated with the test compounds at IC<sub>50</sub>. Fluorescence microscopy using double-staining with Hoechst 33258 fluorochromes and propidium iodide. * <span class="html-italic">p</span> &lt; 0.05 vs. control (PBS-treated cells). Designation of compounds on the chart: <b>1</b>; <b>3</b>; <b>5</b>.</p>
Full article ">Figure 5
<p>Induction of apoptotic and necrotic death of cancer cells of the MCF-7, MDA-MB-231, HCC38, Ishikawa and Hec-1-A lines treated with investigated compounds at IC50. Fluorescence microscopy using double-staining with Hoechst 33258 fluorochromes and propidium iodide. To visualize changes in cell death, microscopic images were captured with a Nikon Eclipse Te200 microscope with a ZEISS Axiocam 208 color microscope camera. Pictures were taken at a magnification of 20× with a Nikon LWD Ph1 DL 20 × 0.40 lens.</p>
Full article ">Figure 6
<p>Relative amounts of ROS (superoxide anion O<sub>2</sub><sup>•−</sup>; DHE probe) generated in MCF-7, MDA-MB-231, HCC38, Hec-1-A and Ishikawa lines treated with investigated chromanone analogues condensed with pyrazolines at a concentration of IC<sub>50</sub>. The results are presented as percentages of controls; these were calculated by comparing the fluorescence intensity ratio of the test sample and the control (100%). Results represent mean ± SEM of the data from three individual experiments, * <span class="html-italic">p</span> &lt; 0.05 vs. control (untreated cells). Designation of compounds on the chart: <b>1</b>; <b>3</b>; <b>5</b>.</p>
Full article ">Figure 7
<p>Relative amounts of ROS (hydrogen peroxide H<sub>2</sub>O<sub>2</sub>; H<sub>2</sub>DCFDA probe) generated in MCF-7, MDA-MB-231, HCC38, Hec-1-A and Ishikawa lines treated with investigated chromanone analogues condensed with pyrazolines at a concentration of IC<sub>50</sub>. The results are presented as percentages of controls; these were calculated by comparing the fluorescence intensity ratio of the test sample and the control (100%). Results represent mean ± SEM of the data from three individual experiments, * <span class="html-italic">p</span> &lt; 0.05 vs. control (untreated cells). Designation of compounds on the chart: <b>1</b>; <b>3</b>; <b>5</b>.</p>
Full article ">Figure 8
<p>Relative amounts of RNS (nitric oxide NO; DAF-FM diacetate probe) generated in MCF-7, MDA-MB-231, HCC38, Hec-1-A and Ishikawa lines treated with investigated chromanone analogues condensed with pyrazolines at a concentration of IC<sub>50</sub>. The results are presented as percentages of controls; these were calculated by comparing the fluorescence intensity ratio of the test sample and the control (100%). Results represent mean ± SEM of the data from three individual experiments, * <span class="html-italic">p</span> &lt; 0.05 vs. control (untreated cells). Designation of compounds on the chart: <b>1</b>; <b>3</b>; <b>5</b>.</p>
Full article ">Figure 9
<p>Comparison of IC50 concentration values for individual compounds in cancer lines MCF-7, MDA-MB-231, HCC38, Hec-1-A and Ishikawa in experimental variants without or with one hour pre-incubation with antioxidants: N-acetylcysteine (NAC) and vitamin E (Trolox). The cells were incubated with the investigated compounds for 24 h. Results are expressed as the mean ± SEM of three repeated experiments. IC50 values (µM) indicate the concentration of a tested compound required to reduce the fraction of surviving cells to 50% compared to the control probe (untreated cell). Results represent the mean ± SEM of the data from three individual experiments, * <span class="html-italic">p</span> &lt; 0.05 vs. tested derivative used without antioxidants. Designation of compounds on the chart: 1; 3; 5.</p>
Full article ">Figure 10
<p>Effects of tested compounds (<b>3</b>; <b>5</b>, <b>β-CD</b> and their combinations <b>β-CD + 3</b> and <b>β-CD + 5</b>) on the morphology of red blood cells in vitro after 24 h incubation at 37 °C. Representative phase-contrast images are shown (magnification = 400 times). Examples of echinocytes, stomatocytes and eryptotic erythrocytes are marked with arrows.</p>
Full article ">Figure 11
<p>The influence of the tested compounds (<b>3</b>; <b>5</b>; <b>β-CD</b> and the combinations <b>β-CD + 3</b> and <b>β-CD + 5</b>) in the concentration range of 1–100 µmol/L on the extrinsic coagulation pathway expressed as prothrombin time (PT). No statistically significant differences were noted compared to controls. Results are presented as mean values ± standard deviation (SD) from three independent experiments.</p>
Full article ">Figure 12
<p>The influence of the tested compounds (<b>3</b>; <b>5</b>; <b>β-CD</b> and their combinations <b>β-CD + 3</b> and <b>β-CD + 5</b>) in the concentration range of 1–100 µmol/L on the intrinsic coagulation pathway expressed as activated partial thromboplastin time (APTT). A statistically significant difference was noted compared to control values *—<span class="html-italic">p</span> = 0.05; **—<span class="html-italic">p</span> = 0.01–0.001; ***—<span class="html-italic">p</span> &lt; 0.001. Results are presented as mean values ± standard deviation (SD) from three independent experiments.</p>
Full article ">Figure 13
<p>The influence of the tested compounds (<b>3</b>; <b>5</b>; <b>β-CD</b> and their combinations <b>β-CD + 3</b> and <b>β-CD + 5</b>) in the concentration range of 1–100 µmol/L on the common pathway, expressed as thrombin time (TT). The results significantly differed with controls *—<span class="html-italic">p</span> = 0.05; **—<span class="html-italic">p</span> = 0.01–0.001; ***—<span class="html-italic">p</span> &lt; 0.001. Results are presented as mean values ± standard deviation (SD) from three independent experiments.</p>
Full article ">Figure 14
<p>Direct effects of interaction between 0.2 mM <b>3</b> and 5 mM cyclodextrin (<span style="color:#388600">■</span>—α-CD, <span style="color:#004F88">■</span>—β-CD, <span style="color:#ED0000">■</span>—HP-β-CD). Solid lines were calculated assuming one type of active site.</p>
Full article ">Figure 15
<p>Direct effects of interaction between 0.2 mM <b>3a</b> and 5 mM cyclodextrin (<span style="color:#388600">■</span>—α-CD, <span style="color:#004F88">■</span>—β-CD, <span style="color:#ED0000">■</span>—HP-β-CD). Solid lines were calculated assuming one type of active site.</p>
Full article ">Figure 16
<p>Direct effects of interaction between 0.2 mM <b>5</b> and 5 mM cyclodextrin (<span style="color:#388600">■</span>—α-CD, <span style="color:#004F88">■</span>—β-CD, <span style="color:#ED0000">■</span>—HP-β-CD). Solid lines were calculated assuming one type of active site.</p>
Full article ">Figure 17
<p>Dependence of increasing the solubility of <b>3</b> in water on the increasing concentration of α-CD (<span style="color:#00B050">•</span>), β-CD (<span style="color:#4472C4">♦</span>) and HP-β-CD (<span style="color:red">■</span>).</p>
Full article ">Figure 18
<p>Dependence of increasing the solubility of <b>3</b> in water on the increasing concentration of α-CD (<span style="color:#00B050">•</span>), β-CD (<span style="color:#4472C4">♦</span>) and HP-β-CD (<span style="color:red">■</span>).</p>
Full article ">Figure 19
<p>Dependence of increasing the solubility of <b>5</b> in water on the increasing concentration of α-CD (<span style="color:#538135">•</span>), β-CD (<span style="color:#4472C4">♦</span>) and HP-β-CD (<span style="color:red">■</span>).</p>
Full article ">Figure 20
<p>Illustration of the docking results based on the example of the most strongly interacting complex. (4a in Ledrob dextrin). The side view is given in the left panel and the top view in the right panel. Hydrogen atoms in the ligand have been omitted for clarity.</p>
Full article ">Figure 21
<p>Calibration curve of ligands.</p>
Full article ">Scheme 1
<p>The structures of tested compounds.</p>
Full article ">
22 pages, 2341 KiB  
Article
Anti-Platelet Activity of Sea Buckthorn Seeds and Its Relationship with Thermal Processing
by Natalia Sławińska, Jerzy Żuchowski, Anna Stochmal and Beata Olas
Foods 2024, 13(15), 2400; https://doi.org/10.3390/foods13152400 - 29 Jul 2024
Viewed by 507
Abstract
Sea buckthorn (Hippophae rhamnoides L.) is a tree or shrub with small, orange berries. Sea buckthorn seeds have shown many properties beneficial to human health, including antioxidant, anti-hypertensive, anti-hyperlipidemic, and retinoprotective activities. Seeds, as a component of food, are often exposed to [...] Read more.
Sea buckthorn (Hippophae rhamnoides L.) is a tree or shrub with small, orange berries. Sea buckthorn seeds have shown many properties beneficial to human health, including antioxidant, anti-hypertensive, anti-hyperlipidemic, and retinoprotective activities. Seeds, as a component of food, are often exposed to high temperatures, which can increase or decrease their biological activity. In our previous study, we showed that both raw and roasted sea buckthorn seeds had significant antioxidant activity, which was measured in human plasma in vitro. In this paper, we evaluated the effect of extracts from raw and roasted sea buckthorn seeds on several parameters of hemostasis in vitro, including thrombus formation in full blood (measured by the Total Thrombus formation Analysis System—T-TAS), blood platelet activation (based on the exposition of P-selectin, the active form of GPIIb/IIIa on their surface and platelet-derived microparticles formation), aggregation (measured with impedance aggregometry), adhesion to fibrinogen and collagen, arachidonic acid metabolism in washed platelets stimulated by thrombin, and COX-1 activity. We also measured the levels of free 8-isoprostane in plasma and the total non-enzymatic antioxidant status of plasma. The extract from roasted seeds (50 µg/mL) significantly prolonged the time of occlusion measured by T-TAS—the AUC10 (area under the curve) value was decreased by approximately 18%. Both extracts decreased the exposition of the active form of GPIIb/IIIa on the surface of platelets activated with 10 μM ADP (by 38.4–62.2%) and 20 μM ADP (by 39.7–51.3%). Moreover, the extract from raw seeds decreased the exposition of P-selectin on the surface of platelets stimulated with 20 μM ADP (by 31.2–34.9%). The adhesion of thrombin-stimulated platelets to fibrinogen and collagen was inhibited only by the extract from roasted sea buckthorn seeds (by 20–30%). Moreover, the extract from raw seeds inhibited the level of TBARS (thiobarbituric acid-reactive substances, an indicator of enzymatic peroxidation of arachidonic acid) in washed platelets stimulated with thrombin; the activity of COX-1 was inhibited by both extracts, although the effect of the extract from raw seeds was stronger. These results indicate that sea buckthorn seeds have anti-platelet activity that is not decreased by thermal processing, but more research is needed to determine which exact chemical compounds and mechanisms are responsible for this phenomenon. Full article
(This article belongs to the Section Food Engineering and Technology)
Show Figures

Figure 1

Figure 1
<p>Effect of the extracts from raw and roasted sea buckthorn seeds (at concentrations of 0.5–50 μg/mL) on thrombus formation in whole blood (<b>A</b>,<b>B</b>) (<span class="html-italic">n</span> = 6) and blood platelet aggregation in whole blood (<b>C</b>) (<span class="html-italic">n</span> = 6). The samples (<b>A</b>,<b>B</b>) were analyzed with the T-TAS PL chip at the shear stress rates of 1500/s. The results (<b>A</b>,<b>B</b>) are calculated as AUC<sub>10</sub> (area under the curve). In the graph (<b>A</b>), AUC<sub>10</sub> is expressed as a percentage of the control sample (blood without the tested extract). The data are expressed as means ± SD (<b>A</b>) or median and interquartile range (<b>C</b>). The results were considered significant at <span class="html-italic">p</span> &lt; 0.05 (* <span class="html-italic">p</span> &lt; 0.05). The number above the significant result is the % of inhibition of thrombus formation (<b>A</b>). (<b>B</b>) demonstrates a selected diagram of the pressure recorded inside the PL chip for 10 min (OT—occlusion time) (for the extract from roasted seeds).</p>
Full article ">Figure 2
<p>Effect of the extracts from raw and roasted sea buckthorn seeds (at concentrations of 0.5–50 μg/mL) on the exposition of the active form of GPIIb/IIIa on 10 µM ADP-stimulated blood platelets (<b>A</b>,<b>B</b>) and the exposition of P-selectin on 10 µM ADP-stimulated blood platelets (<b>C</b>,<b>D</b>) in whole blood. Blood platelets were gated based on their size and the exposition of CD61. In each sample, 5000 CD61-positive objects were acquired. To assess the exposition of GPIIb/IIIa, fluorescently conjugated monoclonal antibody PAC-1/FITC was used. Results are expressed as the percentage of platelets binding PAC-1/FITC. To assess the exposition of P-selectin, a fluorescently conjugated monoclonal antibody CD62P/PE was used. Results are expressed as the percentage of platelets binding CD62P/PE. Data represent the means ± SD. The blood samples were drawn from 5–6 healthy volunteers. The activity of the tested extract was compared to the control sample. The results were considered significant at <span class="html-italic">p</span> &lt; 0.05 (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001); ns—not significant.</p>
Full article ">Figure 3
<p>Effect of the extracts from raw and roasted sea buckthorn seeds (at concentrations of 0.5–50 μg/mL) on the exposition of the active form of GPIIb/IIIa on 20 µM ADP-stimulated blood platelets (<b>A</b>,<b>B</b>), and the exposition of P-selectin on 20 µM ADP-stimulated blood platelets (<b>C</b>,<b>D</b>) in whole blood. Blood platelets were gated based on their size and the exposition of CD61. For each sample, 5000 CD61-positive objects were acquired. To assess the exposition of GPIIb/IIIa, a fluorescently conjugated monoclonal antibody PAC-1/FITC was used. Results are expressed as the percentage of platelets binding PAC-1/FITC. To assess the exposition of P-selectin, fluorescently conjugated monoclonal antibody CD62P/PE was used. Results are expressed as the percentage of platelets binding CD62P/PE. Data represent the means ± SD. The blood samples were drawn from 5–6 healthy volunteers. The activity of the tested extract was compared to the control samples. The results were considered significant at <span class="html-italic">p</span> &lt; 0.05 (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01); ns—not significant.</p>
Full article ">Figure 4
<p>Effect of the extracts from raw and roasted sea buckthorn seeds (at concentrations of 0.5–50 μg/mL) on the adhesion of thrombin-activated platelets to fibrinogen (<b>A</b>), ADP-activated platelets to fibrinogen (<b>B</b>), thrombin-activated platelets to collagen (<b>C</b>), and the adhesion of unstimulated platelets to collagen (<b>D</b>) (<span class="html-italic">n</span> = 7). In the graphs, platelet adhesion is expressed as a percentage of the control sample (blood platelets without the tested extract). The data are expressed as medians and interquartile ranges. The results were considered significant at <span class="html-italic">p</span> &lt; 0.05 (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001); ns—not significant.</p>
Full article ">Figure 5
<p>Effect of the extracts from raw and roasted sea buckthorn seeds (at concentrations of 0.5–50 μg/mL) on arachidonic acid metabolism in blood platelets stimulated by thrombin (5 U/mL) (<span class="html-italic">n</span> = 8) (<b>A</b>), and the activity of COX-1 (<b>B</b>). In (<b>A</b>), the results are presented as TBARS/2 × 10<sup>8</sup> blood platelets (percent of the control sample). The data are expressed as medians and interquartile ranges. The results were considered significant at <span class="html-italic">p</span> &lt; 0.05 (*** <span class="html-italic">p</span> &lt; 0.001). The differences between negative and positive control were statistically significant (<span class="html-italic">p</span> &lt; 0.001). In (<b>B</b>), the results are expressed as % of COX-1 inhibition (in comparison to 100% initial activity sample—sample where inhibitor vehicle (50% DMSO) was added instead of the extracts). The extracts were incubated with COX-1 for 10 or 30 min at the concentrations of 1, 10, 50, and 100 μg/mL. The data are shown as the means and SD of two dilutions of the sample (each measured in duplicate).</p>
Full article ">
Back to TopTop