Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Pharmacol Rep ; 76(2): 328-337, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38438661

ABSTRACT

Optical aggregometry by 96-well plate assay, the microplate method, is a fast, efficient, and readily available method for measuring the pharmacological effects of antiplatelet drugs. Even though recent years have witnessed growing interest in adopting the microplate method for widespread use, it remains in the shadow of the standard light transmission aggregometry (LTA). Regardless of the method used, the results of platelet aggregation depend on a variety of factors and often vary among laboratories worldwide. While several methodological papers have examined the microplate method, no standards have been established, most likely because the approach is not used as a diagnostic tool. Currently, the microplate method is recommended by researchers to be used in conjunction with LTA or as an adjunct to LTA. This raises the question of whether an optimal protocol exists for microplate aggregometry, and what are the key considerations in a good experimental protocol for obtaining reliable results? This article attempts to address these questions by summarizing the knowledge accumulated in this field over the last three decades.


Subject(s)
Platelet Aggregation , Platelet Function Tests , Platelet Function Tests/methods , Platelet Aggregation Inhibitors/pharmacology , Reference Standards , Blood Platelets
2.
Br J Pharmacol ; 181(4): 547-563, 2024 02.
Article in English | MEDLINE | ID: mdl-37218380

ABSTRACT

Traditionally, platelets are known to play an important role in haemostasis and thrombosis; however, they serve also as important modulators of inflammation and immunity. Platelets secrete adhesion molecules and cytokines, interact with leukocytes and endothelium, and express toll-like receptors involved in a direct interaction with pathogens. Platelets express A2A and A2B subtypes of receptors for adenosine. The activation of these receptors leads to an increase in cAMP concentration in the cytoplasm, thereby resulting in inhibited secretion of pro-inflammatory mediators and reduced cell activation. Therefore, platelet adenosine receptors could be a potential target for inhibiting platelet activation and thus down-regulating inflammation or immunity. The biological effects of adenosine are short-lasting, because the compound is rapidly metabolized; hence, its lability has triggered efforts to synthesize new, longer-lasting adenosine analogues. In this article, we have reviewed the literature regarding the pharmacological potential of adenosine and other agonists of A2A and A2B receptors to affect platelet function during inflammation. LINKED ARTICLES: This article is part of a themed issue on Platelet purinergic receptor and non-thrombotic disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.4/issuetoc.


Subject(s)
Blood Platelets , Thrombosis , Humans , Adenosine/pharmacology , Adenosine/metabolism , Receptors, Purinergic P1/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Platelet Activation , Thrombosis/metabolism
3.
Macromol Biosci ; 23(7): e2300052, 2023 07.
Article in English | MEDLINE | ID: mdl-37084188

ABSTRACT

A commercial nonbinding surface effectively prevents protein adsorption; however, the platelet phenotype on this surface has yet to be defined. This study evaluates platelet adhesion and adsorption of several plasma/extracellular matrix (ECM) proteins to the nonbinding surface compared to other commonly used nontreated and high-binding surfaces. Platelet adhesion to uncoated microplates and those coated with fibrinogen or collagen is quantified by colorimetric assay. The binding capacity of the examined surfaces for plasma/ECM proteins is evaluated by measuring the relative and absolute protein adsorption. Compared to other surfaces, the nonbinding surface effectively prevents platelet adsorption, i.e. by 61-93% (Enzyme-Linked Immunosorbent Assay, ELISA), and reduces platelet adhesion, i.e. by 92%, when not coated with any protein. The nonbinding surface also decreases platelet deposition on collagen (up to 31%), but not fibrinogen. The nonbinding surface seems to be more of a low-fouling than nonfouling material, as it is able to reduce fibrinogen adsorption but not prevent platelet adhesion to fibrinogen. This feature should be considered when using the nonbinding surface for in vitro platelet testing.


Subject(s)
Fibrinogen , Hemostatics , Fibrinogen/metabolism , Adsorption , Platelet Adhesiveness , Blood Platelets/metabolism , Hemostatics/pharmacology , Surface Properties
4.
Int J Mol Sci ; 24(6)2023 Mar 10.
Article in English | MEDLINE | ID: mdl-36982387

ABSTRACT

Several studies report elevated blood platelet activation and altered platelet count in COVID-19 patients, but the role of the SARS-CoV-2 spike protein in this process remains intriguing. Additionally, there is no data that anti-SARS-CoV-2 neutralizing antibodies (nAb) may attenuate spike protein activity toward blood platelets. Our results indicate that under in vitro conditions, the spike protein increased the collagen-stimulated aggregation of isolated platelets and induced the binding of vWF to platelets in ristocetin-treated blood. The spike protein also significantly reduced collagen- or ADP-induced aggregation or decreased GPIIbIIIa (fibrinogen receptor) activation in whole blood, depending on the presence of the anti-spike protein nAb. Our findings suggest that studies on platelet activation/reactivity in COVID-19 patients or in donors vaccinated with anti-SARS-CoV-2 and/or previously-infected COVID-19 should be supported by measurements of spike protein and IgG anti-spike protein antibody concentrations in blood.


Subject(s)
COVID-19 , Humans , COVID-19/metabolism , Spike Glycoprotein, Coronavirus/metabolism , SARS-CoV-2/metabolism , Blood Platelets/metabolism , Antibodies, Viral , Antibodies, Neutralizing
5.
Pharmacol Rep ; 75(2): 423-441, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36646965

ABSTRACT

BACKGROUND: Clinical trials indicate that fentanyl, like morphine, may impair intestinal absorption and thus decrease the efficacy of oral P2Y12 inhibitors, such as clopidogrel, ticagrelor, and prasugrel. However, the ability of fentanyl to directly negate or reduce the inhibitory effect of P2Y12 receptor antagonists on platelet function has not been established. A series of in vitro experiments was performed to investigate the ability of fentanyl to activate platelets, potentiate platelet response to ADP, and/or diminish platelet sensitivity to prasugrel metabolite (R-138727) in agonist-stimulated platelets. The selectivity and specificity of fentanyl toward major carrier proteins has been also studied. METHODS: Blood was obtained from healthy volunteers (19 women and 12 men; mean age 40 ± 13 years). Platelet function was measured in whole blood, platelet-rich plasma and in suspensions of isolated platelets by flow cytometry, impedance and optical aggregometry. Surface plasmon resonance and molecular docking were employed to determine the binding kinetics of fentanyl to human albumin, α1-acid glycoprotein, apolipoprotein A-1 and apolipoprotein B-100. RESULTS: When applied at therapeutic and supratherapeutic concentrations under various experimental conditions, fentanyl had no potential to stimulate platelet activation and aggregation, or potentiate platelet response to ADP, nor did it affect platelet susceptibility to prasugrel metabolite in ADP-stimulated platelets. In addition, fentanyl was found to interact with all the examined carrier proteins with dissociation constants in the order of 10-4 to 10-9 M. CONCLUSIONS: It does not seem that the delayed platelet responsiveness to oral P2Y12 inhibitors, such as prasugrel, in patients undergoing percutaneous coronary intervention, results from direct interactions between fentanyl and blood platelets. Apolipoproteins, similarly to albumin and α1-acid glycoprotein, appear to be important carriers of fentanyl in blood.


Subject(s)
Blood Platelets , Platelet Aggregation Inhibitors , Male , Humans , Female , Adult , Middle Aged , Prasugrel Hydrochloride/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Orosomucoid/metabolism , Orosomucoid/pharmacology , Fentanyl/pharmacology , Molecular Docking Simulation , Platelet Aggregation , Purinergic P2Y Receptor Antagonists/pharmacology
6.
Toxicol In Vitro ; 83: 105407, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35659575

ABSTRACT

INTRODUCTION: Commercially-available resazurin-based reagents used for cell viability assessment contain varying amounts of resorufin; these may contribute to differences in autofluorescence, signal-to-background (S/B) ratio and the dynamic range of the assay. OBJECTIVES: This in vitro study compares the sensitivity of a new, high-sensitivity PrestoBlue (hs-PB) assay with standard PrestoBlue (PB) in assessing the efficacy of valinomycin and antimycin A in human vascular endothelial EA.hy926 cells, as well as cell viability. METHODS: The metabolic activity of EA.hy926 was evaluated based on resorufin fluorescence (PB assays) or formazan absorbance (MTT assay). RESULTS: The hs-PB assay demonstrated lower resorufin autofluorescence than the PB, resulting in a ≥ 1.4-fold increase in S/B ratio in hs-PB compared to PB. Valinomycin was more potent cytotoxic agent than antimycin A. The hs-PB, PB and MTT produced similar IC50 values for valinomycin. Antimycin A showed significantly higher potency in the MTT than in the resazurin-based assays. The EA.hy926 cells demonstrated higher metabolic activity in the presence of the antimycin A solvent - DMSO. CONCLUSION: All the examined methods may be used interchangeably to analyze drug cytotoxicity. Any differences in drug cytotoxicity observed between the assays may be due to relatively low drug potency and/or the influence of solvent on metabolism of assay reagent. The hs-PB assay appears to more effectively detect cell viability and produce a stronger signal than its conventional counterpart.


Subject(s)
Endothelial Cells , Antimycin A/metabolism , Antimycin A/toxicity , Cell Survival , Humans , Indicators and Reagents/pharmacology , Solvents/pharmacology , Valinomycin/metabolism , Valinomycin/pharmacology
7.
Thromb Res ; 211: 88-97, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35134667

ABSTRACT

INTRODUCTION: The combination index (CI), a common quantitative indicator of the degree of synergy/antagonism, may be determined using different regression methods. However, any analysis with constraints has the potential for underestimating the combined effect of multiple drugs. OBJECTIVES: This in vitro study describes the combined effects of selected platelet antagonists on ADP-induced platelet activation in different regression models. METHODS: The inhibitory effects of P2Y12 receptor antagonists in combination with P2Y1 receptor antagonists (i.e. cangrelor with MRS 2279, prasugrel metabolite with MRS 2179 and PSB 0739 with MRS 2179) were characterized with the aid of three software packages: CompuSyn (for linear regression with constraints), CISNE (for non-linear regression with constraints) and GraphPad Prism (for non-linear regression without constraints). The synergism between P2Y12 and P2Y1 inhibitors was quantified by CI and synergy area. RESULTS: MRS 2279 and MRS 2179 were found to act synergistically with selected P2Y12 receptor antagonists to potentiate their antiplatelet effect. The models of regression with constraints, linear regression in particular, demonstrated a worse fit for experimental data than non-linear regression without constraints; this resulted in an incorrect estimation of the combined effects of two antiplatelet drugs, i.e., underestimating the CI and overestimating the synergy area. Also, the synergy area was found to better reflect the differences among models than the CI. CONCLUSIONS: These findings suggest that non-linear regression without constraints offers more precise quantitative determination of combined effects between two drugs compared to the regression models with constraints.


Subject(s)
Platelet Activation , Platelet Aggregation , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Blood Platelets/metabolism , Humans , Platelet Aggregation Inhibitors/pharmacology , Prasugrel Hydrochloride/pharmacology , Prasugrel Hydrochloride/therapeutic use , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y1/metabolism , Receptors, Purinergic P2Y12/metabolism
9.
Front Pharmacol ; 12: 638257, 2021.
Article in English | MEDLINE | ID: mdl-33776774

ABSTRACT

Concurrent administration of two drugs may complicate the management of acute coronary syndromes: competitive drug displacement diminishes drug binding and alters drug pharmacodynamics. We investigated the interaction of two antiplatelet compounds (PSB 0777 and cangrelor) with human serum albumin (HSA) to determine whether they compete with one another for the binding to albumin. Both examined compounds have been earlier claimed to bind to HSA (PSB 0777) or plasma proteins (cangrelor). Fluorescence spectroscopy, surface plasmon resonance spectroscopy and molecular modeling indicated that PSB 0777 and cangrelor interacted with HSA with moderate affinity (KD∼10-5 M). The binding of cangrelor to HSA involved primarily hydrophobic interactions, while the interaction of PSB 0777 with HSA was driven by hydrophobic and electrostatic forces. It was found that PSB 0777 and cangrelor do not share the same binding site on the protein. Our findings highlight the importance of albumin in the transport of PSB 0777 and cangrelor and suggest that the antiplatelet activity of the examined compounds used in combination is not affected by competition-induced changes in drug binding to HSA.

10.
J Cardiovasc Pharmacol ; 76(3): 267-275, 2020 09.
Article in English | MEDLINE | ID: mdl-32649576

ABSTRACT

Fentanyl is a potent synthetic opioid used to alleviate severe and chronic pain, as well as an adjunct to general or local anesthesia. Although fentanyl has been used for decades, its full effects are still unknown. Its analgesic and anesthetic activity arises from the stimulation of µ-opioid receptors, resulting in the inhibition of adenyl cyclase and downregulation of cyclic adenosine 3',5'-monophosphate (cAMP), as well as decreased calcium channel activity and increased potassium channel activity. The µ-opioid receptors are abundantly distributed within the central nervous system, where they mediate analgesia, and in the nerve cells of the intestines, where they regulate gastrointestinal tract motility in the secretion or transport of fluids and electrolytes. They are also expressed in blood cells, blood vessel cells, and skin. Given the widespread distribution of µ-opioid receptors, it is likely that fentanyl may also regulate the activity of many other cells, including platelets. Recent findings indicate that it may impair the action of ticagrelor: an oral P2Y12 receptor inhibitor acting as an antiplatelet drug. It could pose a risk of insufficient platelet inhibition and result in thrombotic complications in patients with coronary artery disease. This article tackles the issue of fentanyl interactions with antiplatelet drugs. The mechanism of this phenomenon is not fully understood. Similarly, the biological effects exerted by fentanyl on platelets and the presence of opioid receptors on the platelet surface remain an open question.


Subject(s)
Analgesics, Opioid/therapeutic use , Blood Platelets/drug effects , Fentanyl/therapeutic use , Platelet Aggregation Inhibitors/therapeutic use , Platelet Aggregation/drug effects , Purinergic P2Y Receptor Antagonists/therapeutic use , Receptors, Opioid, mu/agonists , Analgesics, Opioid/adverse effects , Analgesics, Opioid/pharmacokinetics , Animals , Blood Platelets/metabolism , Drug Interactions , Fentanyl/adverse effects , Fentanyl/pharmacokinetics , Humans , Patient Safety , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/pharmacokinetics , Purinergic P2Y Receptor Antagonists/adverse effects , Purinergic P2Y Receptor Antagonists/pharmacokinetics , Receptors, Opioid, mu/metabolism , Risk Assessment , Risk Factors , Signal Transduction
11.
Biomolecules ; 10(6)2020 06 07.
Article in English | MEDLINE | ID: mdl-32517350

ABSTRACT

BACKGROUND: Fibrin formation and structure may be affected by a plethora of factors, including both genetic and posttranslational modifications, such as glycation, nitration or acetylation. METHODS: The present study examines the effect of fibrinogen glycation on fibrin polymerization, measured in fibrinogen concentration-standardized plasma of subjects with type 2 diabetes mellitus (T2DM) and in a solution of human fibrinogen exposed to 30 mM glucose for four days. RESULTS: The fibrin polymerization velocity (Vmax) observed in the T2DM plasma (median 0.0056; IQR 0.0049‒0.0061 AU/s) was significantly lower than in non-diabetic plasma (median 0.0063; IQR 0.0058‒0.0071 AU/s) (p < 0.05). Furthermore, significantly lower Vmax was observed for glucose-treated fibrinogen (Vmax 0.046; IQR 0.022‒0.085 AU/s) compared to control protein incubated with a pure vehicle (Vmax 0.053; IQR 0.034‒0.097 AU/s) (p < 0.05). The same tendency was observed in the fibrinogen samples supplemented with 6 mM glucose just before measurements. It is assumed that glucose may affect the ability of fibrinogen to form a stable clot in T2DM subjects, and that this impairment is likely to influences the outcomes of some diagnostic assays. As the example, the impaired clotting ability of glycated fibrinogen may considerably influence the results of the standard Clauss method, routinely used to determine fibrinogen concentration in plasma. The stoichiometric analysis demonstrated that spontaneous glycation at both the sites with high and low glycation potential clearly dominated in T2DM individuals in all fibrinogen chains.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Fibrin/metabolism , Fibrinogen/metabolism , Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Female , Fibrin/chemistry , Fibrinogen/analysis , Glucose/chemistry , Humans , Male , Middle Aged , Polymerization
12.
Int J Mol Sci ; 21(9)2020 May 02.
Article in English | MEDLINE | ID: mdl-32370146

ABSTRACT

Blood platelets play a crucial role in the early stages of atherosclerosis development. The process is believed to require firm adhesion of platelets to atherosclerosis-prone sites of the artery. However, little evidence exists regarding whether the blood platelets of individuals with pathological conditions associated with atherosclerosis have higher potential for adhesion. This process is to a large extent dependent on receptors present on the platelet membrane. Therefore, the aim of the presented study was to determine whether blood platelets from diabetic patients have higher capacity of adhesion under flow conditions and how diabetes affects one of the crucial platelet receptors involved in the process of adhesion-GPIIIa. The study compares the ability of platelets from non-diabetic and diabetic humans to interact with fibrinogen and von Willebrand factor, two proteins found in abundance on an inflamed endothelium, under flow conditions. The activation and reactivity of the blood platelets were also characterized by flow cytometry. Platelets from diabetic patients did not demonstrate enhanced adhesion to either studied protein, although they presented increased basal activation and responsiveness towards low concentrations of agonists. Platelets from diabetic patients were characterized by lower expression of GPIIIa, most likely due to an enhanced formation of platelet-derived microparticles PMPs, as supported by the observation of elevated concentration of this integrin and of GPIIIa-positive PMPs in plasma. We conclude that altered functionality of blood platelets in diabetes does not increase their adhesive potential. Increased glycation and decrease in the amount of GPIIIa on platelets may be partially responsible for this effect. Therefore, higher frequency of interactions of platelets with the endothelium, which is observed in animal models of diabetes, is caused by other factors. A primary cause may be a dysfunctional vascular wall.


Subject(s)
Blood Platelets/metabolism , Diabetes Mellitus, Type 2/blood , Hyperglycemia/blood , Integrin beta3/biosynthesis , Platelet Adhesiveness , Adult , Aged , Cell-Derived Microparticles/metabolism , Female , Flow Cytometry/methods , Humans , Male , Middle Aged , Platelet Activation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism
13.
Biochem Pharmacol ; 174: 113827, 2020 04.
Article in English | MEDLINE | ID: mdl-31987853

ABSTRACT

Adenosine analogues have high affinity and selectivity for adenosine receptors (AR), and exhibit anti-platelet activity. Plasma proteins play an important role in the regulation of platelet function and may influence the action of anti-platelet compounds. Little is known about the interactions of AR agonists with plasma proteins. This study investigates the interplay between AR agonists and plasma proteins and the consequences of those interactions. Surface plasmon resonance was employed together with molecular docking study to determine the binding kinetics of four selected ARagonists (PSB0777, Cl-Ado, MRE0094, UK432097) to several carrier proteins and to clarify the nature of these interactions. The influence of a whole plasma and of some plasma components on the effectiveness of ARagonists in the inhibition of platelet function was assessed by flow cytometry (platelet activation) and ELISA (platelet adhesion). Plasma proteins remarkably diminished the effectiveness of ARagonists in inhibiting platelet activation and adhesion in vitro. ARagonists were found to strongly bind to human serum albumin (HSA) and the protein components of lipoproteins - apolipoproteins; HSA was essential for the binding of water-soluble PSB0777, whereas apolipoproteins were needed for interactions with poorly-water soluble compounds such as UK432097 and MRE0094. In addition, HSA was shown to significantly reduce the effectiveness of PSB0777 in inhibiting ADP-induced platelet activation. In conclusion, HSA and lipoproteins are important carriers for ARagonists, which can affect pharmacodynamics of ARagonists used as platelet inhibitors.


Subject(s)
Adenosine A2 Receptor Agonists/pharmacology , Adenosine/pharmacology , Blood Platelets/drug effects , Blood Platelets/metabolism , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Adenosine/analogs & derivatives , Adenosine/chemistry , Adenosine A2 Receptor Agonists/chemistry , Adult , Female , Furans/chemistry , Furans/pharmacology , Humans , Male , Molecular Docking Simulation/methods , Platelet Activation/physiology , Platelet Aggregation/drug effects , Platelet Aggregation/physiology , Protein Binding/drug effects , Protein Binding/physiology , Receptors, Purinergic P1/metabolism , Young Adult
14.
Molecules ; 24(11)2019 May 30.
Article in English | MEDLINE | ID: mdl-31151201

ABSTRACT

C-reactive protein (CRP) is an intriguing protein which plays a variety of roles in either physiological or pathophysiological states. For years it has been regarded merely as a useful biomarker of infection, tissue injury and inflammation, and it was only in the early 80s that the modified isoforms (mCRP) of native CRP (nCRP) appeared. It soon became clear that the roles of native CRP should be clearly discriminated from those of the modified form and so the impacts of both isoforms were divided to a certain degree between physiological and pathophysiological states. For decades, CRP has been regarded only as a hallmark of inflammation; however, it has since been recognised as a significant predictor of future episodes of cardiovascular disease, independent of other risk factors. The existence of modified CRP isoforms and their possible relevance to various pathophysiological conditions, suggested over thirty years ago, has prompted the search for structural and functional dissimilarities between the pentameric nCRP and monomeric mCRP isoforms. New attempts to identify the possible relevance between the diversity of structures and their opposing functions have initiated a new era of research on C-reactive protein. This review discusses the biochemical aspects of CRP physiology, emphasizing the supposed relevance between the structural biology of CRP isoforms and their differentiated physiological and pathophysiological roles.


Subject(s)
C-Reactive Protein/chemistry , C-Reactive Protein/metabolism , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Disease Susceptibility , Animals , Biomarkers , Glycosylation , Humans , Protein Binding , Protein Conformation , Protein Multimerization , Structure-Activity Relationship
15.
Molecules ; 25(1)2019 Dec 28.
Article in English | MEDLINE | ID: mdl-31905703

ABSTRACT

Large inter-individual variation in platelet response to endogenous agonists and pharmacological agents, including resistance to antiplatelet therapy, prompts a search for novel platelet inhibitors and development new antithrombotic strategies. The present in vitro study evaluates the beneficial effects of three adenosine receptor (AR) agonists (regadenoson, LUF 5835 and NECA), different in terms of their selectivity for platelet adenosine receptors, when used alone and in combination with P2Y12 inhibitors, such as cangrelor or prasugrel metabolite. The anti-platelet effects of AR agonists were evaluated in healthy subjects (in the whole group and after stratification of individuals into high- and low-responders to P2Y12 inhibitors), using whole blood techniques, under flow (thrombus formation) and static conditions (study of platelet activation and aggregation). Compared to P2Y12 antagonists, AR agonists were much less or not effective under static conditions, but demonstrated similar antiplatelet activity in flow. In most cases, AR agonists significantly enhanced the anti-platelet effect of P2Y12 antagonists, despite possessing different selectivity profiles and antiplatelet activities. Importantly, their inhibitory effects in combination with P2Y12 antagonists were similar in high- and low-responders to P2Y12 inhibitors. In conclusion, a combination of anti-platelet agents acting via the P1 and P2 purinergic receptors represents a promising alternative to existing antithrombotic therapy.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Drug Resistance/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Prasugrel Hydrochloride/pharmacology , Purinergic P1 Receptor Agonists/pharmacology , Receptors, Purinergic P2Y12/metabolism , Adenosine Monophosphate/pharmacology , Female , Humans , Male , Thrombosis/drug therapy , Thrombosis/metabolism , Thrombosis/pathology
16.
Basic Clin Pharmacol Toxicol ; 124(2): 199-210, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30137675

ABSTRACT

Mouse 3T3 fibroblasts are commonly used for in vitro toxicity testing; however, their sensitivity to stimuli is not well defined. To assess the sensitivity of the 3T3 cell line, the study compared the changes in mitochondrial membrane potential (MMP) occurring after exposure to eight chemicals known to demonstrate pro-apoptotic activity (glycerol, isopropanol, ethanol, paracetamol, propranolol, cobalt chloride, formaldehyde and atropine). Five cell lines were used as follows: mouse 3T3 fibroblasts, human epithelial cells (A549, Caco-2 and HepG2) and human endothelial cells (HMEC-1). Cell sensitivity was assessed based on the total area under and over the dose-response curves (AUOC) in relation to baselines. The 3T3 fibroblasts had the highest AUOC values and were the most sensitive to the action of all the examined chemicals, with the exception of formaldehyde. Significant changes in MMP between the 3T3 cell line and other cells were observed after cell treatment with atropine (A549, Caco-2 or HMEC-1 cells vs 3T3 cells, P < 0.05), propranolol (A549 vs 3T3 cells, P < 0.01; HepG2 vs 3T3 cells, P < 0.05), cobalt chloride (A549 cells vs 3T3 cells, P < 0.01) or ethanol (HMEC-1 vs 3T3, P < 0.05). Formaldehyde appeared the most toxic compound for Caco-2 cells (Caco-2 vs 3T3 cells, P < 0.05). The surface areas (AUOC) calculated for each other chemical and obtained for HepG2, Caco-2, A549 and HMEC-1 did not differ significantly between cell lines. We postulate that mouse 3T3 fibroblasts demonstrate significantly higher relative sensitivity to many agents with toxic potential.


Subject(s)
Acetaminophen/pharmacology , Atropine/pharmacology , Cobalt/pharmacology , Ethanol/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Propranolol/pharmacology , 3T3-L1 Cells , A549 Cells , Animals , Caco-2 Cells , Cell Count , Cell Survival/drug effects , Endothelial Cells/drug effects , Hep G2 Cells , Humans , Mice , Mitochondria/metabolism
17.
Vascul Pharmacol ; 113: 47-56, 2019 02.
Article in English | MEDLINE | ID: mdl-30471364

ABSTRACT

Several adenosine receptor (AR) agonists have been shown in the past to possess anti-platelet potential; however, the adjunctive role of AR agonists in anti-platelet therapy with the use of P2Y12 receptor inhibitors has not been elucidated so far. This in vitro aggregation-based study investigates whether the inhibition of platelet function mediated by cangrelor or prasugrel metabolite can be potentiated by AR agonists. It evaluates the effect of non-selective (2-chloroadenosine), A2A-selective (UK 432097, MRE 0094, PSB 0777) and A2B-selective AR agonists (BAY 60-6583) on platelet function in relation to their toxicity, specificity towards adenosine receptor subtypes, structure and solubility. UK 432097, 2-chloroadenosine, MRE 0094 and PSB 0777 were found to be more or less potent inhibitors of ADP-induced platelet aggregation when acting alone, and that they remained non-cytotoxic to the cells. These AR agonists were also effective in the potentiation of the effects exerted by P2Y12 antagonists. Considering the estimated IC50 value, UK 432097, showing a relatively high binding affinity to the A2A adenosine receptor, has been identified as the most potent anti-aggregatory agent. This compound diminished platelet aggregation at nanomolar concentrations and further augmented platelet inhibition by P2Y12 antagonists by approx. 60% (P < .01). Our results indicate the importance of adenosine receptors as therapeutic targets and point out challenges and potential benefits of therapeutic use of a combined therapy of P2Y12 antagonist and AR agonist in cardioprotection. Our comparative analysis of the effects of AR agonists on platelet response in plasma and whole blood may indirectly suggest that other blood morphology elements contribute little to the inhibition of platelet function by AR agonists.


Subject(s)
Adenosine A2 Receptor Agonists/pharmacology , Blood Platelets/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y12/drug effects , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Adult , Blood Platelets/metabolism , Dose-Response Relationship, Drug , Female , Furans/pharmacology , Humans , Male , Prasugrel Hydrochloride/pharmacology , Receptors, Purinergic P2Y12/blood , Signal Transduction/drug effects , Young Adult
18.
Int J Biol Macromol ; 107(Pt B): 2701-2714, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29111269

ABSTRACT

We examined the structural and functional consequences of oxidative modification of C-reactive protein (CRP) by hypochlorous acid (HOCl), which can be generated in vivo via the myeloperoxidase/H2O2/Cl- system. HOCl exposure resulted in the oxidation and chlorination of CRP amino acid residues, leading to protein unfolding, greater surface hydrophobicity and the formation of aggregates. After treatment of isolated platelets with 50µg/ml HOCl-CRP, the modified CRP significantly stimulated platelet activation (over 10-fold increase in the fraction of CD62-positive platelets compared to controls, P<0.008), enhanced deposition of platelets onto immobilized fibrinogen (two-fold rise in platelet adhesion compared to controls, P<0.0001), and induced platelet aggregation by up to 79.5%. The ability of HOCl-CRP to interact with several platelet receptors (TLR-4, GPIIbIIIa) and plasma proteins (C1q, IgG) strongly indicates that HOCl-modification leads to structural changes of CRP resulting in the formation of new ligand binding sites, which is characteristic of the monomeric form of CRP exerting pro-inflammatory effects on a variety of cells. Overall, the oxidation of native CRP by HOCl seems to represent an alternative mechanism of CRP modification, by which CRP reveals its pro-inflammatory and pro-thrombotic properties, and as such, it might be of causal relevance in the pathogenesis of atherosclerosis.


Subject(s)
C-Reactive Protein/metabolism , Hypochlorous Acid/pharmacology , Platelet Activation/drug effects , Adenosine Diphosphate/pharmacology , Adsorption , Adult , Amino Acid Sequence , Benzothiazoles , C-Reactive Protein/chemistry , Collagen/pharmacology , Female , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Ligands , Male , Oxidation-Reduction , P-Selectin/metabolism , Phosphorylation/drug effects , Platelet Adhesiveness/drug effects , Polystyrenes/chemistry , Protein Aggregates , Spectrometry, Fluorescence , Surface Plasmon Resonance , Surface Properties , Tandem Mass Spectrometry , Thiazoles/metabolism
19.
PLoS One ; 12(6): e0180022, 2017.
Article in English | MEDLINE | ID: mdl-28662177

ABSTRACT

The toxicity of in vitro tested compounds is usually evaluated based on AC50 values calculated from dose-response curves. However, there is a large group of compounds for which a standard four-parametric sigmoid curve fitting may be inappropriate for estimating AC50. In the present study, 22 polyphenol-rich compounds were prioritized from the least to the most toxic based on the total area under and over the dose-response curves (AUOC) in relation to baselines. The studied compounds were ranked across three key cell indicators (mitochondrial membrane potential, cell membrane integrity and nuclear size) in a panel of five cell lines (HepG2, Caco-2, A549, HMEC-1, and 3T3), using a high-content screening (HCS) assay. Regarding AUOC score values, naringin (negative control) was the least toxic phenolic compound. Aronox, spent hop extract and kale leaf extract had very low cytotoxicity with regard to mitochondrial membrane potential and cell membrane integrity, as well as nuclear morphology (nuclear area). Kaempferol (positive control) exerted strong cytotoxic effects on the mitochondrial and nuclear compartments. Extracts from buckthorn bark, walnut husk and hollyhock flower were highly cytotoxic with regard to the mitochondrion and cell membrane, but not the nucleus. We propose an alternative algorithm for the screening of a large number of agents and for identifying those with adverse cellular effects at an early stage of drug discovery, using high content screening analysis. This approach should be recommended for series of compounds producing a non-sigmoidal cell response, and for agents with unknown toxicity or mechanisms of action.


Subject(s)
High-Throughput Screening Assays/methods , Plant Extracts/toxicity , Polyphenols/toxicity , Animals , Cell Line , Humans , Mice
20.
Platelets ; 27(5): 433-9, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26836594

ABSTRACT

Recently, polyphenols have gained attention as potential natural cardioprotective therapeutics, due to their antiplatelet, anti-inflammatory and anticoagulant activity. Species belonging to the genus Rubus sp. have been reported to be a source of polyphenolic compounds with antioxidative proprieties and beneficial biological activities. This study investigates the effects of leaf extracts obtained from red raspberry (Rubus idaeus L.) and European dewberry (Rubus caesius L.) on the reactivity of blood platelets. In ADP-stimulated blood, raspberry and dewberry extracts (15 µg/ml) markedly decreased platelet surface membrane expression of activated GPIIbIIIa receptor by 16% and 21%, respectively (P < 0.01) and significantly inhibited platelet aggregation (by 31-41% for raspberry and by 38-55% for dewberry, P < 0.01). In platelet-rich plasma (PRP), the extracts had no effect on ADP-induced platelet aggregation. The effectiveness of the extracts in whole blood and the lack of their activity in PRP indicate that leukocytes are likely to participate in the platelet response to the extracts. Our experiments show that the extracts significantly reduced the amount of free radicals released by activated neutrophils in whole blood (P < 0.001), as well as in suspensions of isolated neutrophils (P < 0.05). Moreover, the reduced number of neutrophils leads to the decreased efficiency of the extracts in the inhibition of platelet aggregation. In summary, our findings show that the raspberry and dewberry leaf extracts considerably modulated blood platelet reactivity in whole blood: they influenced blood platelet aggregation, possibly via the modulation of the redox status dependent on the oxidative activity of neutrophils.


Subject(s)
Blood Platelets/drug effects , Blood Platelets/metabolism , Plant Extracts/pharmacology , Platelet Aggregation/drug effects , Rubus/chemistry , Antioxidants/metabolism , Biomarkers , Cell Communication/drug effects , Cell Survival/drug effects , Humans , Leukocyte Count , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Phosphorylation , Platelet Activation/drug effects , Reactive Oxygen Species/metabolism , Respiratory Burst/drug effects , Respiratory Burst/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...