Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Int J Mol Sci ; 23(7)2022 Mar 27.
Article in English | MEDLINE | ID: mdl-35409027

ABSTRACT

Blood platelet dysfunctions are strongly involved in the development of the micro- and macrovascular complications in diabetes mellitus (DM). However, the molecular causes of abnormal platelet activation in DM remain unclear. Experimental data suggests that platelet mitochondria can regulate the prothrombotic phenotype of platelets, and changes in these organelles may influence platelet activation and modify platelet responses to stimulation. The present study evaluates the impact of DM on mitochondrial respiratory parameters and blood platelet activation/reactivity in a rat model of experimental diabetes following 1, 2.5 and 5 months of streptozotocin (STZ)-induced diabetes. Moreover, a mild inhibition of the mitochondrial respiratory chain with the use of metformin under in vitro and in vivo conditions was tested as a method to reduce platelet activation and reactivity. The platelets were studied with a combination of flow cytometry and advanced respirometry. Our results indicate that prolonged exposure of blood platelets to high concentrations of glucose, as in diabetes, can result in elevated blood platelet mitochondrial respiration; this may be an effect of cell adaptation to the high availability of energy substrates. However, as these alterations occur later than the changes in platelet activation/reactivity, they may not constitute the major reason for abnormal platelet functioning in DM. Moreover, metformin was not able to inhibit platelet activation and reactivity under in vitro conditions despite causing a decrease in mitochondrial respiration. This indicates that the beneficial effect of metformin on the coagulation system observed in vivo can be related to other mechanisms than via the inhibition of platelet activation.


Subject(s)
Diabetes Mellitus, Experimental , Metformin , Animals , Blood Platelets/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Energy Metabolism , Metformin/metabolism , Metformin/pharmacology , Mitochondria/metabolism , Platelet Activation , Rats
2.
Int J Mol Sci ; 22(17)2021 Aug 28.
Article in English | MEDLINE | ID: mdl-34502240

ABSTRACT

Blood platelets are considered as promising candidates as easily-accessible biomarkers of mitochondrial functioning. However, their high sensitivity to various stimulus types may potentially affect mitochondrial respiration and lead to artefactual outcomes. Therefore, it is crucial to identify the factors associated with platelet preparation that may lead to changes in mitochondrial respiration. A combination of flow cytometry and advanced respirometry was used to examine the effect of blood anticoagulants, the media used to suspend isolated platelets, respiration buffers, storage time and ADP stimulation on platelet activation and platelet mitochondria respiration. Our results clearly show that all the mentioned factors can affect platelet mitochondrial respiration. Briefly, (i) the use of EDTA as anticoagulant led to a significant increase in the dissipative component of respiration (LEAK), (ii) the use of plasma for the suspension of isolated platelets with MiR05 as a respiration buffer allows high electron transfer capacity and low platelet activation, and (iii) ADP stimulation increases physiological coupling respiration (ROUTINE). Significant associations were observed between platelet activation markers and mitochondrial respiration at different preparation steps; however, the fact that these relationships were not always apparent suggests that the method of platelet preparation may have a greater impact on mitochondrial respiration than the platelet activation itself.


Subject(s)
Anticoagulants/pharmacology , Blood Platelets/physiology , Cell Respiration/physiology , Culture Media/pharmacology , Mitochondria/physiology , Platelet Activation , Blood Platelets/drug effects , Flow Cytometry , Humans , Mitochondria/drug effects
3.
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
4.
Aging (Albany NY) ; 12(6): 5091-5120, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32191918

ABSTRACT

Aging has become a significant risk factor for several diseases, including breast cancer.Platelet activation and platelet-cancer cell aggregate fractions were found to increase with tumor progression in a mouse model of breast cancer. At advanced stages of tumor development, platelets from mice with breast cancer were hyperreactive to low agonist concentrations and hyporeactive to high ones. Platelet activation and reactivity were strongly associated with breast cancer metastasis in the lungs and extramedullary hematopoiesis in the liver. A greater fraction of platelet aggregates was observed in 4T1-injected mice at the advanced stages of breast cancer. In vitro, platelet activation was elevated after incubation with 4T1 cells, and thrombin-stimulated platelets formed aggregates with 4T1 cells. Neither GPIbα, nor GPIIb/IIIa blocking antibodies, were able to affect platelet-cancer cell aggregation in vitro.The primed circulating platelets became more sensitive to subthreshold stimuli at advanced stages of tumor development, and the formation of platelet-cancer cell aggregates increased with cancer progression. Our findings demonstrate that the age-associated progression of breast cancer cells is connected with increased platelet functioning, and that it can be manifested by the increased number of metastases and extramedullary hematopoiesis in a time-dependent-manner.


Subject(s)
Blood Platelets/pathology , Breast Neoplasms/pathology , Hematopoiesis, Extramedullary , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Platelet Activation , Animals , Breast Neoplasms/blood , Cell Line, Tumor , Disease Models, Animal , Female , Liver Neoplasms/blood , Lung Neoplasms/blood , Mice
5.
Int J Exp Pathol ; 100(1): 41-48, 2019 02.
Article in English | MEDLINE | ID: mdl-30811756

ABSTRACT

Diabetes is associated with an increased risk of cardiovascular disease. This is partially attributed to an altered activation status of blood platelets in this disease. Previously, alterations have been shown in COX-1 and protease activated receptor (PAR)-3 receptor expression in platelets in two animal models of diabetes, there have not been studies which address expression of these proteins in mice with long-term streptozotocin (STZ)-induced diabetes. We have also addressed the effect of diabetes on platelet adhesion under flow conditions. With the use of flow cytometry, we have shown that certain markers of platelet basal activation, such as active form of αIIb ß3 and of CD40L were increased in STZ-induced diabetic mice. Platelets from STZ-induced diabetic mice were also more reactive when stimulated with PAR-4 activating peptide as revealed by higher expression of active form of αIIb ß3 , membrane-bound on vWillebrand Factor and binding of exogenous fluorescein isothyanate-labelled fibrinogen. Expression of COX-1 and production of thromboxane A2 in platelets of STZ-induced diabetic mice were higher than in control animals. We observed no effect of diabetes on ability of platelets to form stable adhesions with fibrinogen in flow conditions. We conclude that although certain similarities exist between patterns of activation of platelets in animal models of diabetes, the differences should also be taken into account.


Subject(s)
Blood Platelets/enzymology , Cyclooxygenase 1/blood , Diabetes Mellitus, Experimental/blood , Membrane Proteins/blood , Platelet Adhesiveness , Streptozocin , Animals , CD40 Ligand/blood , Coronary Vessels/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/enzymology , Epoprostenol/metabolism , Male , Mice, Inbred C57BL , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Receptors, Proteinase-Activated/blood , Thromboxane A2/blood , von Willebrand Factor/metabolism
6.
Int J Chron Obstruct Pulmon Dis ; 13: 3707-3717, 2018.
Article in English | MEDLINE | ID: mdl-30519014

ABSTRACT

BACKGROUND: COPD represents a major global health issue, which is often accompanied by cardiovascular diseases. A considerable body of evidence suggests that cardiovascular risk is elevated by the activation of blood platelets, which in turn is exacerbated by inflammation. As reactive oxygen species are believed to be an important factor in platelet metabolism and functioning, the aim of our study was to perform a complex assessment of mitochondrial function in platelets in chronic smoke exposed animals with COPD-like lung lesions. MATERIALS AND METHODS: Eight-week-old, male Dunkin Hartley guinea pigs (the study group) were exposed to the cigarette smoke from commercial unfiltered cigarettes (0.9 mg/cig of nicotine content) or to the air without cigarette smoke (control group), using the Candela Constructions® exposure system. The animals were exposed for 4 hours daily, 5 days a week, with 2×70 mL puff/minute, until signs of dyspnea were observed. The animals were bled, and isolated platelets were used to monitor blood platelet respiration. The mitochondrial respiratory parameters of the platelets were monitored in vitro based on continuous recording of oxygen consumption by high-resolution respirometry. RESULTS: An elevated respiration trend was observed in the LEAK-state (adjusted for number of platelets) in the smoke-exposed animals: 6.75 (5.09) vs 2.53 (1.28) (pmol O2/[s ⋅ 1108 platelets]); bootstrap-boosted P 1α=0.04. The study group also demonstrated lowered respiration in the ET-state (normalized for protein content): 12.31 (4.84) vs 16.48 (1.72) (pmol O2/[s ⋅ mg of protein]); bootstrap-boosted P 1α=0.049. CONCLUSION: Our results suggest increased proton and electron leak and decreased electron transfer system capacity in platelets from chronic smoke-exposed animals. These observations may also indicate that platelets play an important role in the pathobiology of COPD and its comorbidities and may serve as a background for possible therapeutic targeting. However, these preliminary outcomes should be further validated in studies based on larger samples.


Subject(s)
Blood Platelets/pathology , Cigarette Smoking/pathology , Mitochondria/pathology , Pulmonary Disease, Chronic Obstructive/pathology , Smoke , Animals , Blood Platelets/metabolism , Cell Respiration , Cigarette Smoking/blood , Cigarette Smoking/physiopathology , Disease Models, Animal , Guinea Pigs , Inhalation Exposure , Lung/metabolism , Lung/pathology , Lung/physiopathology , Male , Mitochondria/metabolism , Pilot Projects , Pulmonary Disease, Chronic Obstructive/blood , Reactive Oxygen Species/blood
7.
Blood Cells Mol Dis ; 65: 16-22, 2017 06.
Article in English | MEDLINE | ID: mdl-28460264

ABSTRACT

INTRODUCTION: Recent studies have shown that it may be the concentration of thrombin, which is discriminative in determining of the mechanism of platelet activation via protease activated receptors (PARs). Whether the observed phenomenon of differentiated responses of mouse platelets to various thrombin concentrations in non-diabetic db/+ and diabetic db/db mice depends upon the concerted action of various PARs, remains to be established. RESULTS: We found elevated reactivity of platelets, as well as the enhanced PAR-3 expression in response to both the used concentrations of AYPGKF in db/db mice, as compared to db/+ heterozygotes. At low concentration of thrombin platelets from diabetic mice demonstrated hyperreactivity, reflected by higher expression of PAR-3. For higher thrombin concentration, blood platelets from db/db mice appeared hyporeactive, compared to db/+ animals, while no significant differences in PAR-3 expression were observed between diabetic and non-diabetic mice. CONCLUSIONS: The novel and previously unreported finding resulting from our study is that the increased expression of PAR-3 in response to either TRAP for PAR-4 or low thrombin (when PAR-4 is not the efficient thrombin receptor) may be one of the key events contributing to higher reactivity of platelets in db/db mice.


Subject(s)
Blood Platelets/metabolism , Cell Adhesion Molecules/metabolism , Platelet Activation , Tartrate-Resistant Acid Phosphatase/metabolism , Thrombin/metabolism , Adaptor Proteins, Signal Transducing , Animals , Biomarkers , Blood Platelets/drug effects , Cell Adhesion Molecules/genetics , Cell Cycle Proteins , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Flow Cytometry , Gene Expression , Male , Mice , Oligopeptides/pharmacology , Platelet Activation/drug effects , Platelet Activation/genetics
8.
Chem Biol Interact ; 253: 78-92, 2016 Jun 25.
Article in English | MEDLINE | ID: mdl-27083140

ABSTRACT

Due to its ability to inhibit the blood platelet PGHS-1, acetylsalicylic acid (ASA, Aspirin(®)) is widely used as a preventive agent in atherothrombotic diseases. However, its beneficial effects seem to be lower in diabetic patients, suggesting that protein glycation may impair effective ASA-mediated acetylation process. On the other hand, it is proposed that ASA can prevent some of the late complications of diabetes by lowering the extent of glycation at protein free amino groups. The aim of this work was to evaluate the extents of non-enzymatic N-glycosylation (glycation) and acetylation of blood platelet PGHS-1 (COX-1) and the competition between glycation and acetylation was investigated in order to demonstrate how these two reactions may compete against platelet PGHS-1. When PGHS-1 was incubated with glycating/acetylating agents (glucose, Glu; 1,6-bisphosphofructose, 1,6-BPF; methylglyoxal, MGO, acetylsalicylic acid, ASA), the enzyme was modified in 13.4 ± 1.6, 5.3 ± 0.5, 10.7 ± 1.2 and 6.4 ± 1.1 mol/mol protein, respectively, and its activity was significantly reduced. The prior glycation/carbonylation of PGHS-1 with Glu, 1,6-BPF or MGO decreased the extent of acetylation from 6.4 ± 1.1 down to 2.5 ± 0.2, 3.6 ± 0.3 and 5.2 ± 0.2 mol/mol protein, respectively, but the enzyme still remained susceptible to the subsequent inhibition of its activity with ASA. When PGHS-1 was first acetylated with ASA and then incubated with glycating/carbonylating agents, we observed the following reductions in the enzyme modifications: from 13.4 ± 1.6 to 8.7 ± 0.6 mol/mol protein for Glu, from 5.3 ± 0.5 to 3.9 ± 0.3 mol/mol protein for 1,6-BPF and from 10.7 ± 1.2 to 7.5 ± 0.5 mol/mol protein for MGO, however subsequent glycation/carbonylation did not significantly affect PGHS-1 function. Overall, our outcomes allow to better understand the structural aspects of the chemical competition between glycation and acetylation of PGHS-1.


Subject(s)
Aspirin/pharmacology , Cyclooxygenase 1/metabolism , Protein Processing, Post-Translational/drug effects , Acetylation/drug effects , Blood Platelets/cytology , Blood Platelets/drug effects , Blood Platelets/metabolism , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Glycosylation/drug effects , Humans , Isotope Labeling , Peptides/analysis , Tandem Mass Spectrometry
10.
Life Sci ; 148: 293-304, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26872978

ABSTRACT

AIMS: The high glucose concentration observed in diabetic patients is a recognized factor of mitochondrial damage in various cell types. Its impact on mitochondrial bioenergetics in blood platelets remains largely vague. The aim of the study was to determine how the metabolism of carbohydrates, which has been impaired by streptozotocin-induced diabetes may affect the functioning of platelet mitochondria. MATERIALS AND METHODS: Diabetes was induced in Sprague Dawley rats by intraperitoneal injection of streptozotocin. Platelet mitochondrial respiratory capacity was monitored as oxygen consumption (high-resolution respirometry). Mitochondrial membrane potential was assessed using a fluorescent probe, JC-1. Activation of circulating platelets was monitored by flow cytometry measuring of the expressions of CD61 and CD62P on a blood platelet surface. To determine mitochondrial protein density in platelets, Western Blot technique was used. KEY FINDINGS: The results indicate significantly elevated mitochondria mass, increased mitochondrial membrane potential (ΔΨm) and enhanced respiration in STZ-diabetic animals, although the respiration control ratios appear to remain unchanged. Higher ΔΨm and elevated mitochondrial respiration were closely related to the excessive activation of circulating platelets in diabetic animals. SIGNIFICANCE: Long-term diabetes can result in increased mitochondrial mass and may lead to hyperpolarization of blood platelet mitochondrial membrane. These alterations may be a potential underlying cause of abnormal platelet functioning in diabetes mellitus and hence, a potential target for antiplatelet therapies in diabetes.


Subject(s)
Blood Platelets/metabolism , Diabetes Mellitus, Experimental/metabolism , Membrane Potential, Mitochondrial/physiology , Mitochondria/metabolism , Oxygen Consumption/physiology , Animals , Cell Respiration/physiology , Male , Rats , Rats, Sprague-Dawley
11.
Int J Pharm ; 500(1-2): 379-89, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26772535

ABSTRACT

We explored the hypothesis that zeta potential altered by polycations affects blood platelet activation and reactivity, the phenomena associated with membrane lipid fluidity and platelet mitochondrial bioenergetics. PAMAM dendrimers generation- and dose-dependently enhanced zeta potential of platelets (from -10.7 mV to -4.3 mV). Increased expressions of activation markers, P-selectin and the active complex αIIbß3, as well as significantly enhanced fibrinogen binding occurred upon the in vitro incubation of blood platelets in the presence of PAMAMs G3 and G4 (resp. 62.1% and 69.4% vs. 1.4% and 2.7% in control for P-selectin, P<0.0001). PAMAM dendrimers increased fluidity of platelet membrane lipid bilayer, while they did not affect platelet mitochondria respiration. Increased platelet activation and their responses to agonists in vitro were statistically associated with the revealed alterations in zeta potential. Our results support the hypothesis that polycation-mediated "neutralized" zeta potential may underlie the activating effects of PAMAMs on blood platelets.


Subject(s)
Blood Platelets/drug effects , Dendrimers/pharmacology , Platelet Activation/drug effects , Adult , Blood Platelets/physiology , Cell Membrane , Electron Spin Resonance Spectroscopy , Female , Humans , Male , Mitochondria/drug effects , Mitochondria/metabolism , Oxygen/metabolism , Young Adult
12.
Platelets ; 27(3): 203-11, 2016.
Article in English | MEDLINE | ID: mdl-26325148

ABSTRACT

In diabetes-related states of chronic hyperglycaemia elevated concentrations of glucose may alter the functioning of platelet enzymes involved in arachidonic acid metabolism, including prostaglandin H2 synthase (cyclooxygenase) (PGHS, COX). Therefore, the principal aim of this study was to assess the effects of experimental chronic hyperglycaemia on platelet PGHS-1 (COX-1) expression and activity. Blood platelet activation and reactivity were assessed in Sprague-Dawley rats with the 5-month streptozotocin (STZ) diabetes. The PGHS-1 abundance in platelets was evaluated with flow cytometry and Western blotting, while its activity monitored using a high resolution respirometry and the peroxidase fluorescent assay. The production of prostaglandin E2 (PGE2) and thromboxane B2 (TXB2) in platelets were assayed immunoenzymatically. Circulating platelets from diabetic were characterised by increased size, elevated 'priming' and altered reactivity, compared to non-diabetic animals. Both Western blot analysis and flow cytometry revealed significantly elevated expressions of platelet PGHS-1 in STZ-diabetic rats (p < 0.05). We also observed significantly elevated platelet PGHS-1-related arachidonic acid metabolism in diabetic vs. non-diabetic animals, with the use of polarographic (p < 0.05) and total activity assay (p < 0.001). Such increases were accompanied by the elevated production of PGE2 (p < 0.001) and TXB2 (p < 0.05) in diabetic animals. The increased PGHS-1-dependent oxygen consumption and the total activity of PGHS-1 in diabetic animals remained very significant (p < 0.001) also upon adjusting for blood platelet PGHS-1 abundance. Therefore, our results further contribute to the explanation of the increased metabolism of arachidonic acid observed in diabetes.


Subject(s)
Blood Platelets/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Gene Expression , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Arachidonic Acid/pharmacology , Biomarkers , Blood Platelets/drug effects , Cyclooxygenase 1/metabolism , Diabetes Mellitus, Experimental/blood , Dinoprostone/biosynthesis , Disease Models, Animal , Enzyme Activation , Male , Platelet Activation , Platelet Aggregation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Thromboxane B2/biosynthesis
13.
Thromb Res ; 133(6): 1097-104, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24731558

ABSTRACT

Currently, there are several animal models of diabetes mellitus and hypertension, but relatively little is known about blood platelet function in these models. The aim of this work was to characterise and compare platelet reactivity and activation in db/db mice (mouse model of diabetes) and mice receiving L-NAME (model of chronic inhibition of NO synthesis), using various platelet function assays. We found higher platelet activation (circulating resting platelets) in db/db mice than in db/+heterozygotes, as evidenced by elevated expressions of CD62P and CD40L and a lower expression of CD42b. The expression of COX-1 was significantly increased, and the phosphorylation of vasodilator stimulated phosphoprotein (VASP) Ser(157) significantly reduced in platelets from db/db mice. Similarly, we observed platelet hyperreactivity in db/db mice following the in vitro responses to 20µg/ml collagen (reflected by increased expressions of CD62P and CD40L, and reduced CD42b), 20µM ADP (reduced CD42b) and lower concentrations of thrombin (0.025 U/ml) (increased CD62P, JON/A, bound vWF, and bound fibrinogen). Otherwise, platelet hyporeactivity was revealed for higher thrombin (0.25 U/ml) (reduced CD62P and bound vWF), while hyperreactivity occurred for CD40L and bound Fg in db/db mice compared to non-diabetic control, db/+. Plasma levels of sCD40L, but not of sCD62P, were increased in db/db mice; also plasma TXB2 concentrations were over 3.5-fold higher in this group than in the heterozygous db/+mice (P<0.01). In contrast, in the mice administered with L-NAME, no statistical differences in expressions of platelet activation markers were found between mice supplemented with L-NAME and controls. Likewise, the TXB2 level did not differ between L-NAME mice and controls, but L-NAME mice had significantly higher plasma levels of sCD62P and sCD40L than controls. In conclusion, these two studied models differ in the overall picture of blood platelet activation and reactivity, as they demonstrated opposite time sequence patterns of platelet activation in circulating blood. More generally, our study provides another argument for the opinion that multiparametric analysis of platelet function offers a much better tool for investigation and minimizes the likelihood of artefacts.


Subject(s)
Blood Platelets/physiology , Diabetes Mellitus, Experimental/blood , Nitric Oxide Synthase/antagonists & inhibitors , Platelet Activation/physiology , Platelet Aggregation/physiology , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , CD40 Ligand/blood , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/enzymology , Hypertension/blood , Hypertension/enzymology , Male , Mice , Mice, Inbred C57BL , NG-Nitroarginine Methyl Ester/pharmacology , P-Selectin/blood , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Random Allocation , Thromboxane B2/blood
14.
Molecules ; 18(11): 13769-811, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24213655

ABSTRACT

Diabetes mellitus, which is characterised by high blood glucose levels and the burden of various macrovascular and microvascular complications, is a cause of much human suffering across the globe. While the use of exogenous insulin and other medications can control and sometimes prevent various diabetes-associated sequelae, numerous diabetic complications are still commonly encountered in diabetic patients. Therefore, there is a strong need for safe and effective antihyperglycaemic agents that provide an alternative or compounding option for the treatment of diabetes. In recent years, amino-terminated poly(amido)amine (PAMAM) dendrimers (G2, G3 and G4) have attracted attention due to their protective value as anti-glycation and anti-carbonylation agents that can be used to limit the nonenzymatic modifications of biomacromolecules. The focus of this review is to present a detailed survey of our own data, as well as of the available literature regarding the toxicity, pharmacological properties and overall usefulness of PAMAM dendrimers. This presentation pays particular and primary attention to their therapeutic use in poorly controlled diabetes and its complications, but also in other conditions, such as Alzheimer's disease, in which such nonenzymatic modifications may underlie the pathophysiological mechanisms. The impact of dendrimer administration on the overall survival of diabetic animals and on glycosylation, glycoxidation, the brain-blood barrier and cellular bioenergetics are demonstrated. Finally, we critically discuss the potential advantages and disadvantages accompanying the use of PAMAM dendrimers in the treatment of metabolic impairments that occur under conditions of chronic hyperglycaemia.


Subject(s)
Dendrimers/therapeutic use , Animals , Dendrimers/adverse effects , Diabetes Mellitus/drug therapy , Humans , Hyperglycemia/drug therapy , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/therapeutic use
15.
Pharmacol Rep ; 65(4): 870-83, 2013.
Article in English | MEDLINE | ID: mdl-24145081

ABSTRACT

BACKGROUND: The effects of blood platelet inhibitors are often not quite equivalent under in vivo and in vitro conditions. Amongst various models of human pathology using laboratory animals, mice offer several benefits that make them convenient tools for studying the putative therapeutic value of various compounds. However, despite its advantages, the mouse model has methodological limitations concerning the small amount of blood available and technical difficulties with its collection. Among the variety of available methods used to study blood platelet activation and/or reactivity, flow cytometry seems an attractive technique that largely minimizes the constraints of using small rodents and enables outcomes of laboratory research to be transferred successfully to clinical practice. In this study we aimed at a critical evaluation of the optimal discriminative flow cytometric protocol, useful for reliable studies of the effect of cangrelor, a P2Y12 receptor antagonist, on mouse platelets under in vitro and in vivo conditions. METHODS: Blood samples were drawn from two-month-old female BALB/c mice. Protocols differing in methods of anesthesia, blood withdrawal, anticoagulation, gating antibodies, blood preparation and fixation were tested to optimize the one best suited to discrimination between resting and activated platelets. The antiplatelet capabilities of cangrelor were tested in vitro (140 µM in whole blood) and in vivo (7.8 mg/kg b.w. administered once, directly into the bloodstream through the vena cava of the anesthetized animal, 15 min prior to blood withdrawal). Expressions of P-selectin, activated α(IIb)ß3 complex and GPIba were monitored using two-color flow cytometry. RESULTS: "Washed blood" anticoagulated with low molecular weight heparin demonstrated the best discrimination between circulating (resting) platelets and upon their in vitro response to thrombin, collagen or ADP in freshly-stained unfixed cell suspensions. Cangrelor inhibited the expression of the active form of the integrin a(IIb)ß3 to approximately the same extent under in vitro and in vivo conditions (84.5 ± 7.7% vs. 75.4 ± 19.5% for the in vitro and in vivo approaches, respectively, n.s.). CONCLUSIONS: The agreement between the in vivo and in vitro approaches with respect to cangrelor-inhibited hallmarks of blood platelet activation and reactivity supports our proposal that flow cytometry is useful and reliable for determining the effects of antiplatelet agents on the activation of circulating platelets in the mouse model, as well as the in vitro response of platelets to agonists.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Blood Platelets/drug effects , Flow Cytometry , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Adenosine Diphosphate/pharmacology , Adenosine Monophosphate/pharmacology , Animals , Blood Platelets/metabolism , Collagen/pharmacology , Female , Mice , Models, Animal , P-Selectin/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Purinergic P2Y Receptor Antagonists/pharmacology , Thrombin/pharmacology
16.
Thromb Haemost ; 108(1): 107-18, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22627503

ABSTRACT

Leptin is an adipokine that in vitro enhances agonist-induced platelet aggregation and adipokine expression. Hyperleptinaemia represents a risk factor for cardiovascular disease. We conducted a prospective evaluation of the potential link between blood platelet activation and plasma leptin levels in post-stroke patients. Using five-colour flow cytometry, the platelet surface expression of CD40L, CD62P, the subpopulations of monocyte-platelet aggregates and platelet-derived microparticles (PMPs) as well as the plasma leptin, soluble leptin receptor (sOb-R), leptin/sOb-R ratio, the plasma adiponectin, and leptin/adiponectin ratio were assessed in 98 stroke patients on the first (V0), 10th (V1 ) and 90th (V2) day after stroke and once in 78 age-, gender- and vascular risk factor-matched disease controls. We demonstrated that at V0 leptin resistance, defined as leptin/sOb-R ratio, was higher than in the controls [1.1 (0.5-1.8 vs. 0.5 (0.2-1.1); p=0.02]. After adjustment according to the factors which influence platelet activation, we confirmed the relationship between percentage of circulating PMPs and plasma leptin level (B=0.18; p=0.02) or the leptin/sOb-R ratio (B=0.23; p=0.02) in normal-weight subjects in the acute phase of stroke. No correlation could be demonstrated between the adipokine parameters and the percentage of monocyte-platelet aggregates or expression of platelet pro-inflammatory glycoproteins. In conclusion, formation of PMPs on the first day following an ischaemic stroke shows a positive correlation with leptin levels and with resistance to leptin. Leptin level does not seem to affect the expression of platelet surface proinflammatory glycoproteins.


Subject(s)
Blood Platelets/metabolism , Leptin/blood , Platelet Activation , Stroke/diagnosis , Acute Disease , Adiponectin/blood , Aged , Blood Platelets/pathology , CD40 Ligand/metabolism , Cell Separation , Cell-Derived Microparticles/pathology , Disease Progression , Female , Flow Cytometry , Humans , Male , Middle Aged , Monocytes/metabolism , Monocytes/pathology , P-Selectin/metabolism , Prospective Studies , Stroke/pathology , Stroke/physiopathology
17.
Int J Pharm ; 430(1-2): 258-65, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22486957

ABSTRACT

Poly(amido)amine (PAMAM) dendrimer G3 was investigated for its ability to support the proper functioning of rat heart mitochondria exposed to hyperglycemia, in both the in vitro and in vivo experiments. The main aims of this study were to check whether PAMAM G3 dendrimer improves the efficiency of the impaired respiration of rat heart mitochondria. This study showed that mitochondria isolated from animals studied in different seasons respond to G3 (100 µM) exposure to a different extent. Probably, seasonal variations had the impact on rat metabolism and consequently on the received data. The used biological samples formed a heterogenous group and therefore the obtained results were not pooled together but treated separately. Nevertheless, the in vitro part of this study revealed that PAMAM G3 could be successfully used in the protection of heart mitochondria against MG-induced impaired respiratory activity. Despite these promising data, the protective effect of G3 was not confirmed in the in vivo experiment. This study revealed that dendrimer G3 (20 mg/kgbw) is toxic and very high mortality among the animals administered with G3 did not allow to perform a reliable data analysis.


Subject(s)
Dendrimers/toxicity , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Energy Metabolism/drug effects , Mitochondria, Heart/metabolism , Animals , Cell Respiration/drug effects , Cytoprotection , Dendrimers/administration & dosage , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Type 1/chemically induced , Glycation End Products, Advanced/metabolism , Injections, Intraperitoneal , Male , Mitochondria, Heart/drug effects , Pyruvaldehyde/metabolism , Rats , Rats, Wistar , Seasons
18.
Int J Mol Sci ; 12(11): 8013-26, 2011.
Article in English | MEDLINE | ID: mdl-22174647

ABSTRACT

Diabetes is associated with a mitochondrial dysfunction. Hyperglycaemia is also clearly recognized as the primary culprit in the pathogenesis of cardiac complications. In response to glycation and oxidative stress, cardiac mitochondria undergo cumulative alterations, often leading to heart deterioration. There is a continuous search for innovative treatment strategies for protecting the heart mitochondria from the destructive impact of diabetes. Aminoguanidine derivatives have been successfully used in animal model studies on the treatment of experimental diabetes, as well as the diabetes-driven dysfunctions of peripheral tissues and cells. Considerable attention has been paid particularly to ß-resorcylidene aminoguanidine (RAG), often shown as the efficient anti-glycation and anti-oxidant agent in both animal studies and in vitro experiments. The aim of the present study was to test the hypothesis that RAG improves oxidative phosphorylation and electron transport capacity in mitochondria impaired by hyperglycaemia. Diabetes mellitus was induced in Wistar rats by a single intraperitoneal injection of streptozotocin (70 mg/kg body weight). Heart mitochondria were isolated from healthy rats and rats with streptozotocin-diabetes. Mitochondrial respiratory capacity was measured by high resolution respirometry with the OROBOROS Oxygraph-2k according to experimental protocol including respiratory substrates and inhibitors. The results revealed that RAG protects the heart against diabetes-associated injury by improving the mitochondrial bioenergetics, thus suggesting a possible novel pharmacological strategy for cardioprotection.


Subject(s)
Energy Metabolism/drug effects , Guanidines/pharmacology , Heart/drug effects , Hyperglycemia/drug therapy , Mitochondria, Heart/drug effects , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/drug therapy , Electron Transport , Enzyme Inhibitors/pharmacology , Heart/physiology , Male , Mitochondria, Heart/metabolism , Oxidative Phosphorylation/drug effects , Oxidative Stress/drug effects , Oxygen Consumption/drug effects , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...