Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Toxicon ; 228: 107097, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37028563

ABSTRACT

Rhomb-I, a 23-kDa metalloproteinase was isolated from L. m. rhombeata venom. Its dimethylcasein proteolysis was abolished by metal chelators, and slightly enhanced by Ca2+ and Mg2+ ions, but inhibited by Co2+, Zn2+ and α2-macroglobulin. In aqueous solution, rhomb-I autoproteolyzed to a 20- and 11-kDa fragments at 37 °C. The amino acid sequence showed high homology with other snake venom metalloproteinases. Rhomb-I causes hemorrhage that may be ascribed to hydrolysis of essential basement membrane, extracellular matrix and plasma proteins. It preferentially cleaves the α-chains of fibrin (ogen). Rhomb-I inhibited convulxin- and von Willebrand factor (vWF)-induced aggregation on human platelets without significant effect on collagen-stimulated aggregation or other effectors. It digests vWF into a low-molecular-mass multimers of vWF and a rvWF-A1 domain to a 27-kDa fragment as revealed by western blotting with mouse anti-rvWF A1-domain IgG. Incubation of platelets with rhomb-I resulted in adhesion to and cleavage of platelet receptors glycoprotein (GP)Ibα and GPVI to release a 55-kDa soluble form. Both membrane glycoproteins GPIbα that binds vWF, together with GPVI which binds collagen, play a key role in mediating platelet adhesion/activation and can initiate (patho)physiological thrombus formation. Conclusions: rhomb-I is implicated in the pathophysiology of Lachesis envenoming by disrupting vasculature, hemostasis and platelet aggregation through impairing vWF-GPIb axis and blocking GPVI-collagen binding.


Subject(s)
Platelet Aggregation , von Willebrand Factor , Humans , Animals , Mice , von Willebrand Factor/metabolism , Metalloproteases/metabolism , Blood Platelets , Collagen/metabolism
2.
Comput Methods Programs Biomed ; 231: 107390, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36745955

ABSTRACT

PURPOSE: The objective of this study is to develop a bleeding risk model for assessing device-induced bleeding risk in patients supported with blood contact medical devices (BCMDs). METHODS: The mathematical model for evaluating bleeding risk considers the effects of shear stress on von Willebrand factor (vWF) unfolding, high molecular weight multimers-vWF (HMWM-vWF) degradation, platelet activation and receptor shedding and platelet-vWF binding ability. Functions of the effect of shear stress on the above factors are fitted/employed and solved by the Eulerian transport equation. An axial flow-through Couette device and two clinical VADs which are HeartWare Ventricular Assist Device (HVAD) and HeartMate II (HM II) blood pump were employed to perform the simulation to evaluate platelet receptor shedding (GPIbα and GPIIb/IIIa), loss of HWMW-vWF, platelet-vWF binding ability and bleeding risk for validating the accuracy of our model. RESULTS: The platelet-vWF binding ability after being subjected to high shear region in the axial flow-through Couette device predicted by our bleeding model was highly consistent with reported experimental data. As indicated by our CFD simulation results in the axial flow-through Couette device, it can find that an increase in shear stress led to a decrease in the adhesion ability of platelets on vWF, while the binding ability of vWF with platelets first increase and then decrease as shear stress elevates gradually beyond a threshold. The factor of exposure time can enhance the effect of shear stress. Additionally, the shear-induced bleeding risk predicted by our model increases with increasing shear stress and exposure time in an axial flow-through Couette device. As indicated by our numerical model, the bleeding risk in HVAD was higher than HMII, which is highly consistent with the meta-analysis based on clinical statistics. Our simulation investigations in these two clinical VADs also found that HVAD caused a higher rate of platelet receptor shedding and lower damage to HWMW-vWF than HeartMate II. The high shear stress generated in the narrow and turbulent regions of both VADs was the underlying cause of device-induced bleeding. CONCLUSION: In this study, the shear-induced bleeding risk predicted by our bleeding model in axial flow-through Couette device and two clinical VADs is consistent or highly correlated with experimental and clinical findings, which proves the accuracy of our bleeding model. Our bleeding model can be used to aid the development of new BCMDs with improved functional characteristics and biocompatibility, and help to reduce risk of device-induced adverse events in patients.


Subject(s)
Heart-Assist Devices , von Willebrand Factor , Humans , von Willebrand Factor/analysis , Hemorrhage/etiology , Hemorrhage/metabolism , Platelet Activation , Blood Platelets/chemistry , Blood Platelets/metabolism , Stress, Mechanical , Models, Theoretical
3.
Am J Blood Res ; 13(6): 168-188, 2023.
Article in English | MEDLINE | ID: mdl-38223314

ABSTRACT

Cardiovascular disease (CVD) is a major cause of death worldwide. Although there are many variables that contribute to the development of this disease, it is predominantly the activity of platelets that provides the mechanisms by which this disease prevails. While there are numerous platelet receptors expressed on the surface of platelets, it is largely the consensus that there are 10 main platelet receptors that contribute to a majority of platelet function. Understanding these key platelet receptors is vitally important for patients suffering from myocardial infarction, CVD, and many other diseases that arise due to overactivation or mutations of these receptors. The goal of this manuscript is to review the main platelet receptors that contribute most to platelet activity.

4.
Toxicon, V. 228, 107097, mar. 2023
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: bud-4853

ABSTRACT

Rhomb-I, a 23-kDa metalloproteinase was isolated from L. m. rhombeata venom. Its dimethylcasein proteolysis was abolished by metal chelators, and slightly enhanced by Ca2+ and Mg2+ ions, but inhibited by Co2+, Zn2+ and α2-macroglobulin. In aqueous solution, rhomb-I autoproteolyzed to a 20- and 11-kDa fragments at 37 °C. The amino acid sequence showed high homology with other snake venom metalloproteinases. Rhomb-I causes hemorrhage that may be ascribed to hydrolysis of essential basement membrane, extracellular matrix and plasma proteins. It preferentially cleaves the α-chains of fibrin (ogen). Rhomb-I inhibited convulxin- and von Willebrand factor (vWF)-induced aggregation on human platelets without significant effect on collagen-stimulated aggregation or other effectors. It digests vWF into a low-molecular-mass multimers of vWF and a rvWF-A1 domain to a 27-kDa fragment as revealed by western blotting with mouse anti-rvWF A1-domain IgG. Incubation of platelets with rhomb-I resulted in adhesion to and cleavage of platelet receptors glycoprotein (GP)Ibα and GPVI to release a 55-kDa soluble form. Both membrane glycoproteins GPIbα that binds vWF, together with GPVI which binds collagen, play a key role in mediating platelet adhesion/activation and can initiate (patho)physiological thrombus formation. Conclusions: rhomb-I is implicated in the pathophysiology of Lachesis envenoming by disrupting vasculature, hemostasis and platelet aggregation through impairing vWF-GPIb axis and blocking GPVI-collagen binding.

5.
Urologiia ; (5): 29-33, 2022 Nov.
Article in Russian | MEDLINE | ID: mdl-36382814

ABSTRACT

AIM: of the study is to identify risk factors for the development of acute pyelonephritis after contact urethrolithotripsy (URLLT) and to establish the mechanisms for maintaining inflammation after the withdrawal of NSAIDs. MATERIAL AND METHODS: The study included 21 patients who underwent contact ureterolithotripsy (URLT). The severity of leukocyturia was assessed 1 day after URLT, 2 days (the last appointment of NSAIDs, the total duration of the drug was 9 days) and 3 days (24 hours after NSAID discontinuation). The number of circulating platelet-leukocyte aggregates (PLA) was calculated by microscopy of stained blood smears. Analysis of the functional activity of platelet receptors involved in the modulation of the acute inflammatory response was performed by the turbidimetric method on a ChronoLog analyzer (USA).Statistical analysis was performed using the MedCalc package. RESULTS: After URSL, when NSAIDs were prescribed to patients, the level of leukocyturia decreased (p<0.05) compared to that at the time of hospitalization. A similar dynamics was found by analyzing the amount of TLA in the blood. Similar dynamics was found in the analysis of the amount of TLA in the blood. After 24 hours of NSAIDs cancellation, an increase in the severity of leukocyturia was detected (p<0.001). At the same time, normoreactivity of the 2-adrenergic receptor, GPVI receptor, AT1 receptor, PAT receptor, P2X1 receptor and A2A receptor, as well as hyporeactivity of the 2-adrenergic receptor and P2Y receptors, were revealed. An analysis of correlations made it possible to establish that the 2-adrenoreceptor, AT1 receptor, and GPVI receptor play a key role in the formation of TPA. Incubation of blood cells in vitro with agonists made it possible to establish that the maximum effect of TLA formation was reproduced during the interaction of the 2-adrenergic receptor and the AT1 receptor. CONCLUSION: With the abolition of NSAIDs, activation of the sympathetic-adrenal and renin-angiotensin systems, as well as remodeling of the basement membrane of the vascular wall are risk factors for the development of acute pyelonephritis after URLS.


Subject(s)
Pyelonephritis , Receptor, Angiotensin, Type 1 , Humans , Pyelonephritis/etiology , Risk Factors , Receptors, Adrenergic , Anti-Inflammatory Agents, Non-Steroidal
6.
Int J Mol Sci ; 23(15)2022 Aug 03.
Article in English | MEDLINE | ID: mdl-35955743

ABSTRACT

Glycoprotein (GP) VI is the major platelet collagen receptor and a promising anti-thrombotic target. This was first demonstrated in mice using the rat monoclonal antibody JAQ1, which completely blocks the Collagen-Related Peptide (CRP)-binding site on mouse GPVI and efficiently inhibits mouse platelet adhesion, activation and aggregation on collagen. Here, we show for the first time that JAQ1 cross-reacts with human GPVI (huGPVI), but not with GPVI in other tested species, including rat, rabbit, guinea pig, swine, and dog. We further demonstrate that JAQ1 differently modulates mouse and human GPVI function. Similar to its effects on mouse GPVI (mGPVI), JAQ1 inhibits CRP-induced activation in human platelets, whereas, in stark contrast to mouse GPVI, it does not inhibit the adhesion, activation or aggregate formation of human platelets on collagen, but causes instead an increased response. This effect was also seen with platelets from newly generated human GPVI knockin mice (hGP6tg/tg). These results indicate that the binding of JAQ1 to a structurally conserved epitope in GPVI differently affects its function in human and mouse platelets.


Subject(s)
Platelet Adhesiveness , Platelet Membrane Glycoproteins , Animals , Blood Platelets/metabolism , Collagen/metabolism , Dogs , Epitopes/metabolism , Guinea Pigs , Humans , Mice , Platelet Activation , Platelet Aggregation , Platelet Membrane Glycoproteins/metabolism , Rabbits , Rats
7.
Front Cardiovasc Med ; 9: 854813, 2022.
Article in English | MEDLINE | ID: mdl-35647068

ABSTRACT

Dual antiplatelet therapy comprising of aspirin and oral P2Y12 receptor antagonists are an established cornerstone of therapy in acute coronary syndromes and percutaneous coronary intervention. As a result, the platelet P2Y12 receptor remains a key therapeutic target in cardiovascular medicine since pharmacological antagonists were first developed in the 1990's. With a greater understanding of platelet biology and the role played by the P2Y12 receptor in the amplification of platelet activation and thrombus formation, there has been progressive refinement in the development of P2Y12 receptor antagonists with greater potency and consistency of antiplatelet effect. However, challenges remain in the utilization of these agents particularly in balancing the need for greater protection from ischemic events whilst minimizing the bleeding risk and present a real opportunity for the institution of individualized medicine. Future drug developments will provide clinicians with greater avenues to achieve this.

8.
CNS Spectr ; 27(2): 157-190, 2022 04.
Article in English | MEDLINE | ID: mdl-33092669

ABSTRACT

BACKGROUND: Blood platelets, due to shared biochemical and functional properties with presynaptic serotonergic neurons, constituted, over the years, an attractive peripheral biomarker of neuronal activity. Therefore, the literature strongly focused on the investigation of eventual structural and functional platelet abnormalities in neuropsychiatric disorders, particularly in depressive disorder. Given their impact in biological psychiatry, the goal of the present paper was to review and critically analyze studies exploring platelet activity, functionality, and morpho-structure in subjects with depressive disorder. METHODS: According to the PRISMA guidelines, we performed a systematic review through the PubMed database up to March 2020 with the search terms: (1) platelets in depression [Title/Abstract]"; (2) "(platelets[Title]) AND depressive disorder[Title/Abstract]"; (3) "(Platelet[Title]) AND major depressive disorder[Title]"; (4) (platelets[Title]) AND depressed[Title]"; (5) (platelets[Title]) AND depressive episode[Title]"; (6) (platelets[Title]) AND major depression[Title]"; (7) platelet activation in depression[All fields]"; and (8) platelet reactivity in depression[All fields]." RESULTS: After a detailed screening analysis and the application of specific selection criteria, we included in our review a total of 106 for qualitative synthesis. The studies were classified into various subparagraphs according to platelet characteristics analyzed: serotonergic system (5-HT2A receptors, SERT activity, and 5-HT content), adrenergic system, MAO activity, biomarkers of activation, responsivity, morphological changes, and other molecular pathways. CONCLUSIONS: Despite the large amount of the literature examined, nonunivocal and, occasionally, conflicting results emerged. However, the findings on structural and metabolic alterations, modifications in the expression of specific proteins, changes in the aggregability, or in the responsivity to different pro-activating stimuli, may be suggestive of potential platelet dysfunctions in depressed subjects, which would result in a kind of hyperreactive state. This condition could potentially lead to an increased cardiovascular risk. In line with this hypothesis, we speculated that antidepressant treatments would seem to reduce this hyperreactivity while representing a potential tool for reducing cardiovascular risk in depressed patients and, maybe, in other neuropsychiatric conditions. However, the problem of the specificity of platelet biomarkers is still at issue and would deserve to be deepened in future studies.


Subject(s)
Blood Platelets , Depressive Disorder, Major , Antidepressive Agents/therapeutic use , Biomarkers , Blood Platelets/metabolism , Depressive Disorder, Major/drug therapy , Humans , Serotonin/metabolism
9.
Platelets ; 32(6): 761-769, 2021 Aug 18.
Article in English | MEDLINE | ID: mdl-33646086

ABSTRACT

While current oral antiplatelet therapies benefit many patients, they deregulate the hemostatic balance leaving patients at risk of systemic side-effects such as hemorrhage. Dual antiplatelet treatment is the standard approach, combining aspirin with P2Y12 blockers. These therapies mainly target autocrine activation mechanisms (TxA2, ADP) and, more recently, the use of thrombin or thrombin receptor antagonists have been added to the available approaches. Recent efforts to develop new classes of anti-platelet drugs have begun to focus on primary platelet activation pathways such as through the immunoreceptor tyrosine-based activation motif (ITAM)-containing collagen receptor GPVI/FcRγ-chain complex. There are already encouraging results from targeting GPVI, with reduced aggregation and smaller arterial thrombi, without major bleeding complications, likely due to overlapping activation signaling pathways with other receptors such as the GPIb-V-IX complex. An alternative approach to reduce platelet activation could be to inhibit this signaling pathway by targeting the inhibitory pathways intrinsic to platelets. Stimulation of endogenous negative modulators could provide more specific inhibition of platelet function, but is this feasible? In this review, we explore the potential of the two major platelet immunoreceptor tyrosine-based inhibitory motif (ITIM)-containing inhibitory receptors, G6b-B and PECAM-1, as antithrombotic targets.


Subject(s)
Blood Platelets/metabolism , Platelet Activation/genetics , Receptors, Immunologic/metabolism , Animals , Humans , Ligands , Mice , Mice, Knockout , Platelet Endothelial Cell Adhesion Molecule-1/metabolism
10.
Platelets ; 32(6): 753-760, 2021 Aug 18.
Article in English | MEDLINE | ID: mdl-33560928

ABSTRACT

Receptors are important pharmacological targets on cells. The Triggering Receptor Expressed on Myeloid Cells (TREM) - Like Transcript - 1 is an abundant, yet little understood, platelet receptor. It is a single Ig domain containing receptor isolated in the α-granules of resting platelets and brought to the platelet surface upon activation. On platelets, the integrin αIIbß3 is the major receptor having roughly 80,000 copies. αIIbß3 is a heterodimeric multidomain structure that mediates platelet aggregation through its interaction with the plasma protein fibrinogen. Anti-platelet drugs have successfully targeted αIIbß3 to control thrombosis. Like αIIbß3, TLT-1 also binds fibrinogen, making its role in platelet function somewhat obscure. In this review, we highlight the known structural features of TLT-1 and present the challenges of understanding TLT-1 function. In our analysis of the dynamics of the platelet surface after activation we propose a model in which TLT-1 supports αIIbß3 function as a mechanoreceptor that may direct platelets toward immune function.


Subject(s)
Blood Platelets/metabolism , Myeloid Cells/metabolism , Animals , Disease Models, Animal , Humans , Mice , Models, Molecular
11.
Biochimie ; 184: 1-7, 2021 May.
Article in English | MEDLINE | ID: mdl-33548391

ABSTRACT

Glycoprotein (GP)Ib that binds von Willebrand factor (vWF) and glycoprotein (GP)VI, that binds collagen play a significant role in platelet activation and aggregation, and are potential targets for antithrombotic treatment. They are targeted by snake venom proteinases. The effect of a such proteinase, mutalysin-II, on platelet aggregation was examined using washed human platelets and platelet-rich plasma. Its proteolytic activity on vWF, on its binding partner GPIbα, and on GPVI was analyzed by SDS-PAGE, and immunodetection with the corresponding antibodies after blotting. Dose- and time-dependently, mutalysin-II inhibits aggregation of washed platelets induced by vWF plus ristocetin and by convulxin, but with no significant effect on platelet-rich-plasma. Furthermore, mutalysin-II cleaves vWF into low molecular mass multimers of vWF and a rvWF-A1 domain to realease a ∼27-kDa fragment detectable by SDS-PAGE and blotting with mouse anti-rvWF-A1-domain IgG. Moreover, GPVI was cut by mutalysin-II into a soluble ∼55-kDa ectodomain and a fragment of ∼35-kDa. Thus, mutalysin-II inhibits vWF-induced platelet aggregation via cleavage of bound vWF-A1, and its receptor GPIbα. The additional cleavage of, GPVI, blocks collagen-induced platelets. Our data highlight mutalysin-II as an interesting platelet-directed tool targeting vWF-GPIbα binding and particularly GPVI. Thus, it might be suited for antithrombotic therapy as its combined inactivation of two receptors does not significantly compromise hemostasis, but shows high efficacy and safety. Studies are needed to further develop and demonstrate its potential benefits.


Subject(s)
Blood Platelets/chemistry , Metalloendopeptidases/chemistry , Platelet Aggregation Inhibitors/chemistry , Platelet Glycoprotein GPIb-IX Complex/chemistry , Platelet Membrane Glycoproteins/chemistry , Snake Venoms/chemistry , Animals , Blood Platelets/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Platelet Glycoprotein GPIb-IX Complex/metabolism , Platelet Membrane Glycoproteins/metabolism
12.
Int J Mol Sci ; 23(1)2021 Dec 29.
Article in English | MEDLINE | ID: mdl-35008781

ABSTRACT

In hemostasis and thrombosis, the complex process of thrombus formation involves different molecular pathways of platelet and coagulation activation. These pathways are considered as operating together at the same time, but this has not been investigated. The objective of our study was to elucidate the time-dependency of key pathways of thrombus and clot formation, initiated by collagen and tissue factor surfaces, where coagulation is triggered via the extrinsic route. Therefore, we adapted a microfluidics whole-blood assay with the Maastricht flow chamber to acutely block molecular pathways by pharmacological intervention at desired time points. Application of the technique revealed crucial roles of glycoprotein VI (GPVI)-induced platelet signaling via Syk kinase as well as factor VIIa-induced thrombin generation, which were confined to the first minutes of thrombus buildup. A novel anti-GPVI Fab EMF-1 was used for this purpose. In addition, platelet activation with the protease-activating receptors 1/4 (PAR1/4) and integrin αIIbß3 appeared to be prolongedly active and extended to later stages of thrombus and clot formation. This work thereby revealed a more persistent contribution of thrombin receptor-induced platelet activation than of collagen receptor-induced platelet activation to the thrombotic process.


Subject(s)
Blood Coagulation , Blood Platelets/metabolism , Collagen/pharmacology , Thromboplastin/pharmacology , Thrombosis/pathology , Blood Coagulation/drug effects , Blood Platelets/drug effects , Factor VIIa/metabolism , Fibrin/metabolism , Humans , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, Proteinase-Activated/metabolism , Signal Transduction , Syk Kinase/metabolism , Time Factors
13.
Trends Endocrinol Metab ; 30(8): 532-545, 2019 08.
Article in English | MEDLINE | ID: mdl-31196615

ABSTRACT

Type 2 diabetes mellitus (T2DM) is a multifactorial disease with a dysregulated circulating inflammatory molecule tendency. T2DM is closely associated with systemic inflammation, endothelial dysfunction, cardiovascular risk, and increased clotting susceptibility. Platelets have fundamental roles in the development and propagation of inflammation and cardiovascular risk. They signal through membrane receptors, resulting in (hyper)activation and release of inflammatory molecules from platelet compartments. This review highlights how circulating inflammatory molecules, acting as platelet receptor ligands, interact with platelets, causing platelets to be potent drivers of systemic inflammation. We conclude by suggesting that focused platelet research in T2DM is an important avenue to pursue to identify novel therapeutic targets, and that platelets could be used as cellular activity sensors themselves.


Subject(s)
Blood Platelets/metabolism , Diabetes Mellitus, Type 2/metabolism , Animals , Biomarkers/blood , Blood Platelets/physiology , Diabetes Mellitus, Type 2/blood , Humans , Signal Transduction/physiology
14.
Ter Arkh ; 91(2): 32-39, 2019 Mar 18.
Article in English | MEDLINE | ID: mdl-31094169

ABSTRACT

MATERIALS AND METHODS: 253 patients with chronic hepatitis C (CHC) and liver cirrhosis were included in the study. Assessment of gene polymorphisms of genes involved in inflammatory reactions and antiviral immunity (IL-1ß-511C/T, IL-10 -1082G/A, IL28B C/T, IL28B T/G, TNF-α -238G/A, TGF-ß -915G/C, IL-6 -174G/C), activators of local hepatic fibrosis (AGT G-6A, AGT 235 M/T, ATR1 1166 A/C), hemochromatosis (HFE C282Y, HFE H63D), platelet receptors (ITGA2 807 C/T, ITGB3 1565 T/C), coagulation proteins and endothelial dysfunction (FII 20210 G/A, FV 1691G/A, FVII 10976 G/A, FXIII 103 G/T, eNOS 894 G/T, CYBA 242 C/T, FBG -455 G/A, PAI-675 5G/4G, MTHFR 677 C/T) was carried. Using Bayesian networks we studied the predictor value of clinical and laboratory factors for the following conditions - end points (EP): development of cirrhosis (EP1), fibrosis rate (EP2), presence of portal hypertension (EP3) and cryoglobulins (EP4). RESULTS AND DISCUSSION: In addition to traditional factors we have shown the contribution of the following mutations. Predicting EP1- liver cirrhosis - HFE H63D, C282Y, CYBA 242 C/T, AGT G-6G, ITGB31565 T/C gene mutations were significant. We also found a link between the rate of progression of liver fibrosis and gene polymorphisms of AGT G-6G, AGT M235T, FV 1691G/A, ITGB31565 T/C. Among the genetic factors associated with portal hypertension there are gene polymorphisms of PAI-I-675 5G/4G, FII 20210 G/A, CYBA 242 C/T, HFE H63D and Il-6 174GC. Cryoglobulins and cryoglobuliemic vasculitis (EP4) are associated with gene mutations MTHFR C677T, ATR A1166C and HFE H63D. CONCLUSION: The results obtained allow to detect the major pathophysiological and genetic factors which determine the status of the patient and the outcome of the disease, to clarify their contribution, and to reveal the significance of point mutations of genes that control the main routes of HCV course and progression.


Subject(s)
Hepatitis C, Chronic/physiopathology , Liver Cirrhosis/physiopathology , Polymorphism, Genetic , Bayes Theorem , Hemochromatosis , Hepatitis C, Chronic/genetics , Humans , Interferons , Interleukins , Liver Cirrhosis/genetics , Mutation
15.
J Cardiothorac Surg ; 13(1): 131, 2018 Dec 22.
Article in English | MEDLINE | ID: mdl-30577843

ABSTRACT

BACKGROUND: As the incidence of cardiovascular diseases increases, the use of antiplatelet therapy is widely recognized. This presents clinicians with the challenge of balancing the risk of thrombotic and bleeding complications. Platelet dysfunction is one of the causes of postoperative bleedings and their etiology is not fully understood. Platelets receptors point-of-care investigation is of a remarkable value in assessing patients risk of bleeding. Reliable assessment of platelet function can improve treatment. The aim of this study was to evaluate the activity of platelet receptors in patients qualified for cardiac surgery, taking into account organ dysfunctions and pharmacological therapy applied in these patients. METHODS: Seventy-one cardiac surgical patients were analyzed before surgery using multiple electrode aggregometry with the use of the ADP test and ASPI test. The cut-off values were determined based on the manufacturer's recommendations. Patients were divided into four groups: Group I (33/71 patients, without platelet dysfunctions), Group II (6/71 patients, ADP < 710 AU x min), Group III (13/71 patients, ASPI < 570 AU x min) and Group IV (19 / 71 patients, ADP < 710 AU x min and ASPI < 570 AU x min). Biochemical data defining the efficiency of the liver and kidneys, the list of preoperative drugs used and the requirement for transfusion throughout the study group were collected. RESULTS: The study group included 41 males (57.7%) and 30 females (42.3%), mean age 66 years. The majority of patients (94.4%) had platelet counts within the normal range, but platelet function was impaired in more than half of the studied patients (53.5%). No relationship was found between the biochemical markers of the kidneys and liver and the function of the ADP and ASPI receptors, while receptors activities were related (rs = 0.72, p < 0.001), and both associated with platelet count (rs = 0.55, p < 0.001 and rs = 0.42, p < 0.001, respectively). Platelet receptors activity was not related to the postoperative need for any type of transfusion as well as the applied preoperative pharmacological therapy. CONCLUSIONS: Early identification of patients at high risk of bleeding, using point-of-care platelet function assessment tests, enables a targeted therapeutic pathway. Due to the variety of factors affecting the activity of platelets, finding a specific cause of this pathology is extremely difficult. According to our study, the correlation between platelet receptor disorders and mild to moderate liver and kidney injury has not been demonstrated. However, platelet receptors dysfunction has been shown to be associated with a decreased number of platelets.


Subject(s)
Blood Platelets/physiology , Cardiac Surgical Procedures , Postoperative Hemorrhage/etiology , Aged , Blood Platelets/metabolism , Blood Transfusion , Cardiac Surgical Procedures/adverse effects , Female , Humans , Kidney Function Tests , Liver Function Tests , Male , Platelet Aggregation Inhibitors/therapeutic use , Platelet Count , Platelet Function Tests , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Postoperative Hemorrhage/therapy , Preoperative Period , Purinergic P2Y Receptor Antagonists/therapeutic use , Risk Assessment/methods
16.
J Biochem Mol Toxicol ; : e22165, 2018 Jul 06.
Article in English | MEDLINE | ID: mdl-29979475

ABSTRACT

Cc3 -SPase (30 kDa-proteinase; pI 5.98) was isolated from Cerastes cerastes venom. Its sequence of 271 residues yielded from LC-MALDI-TOF showed high degrees of homology when aligned with other proteinases. Cc3 -SPase cleaved natural and synthetic proteins such as casein and fibrinogen leaving fibrin clots unaffected. Cc3 -SPase was fully abolished by ion chelators, whereas aprotinin, antithrombin III (Sigma Aldrich, Saint-Louis, Missouri, USA), and heparin were ineffective. Affinity of Cc3 -SPase to benzamidine indicated the presence of an aspartate residue in the catalytic site as confirmed by three-dimensional structure consisting of 14 ß-strands and four α-helices. Molecular mechanisms revealed that Cc3 -SPase is capable of promoting dysfunctional platelet aggregation via two signaling pathways mediated by the G-coupled protein receptors and αIIbß3 integrin. Cc3 -SPase is involved in both extrinsic/intrinsic coagulation pathways in deficient plasmas by replacing defective/lacking factors FII, FVII, and FVIII but not FX. Cc3 -SPase could substitute missing factors in blood diseases related to plasma factor deficiencies.

17.
Curr Pharm Des ; 24(5): 628-640, 2018.
Article in English | MEDLINE | ID: mdl-29237371

ABSTRACT

BACKGROUND: Platelet hyperactivity has been implicated in many cardiovascular (CV) events such as ischemic stroke, myocardial infarction and CV death. Genetic variability of platelet receptors has been shown to impact Src family kinases (SFKs) activation and in turn influence platelet activation. SFKs are important signal transmitters in platelets, interacting with several receptors as GPIIB/IIIa, GPIb, PEAR 1, GPIa, GPVI, PECAM and CD148. METHODS: In this review, we focused on genetic variants of platelet receptors whose signals are transmitted mainly by SFKs and may be associated with clinical manifestations of platelet hyperactivation like MI or IS. RESULTS: The genetic variants of platelet receptors, the signals of which are transmitted by SFKs, and the associated clinical manifestations in platelet hyperactivation, have been examined. The most extensively studied receptors were glycoprotein polymorphisms. The greatest numbers of genetic variants were analyzed in GPIb. GPIIb/IIIa receptor polymorphisms were also well analyzed and many studies highlighted their associations with ischemic stroke (IS) and myocardial infarction (MI). However, there are a number of conflicting studies finding that GPIIb/IIIa receptor polymorphisms may not influence platelet hyperactivity. Moreover, variability within some other receptors like GPVI, PECAM, PEAR1, and CD148 was analyzed only in single studies. CONCLUSIONS: Src family kinases are one of the most important signal transmitters in platelets. Some receptors have well documented interactions with SFKs, while other have not been examined in humans or data about its association originated from single studies. Further studies are necessary to confirm the findings and reduce falsepositive associations.


Subject(s)
Genetic Variation/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , src-Family Kinases/genetics , Humans , Platelet Activation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Polymorphism, Genetic/genetics , src-Family Kinases/metabolism
18.
Postepy Kardiol Interwencyjnej ; 13(3): 202-209, 2017.
Article in English | MEDLINE | ID: mdl-29056992

ABSTRACT

INTRODUCTION: Periprocedural bleeding related to coronary angiography (CAG) or percutaneous coronary intervention (PCI) is associated with worse prognosis. Determining genetic variations associated with increased bleeding risk may help to identify high-risk patients. AIM: To analyse the association between single nucleotide polymorphisms (SNPs) of crucial haemostatic platelet receptors (GPIa, GPVI, P2Y12) and the risk of periprocedural bleeding complications related to CAG/PCI. MATERIAL AND METHODS: The population consisted of 73 patients with ischaemic heart disease who developed bleeding complications within 30 days after CAG/PCI and 331 patients without bleeding. The frequency of SNPs of GPIa 807C/T, GPVI 13254T/C, P2Y12 32C/T, and P2Y12 H1/H2 haplotype was analysed using polymerase chain reaction (PCR) hybridization methods. RESULTS: The prevalence of variant alleles GPIa 807T, GPVI 13254C, P2Y12 34T, and P2Y12 H2 haplotype in the total study population was 56.7%, 20.3%, 56.2%, and 24.3%, respectively. The presence of variant alleles was not related to increased risk of periprocedural bleeding: GPIa 807C/T (OR = 1.29, 95% CI: 0.75-2.24, p = 0.334), GPVI 12354T/C (OR = 0.82, 95% CI: 0.40-1.64, p = 0.551), P2Y12 34C/T (OR = 0.71, 95% CI: 0.42-1.22, p = 0.189), P2Y12 H1/H2 haplotype (OR = 0.69, 95% CI: 0.35-1.36, p = 0.258). The frequency of the homozygous form of P2Y12 H2 haplotype was higher in the group of patients who developed bleeding (OR = 2.79, 95% CI: 0.51-13.77, p = 0.161). CONCLUSIONS: No significant association of the SNPs of GPIa 807C/T, GPVI 13254T/C, P2Y12 32C/T, and P2Y12 H1/H2 haplotype with increased risk of periprocedural bleeding was found in patients with ischaemic heart disease undergoing CAG/PCI.

19.
Expert Rev Proteomics ; 13(11): 993-1006, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27718750

ABSTRACT

INTRODUCTION: Unwanted platelet activation is associated with numerous diseases, mainly thrombosis-related. In this context, proteomics has emerged as a novel tool with potential for drug target discovery and to scrutinize the effects of antiplatelet drugs. Areas covered: The present review presents the main findings of platelet proteomic studies to date in the context of drug target discovery and perspectives for the future ahead. It includes data and evidences obtained from literature searches on PubMed as well as commentaries derived from the authors' experience and opinions. Expert commentary: Platelet proteomics applied to drug target discovery is a young field. Recent studies have shown promising data, especially in the context of coronary artery disease. However, challenges remain such as establishing definitive guidelines for blood collection and platelet isolation, essential to guarantee data reproducibility. Recent advances in quantitative platelet proteomics should lead to novel studies with higher clinical impact in the near future.

20.
Platelets ; 27(4): 322-32, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26559117

ABSTRACT

Anti-platelet agents play a central part in the treatment and prevention of acute thrombotic events. Discriminating animal models are needed for the development of novel agents. The chacma baboon has been extensively used as a model to evaluate anti-platelet agents. However, limited data exist to prove the translatability of this species to humans. We aimed to determine the suitability of the chacma baboon in preclinical human targeted GPIIb/IIIa, GPIbα and P2Y12 studies. Light-transmission platelet aggregometry (LTA), whole blood impedance aggregometry, receptor number quantification and genomic DNA sequencing were performed. Baboon ADP and arachidonic acid-induced LTA aggregation results differed significantly from human values, even at increased concentrations. LTA ristocetin-induced agglutination was comparable between species, but baboon platelets needed twice the concentration of ristocetin to elicit a similar response. Citrated baboon blood had significantly less aggregation than humans when evaluated with impedance aggregometry. However, hirudinised baboon whole blood gave similar aggregation as humans at the same agonist concentrations. GPIIb, GPIIIa and GPIbα numbers were significantly more on the baboon platelets. None of the amino acids deemed vital for receptor function, ligand binding or receptor inhibition, were radically different between the species. However, a conservative change in a calcium-binding region of GPIIb may render the baboon platelets more sensitive to calcium-binding agents. The chacma baboon may be used for the evaluation of human-targeted GPIIb/IIIa-, GPIbα- and P2Y12-inhibiting agents. However, the best anticoagulant, optimal agonist concentrations, increase in receptor number and sequence differences must be considered for any future studies.


Subject(s)
Blood Platelets/drug effects , Blood Platelets/metabolism , Platelet Aggregation Inhibitors/pharmacology , Platelet Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Adenosine Diphosphate/pharmacology , Amino Acid Sequence , Amino Acid Substitution , Animals , Arachidonic Acid/pharmacology , Drug Evaluation, Preclinical , Flow Cytometry , Humans , Male , Papio ursinus , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/chemistry , Platelet Membrane Glycoproteins/genetics , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Receptors, Purinergic P2/chemistry , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/metabolism , Ristocetin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...