Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.863
Filter
Add more filters

Publication year range
1.
Mol Cell Proteomics ; 23(2): 100717, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38237698

ABSTRACT

Platelet activation induces the secretion of proteins that promote platelet aggregation and inflammation. However, detailed analysis of the released platelet proteome is hampered by platelets' tendency to preactivate during their isolation and a lack of sensitive protocols for low abundance releasate analysis. Here, we detail the most sensitive analysis to date of the platelet releasate proteome with the detection of >1300 proteins. Unbiased scanning for posttranslational modifications within releasate proteins highlighted O-glycosylation as being a major component. For the first time, we detected O-fucosylation on previously uncharacterized sites including multimerin-1 (MMRN1), a major alpha granule protein that supports platelet adhesion to collagen and is a carrier for platelet factor V. The N-terminal elastin microfibril interface (EMI) domain of MMRN1, a key site for protein-protein interaction, was O-fucosylated at a conserved threonine within a new domain context. Our data suggest that either protein O-fucosyltransferase 1, or a novel protein O-fucosyltransferase, may be responsible for this modification. Mutating this O-fucose site on the EMI domain led to a >50% reduction of MMRN1 secretion, supporting a key role of EMI O-fucosylation in MMRN1 secretion. By comparing releasates from resting and thrombin-treated platelets, 202 proteins were found to be significantly released after high-dose thrombin stimulation. Complementary quantification of the platelet lysates identified >3800 proteins, which confirmed the platelet origin of releasate proteins by anticorrelation analysis. Low-dose thrombin treatment yielded a smaller subset of significantly regulated proteins with fewer secretory pathway enzymes. The extensive platelet proteome resource provided here (larancelab.com/platelet-proteome) allows identification of novel regulatory mechanisms for drug targeting to address platelet dysfunction and thrombosis.


Subject(s)
Proteome , Thrombin , Proteome/metabolism , Thrombin/pharmacology , Thrombin/metabolism , Glycosylation , Blood Platelets/metabolism , Platelet Activation
2.
Exp Cell Res ; 441(2): 114189, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39069151

ABSTRACT

Vascular smooth muscle cell (VSMCs) is one of the important cell types in artery. VSMCs stiffening may regulate vascular stiffness and contribute to the development of vulnerable plaques. Thrombin, an enzyme in coagulation system, is involved in pathological processes of atherosclerosis. Inter-alpha-trypsin inhibitor heavy chain 4 (ITIH4) plays an important role in regulating inflammation and may have cardiovascular protective effect. Therefore, the elucidation of the mechanisms underlying ITIH4-mediated VSMCs stiffening helps to provide new ideas and potential targets for the diagnosis and treatment of atherosclerosis. In this study, we used specific ITIH4 expression vector and siRNA methods to transfect VSMCs. Our results found that ITIH4 expression increased VSMCs stiffness, meanwhile, ITIH4 siRNA decreased VSMCs stiffness. ITIH4 increased acetylated α-tubulin and inhibited ERK1/2 and JNK, but not P38 MAPK. ERK inhibitor (PD98059) or JNK inhibitor (SP600125) treatment increased acetylated α-tubulin expression and cell stiffness in VSMCs. ITIH4 was downregulated by thrombin treatment, ITIH4 partly reversed the effect of thrombin on acetylated α-tubulin and VSMCs stiffness. These results indicated that ITIH4 regulated acetylated α-tubulin expression in VSMCs and was against the effects of thrombin on VSMCs stiffness. JNK and ERK signaling pathways were proved to participate in this process.


Subject(s)
MAP Kinase Signaling System , Muscle, Smooth, Vascular , Thrombin , Thrombin/pharmacology , Thrombin/metabolism , MAP Kinase Signaling System/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Animals , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Vascular Stiffness/drug effects , Cells, Cultured , Rats , Humans , Rats, Sprague-Dawley , Peptide Hormones/metabolism , Peptide Hormones/pharmacology , Peptide Hormones/genetics
3.
Mol Cell ; 65(6): 1014-1028.e7, 2017 Mar 16.
Article in English | MEDLINE | ID: mdl-28262504

ABSTRACT

Ca2+ dynamics and oxidative signaling are fundamental mechanisms for mitochondrial bioenergetics and cell function. The MCU complex is the major pathway by which these signals are integrated in mitochondria. Whether and how these coactive elements interact with MCU have not been established. As an approach toward understanding the regulation of MCU channel by oxidative milieu, we adapted inflammatory and hypoxia models. We identified the conserved cysteine 97 (Cys-97) to be the only reactive thiol in human MCU that undergoes S-glutathionylation. Furthermore, biochemical, structural, and superresolution imaging analysis revealed that MCU oxidation promotes MCU higher order oligomer formation. Both oxidation and mutation of MCU Cys-97 exhibited persistent MCU channel activity with higher [Ca2+]m uptake rate, elevated mROS, and enhanced [Ca2+]m overload-induced cell death. In contrast, these effects were largely independent of MCU interaction with its regulators. These findings reveal a distinct functional role for Cys-97 in ROS sensing and regulation of MCU activity.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling , Calcium/metabolism , Endothelial Cells/metabolism , Ion Channel Gating , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Reactive Oxygen Species/metabolism , Animals , COS Cells , Calcium Channels/chemistry , Calcium Channels/genetics , Calcium Signaling/drug effects , Cell Death , Cell Hypoxia , Chlorocebus aethiops , Cysteine , Endothelial Cells/drug effects , Endothelial Cells/pathology , Energy Metabolism , Glutathione/metabolism , HEK293 Cells , HeLa Cells , Humans , Ion Channel Gating/drug effects , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/pathology , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/pathology , Mutation , Oxidation-Reduction , Protein Multimerization , Protein Processing, Post-Translational , Protein Structure, Quaternary , Structure-Activity Relationship , Thrombin/pharmacology , Time Factors , Transfection
4.
Proteomics ; 24(16): e2400090, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39148210

ABSTRACT

Arterial thrombosis contributes to some of the most frequent causes of mortality globally, such as myocardial infarction and stroke. Platelets are essential mediators of physiological haemostasis and pathological thrombosis. Platelet activation is controlled by a multitude of signalling pathways. Upon activation, platelets shed platelet-derived extracellular vesicles (pEVs). In this Special Issue: Extracellular Vesicles, Moon et al. investigate the impact of various platelet agonists (thrombin, ADP, collagen) on the proteome of pEVs. The study demonstrates that pEVs exhibit an agonist-dependent altered proteome compared to their parent cells, with significant variations in proteins related to coagulation, complement, and platelet activation. The study observes the rapid generation of pEVs following agonist stimulation with specific proteome alterations that underscore an active packaging process. This commentary highlights the implications of their findings and discusses the role of pEV cargo in cardiovascular disease with potential novel therapeutic and diagnostic opportunities.


Subject(s)
Blood Platelets , Extracellular Vesicles , Platelet Activation , Proteome , Humans , Extracellular Vesicles/metabolism , Extracellular Vesicles/drug effects , Blood Platelets/metabolism , Blood Platelets/drug effects , Proteome/metabolism , Platelet Activation/drug effects , Proteomics/methods , Thrombin/metabolism , Thrombin/pharmacology
5.
Proteomics ; 24(11): e2300391, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38556629

ABSTRACT

Arterial thrombosis manifesting as heart attack and stroke is the leading cause of death worldwide. Platelets are central mediators of thrombosis that can be activated through multiple activation pathways. Platelet-derived extracellular vesicles (pEVs), also known as platelet-derived microparticles, are granular mixtures of membrane structures produced by platelets in response to various activating stimuli. Initial studies have attracted interest on how platelet agonists influence the composition of the pEV proteome. In the current study, we used physiological platelet agonists of varying potencies which reflect the microenvironments that platelets experience during thrombus formation: adenosine diphosphate, collagen, thrombin as well as a combination of thrombin/collagen to induce platelet activation and pEV generation. Proteomic profiling revealed that pEVs have an agonist-dependent altered proteome in comparison to their cells of origin, activated platelets. Furthermore, we found that various protein classes including those related to coagulation and complement (prothrombin, antithrombin, and plasminogen) and platelet activation (fibrinogen) are attributed to platelet EVs following agonist stimulation. This agonist-dependent altered proteome suggests that protein packaging is an active process that appears to occur without de novo protein synthesis. This study provides new information on the influence of physiological agonist stimuli on the biogenesis and proteome landscape of pEVs.


Subject(s)
Blood Platelets , Extracellular Vesicles , Platelet Activation , Proteome , Proteomics , Thrombin , Blood Platelets/metabolism , Blood Platelets/drug effects , Humans , Proteome/metabolism , Extracellular Vesicles/metabolism , Extracellular Vesicles/drug effects , Platelet Activation/drug effects , Thrombin/pharmacology , Thrombin/metabolism , Proteomics/methods , Adenosine Diphosphate/pharmacology , Adenosine Diphosphate/metabolism , Collagen/metabolism
6.
J Biol Chem ; 299(12): 105370, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37865315

ABSTRACT

G protein-coupled receptors (GPCRs) are highly druggable and implicated in numerous diseases, including vascular inflammation. GPCR signals are transduced from the plasma membrane as well as from endosomes and controlled by posttranslational modifications. The thrombin-activated GPCR protease-activated receptor-1 is modified by ubiquitin. Ubiquitination of protease-activated receptor-1 drives recruitment of transforming growth factor-ß-activated kinase-1-binding protein 2 (TAB2) and coassociation of TAB1 on endosomes, which triggers p38 mitogen-activated protein kinase-dependent inflammatory responses in endothelial cells. Other endothelial GPCRs also induce p38 activation via a noncanonical TAB1-TAB2-dependent pathway. However, the regulatory processes that control GPCR ubiquitin-driven p38 inflammatory signaling remains poorly understood. We discovered mechanisms that turn on GPCR ubiquitin-dependent p38 signaling, however, the mechanisms that turn off the pathway are not known. We hypothesize that deubiquitination is an important step in regulating ubiquitin-driven p38 signaling. To identify specific deubiquitinating enzymes (DUBs) that control GPCR-p38 mitogen-activated protein kinase signaling, we conducted a siRNA library screen targeting 96 DUBs in endothelial cells and HeLa cells. We identified nine DUBs and validated the function two DUBs including cylindromatosis and ubiquitin-specific protease-34 that specifically regulate thrombin-induced p38 phosphorylation. Depletion of cylindromatosis expression by siRNA enhanced thrombin-stimulated p38 signaling, endothelial barrier permeability, and increased interleukin-6 cytokine expression. Conversely, siRNA knockdown of ubiquitin-specific protease-34 expression decreased thrombin-promoted interleukin-6 expression and had no effect on thrombin-induced endothelial barrier permeability. These studies suggest that specific DUBs distinctly regulate GPCR-induced p38-mediated inflammatory responses.


Subject(s)
Deubiquitinating Enzyme CYLD , Deubiquitinating Enzymes , Endothelial Cells , Thrombin , Humans , Adaptor Proteins, Signal Transducing/metabolism , Deubiquitinating Enzyme CYLD/metabolism , Deubiquitinating Enzymes/metabolism , Endothelial Cells/metabolism , HeLa Cells , Interleukin-6/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , Receptor, PAR-1/metabolism , RNA, Small Interfering/metabolism , Thrombin/pharmacology , Thrombin/metabolism , Ubiquitin/metabolism , Ubiquitin-Specific Proteases/metabolism , Cell Line , Gene Expression Regulation, Enzymologic , Phosphorylation/genetics
7.
Am J Physiol Renal Physiol ; 326(2): F219-F226, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38031732

ABSTRACT

Protease-activated receptor 4 (PAR4) is a G protein-coupled receptor activated by thrombin. In the platelet, response to thrombin PAR4 contributes to the predominant procoagulant microparticle formation, increased fibrin deposition, and initiation of platelet-stimulated inflammation. In addition, PAR4 is expressed in other cell types, including endothelial cells. Under inflammatory conditions, PAR4 is overexpressed via epigenetic demethylation of the PAR4 gene, F2RL3. PAR4 knockout (KO) studies have determined a role for PAR4 in ischemia-reperfusion injury in the brain, and PAR4 KO mice display normal cardiac function but present less myocyte death and cardiac dysfunction in response to acute myocardial infarction. Although PAR4 has been reported to be expressed within the kidney, the contribution of PAR4 to acute kidney injury (AKI) and chronic kidney disease (CKD) is not well understood. Here we report that PAR4 KO mice are protected against kidney injury in two mouse models. First, PAR4 KO mice are protected against induction of markers of both fibrosis and inflammation in two different models of kidney injury: 1) 7 days following unilateral ureter obstruction (UUO) and 2) an AKI-CKD model of ischemia-reperfusion followed by 8 days of contralateral nephrectomy. We further show that PAR4 expression in the kidney is low in the control mouse kidney but induced over time following UUO. PAR4 KO mice are protected against blood urea nitrogen (BUN) and glomerular filtration rate (GFR) kidney function pathologies in the AKI-CKD model. Following the AKI-CKD model, PAR4 is expressed in the collecting duct colocalizing with Dolichos biflorus agglutinin (DBA), but not in the proximal tubule with Lotus tetragonolobus lectin (LTL). Collectively, the results reported in this study implicate PAR4 as contributing to the pathology in mouse models of acute and chronic kidney injury.NEW & NOTEWORTHY The contribution of the thrombin receptor protease-activated receptor 4 (PAR4) to acute kidney injury (AKI) and chronic kidney disease (CKD) is not well understood. Here we report that PAR4 expression is upregulated after kidney injury and PAR4 knockout (KO) mice are protected against fibrosis following kidney injury in two mouse models. First, PAR4 KO mice are protected against unilateral ureter obstruction. Second, PAR4 KO mice are protected against an AKI-CKD model of ischemia-reperfusion followed by contralateral nephrectomy.


Subject(s)
Acute Kidney Injury , Renal Insufficiency, Chronic , Animals , Mice , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Endothelial Cells/metabolism , Fibrosis , Inflammation/pathology , Ischemia/pathology , Kidney/metabolism , Mice, Knockout , Receptors, Thrombin/genetics , Receptors, Thrombin/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Reperfusion Injury/pathology , Thrombin/metabolism , Thrombin/pharmacology
8.
Am J Physiol Lung Cell Mol Physiol ; 326(3): L213-L225, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38113296

ABSTRACT

Neonates with congenital diaphragmatic hernia (CDH) frequently require cardiopulmonary bypass and systemic anticoagulation. We previously demonstrated that even subtherapeutic heparin impairs lung growth and function in a murine model of compensatory lung growth (CLG). The direct thrombin inhibitors (DTIs) bivalirudin and argatroban preserved growth in this model. Although DTIs are increasingly used for systemic anticoagulation clinically, patients with CDH may still receive heparin. In this experiment, lung endothelial cell proliferation was assessed following treatment with heparin-alone or mixed with increasing concentrations of bivalirudin or argatroban. The effects of subtherapeutic heparin with or without DTIs in the CLG model were also investigated. C57BL/6J mice underwent left pneumonectomy and subcutaneous implantation of osmotic pumps. Pumps were preloaded with normal saline, bivalirudin, or argatroban; treated animals received daily intraperitoneal low-dose heparin. In vitro, heparin-alone decreased endothelial cell proliferation and increased apoptosis. The effect of heparin on proliferation, but not apoptosis, was reversed by the addition of bivalirudin and argatroban. In vivo, low-dose heparin decreased lung volume compared with saline-treated controls. All three groups that received heparin demonstrated decreased lung function on pulmonary function testing and impaired exercise performance on treadmill tolerance testing. These findings correlated with decreases in alveolarization, vascularization, angiogenic signaling, and gene expression in the heparin-exposed groups. Together, these data suggest that bivalirudin and argatroban fail to reverse the inhibitory effects of subtherapeutic heparin on lung growth and function. Clinical studies on the impact of low-dose heparin with DTIs on CDH outcomes are warranted.NEW & NOTEWORTHY Infants with pulmonary hypoplasia frequently require cardiopulmonary bypass and systemic anticoagulation. We investigate the effects of simultaneous exposure to heparin and direct thrombin inhibitors (DTIs) on lung growth and pulmonary function in a murine model of compensatory lung growth (CGL). Our data suggest that DTIs fail to reverse the inhibitory effects of subtherapeutic heparin on lung growth and function. Clinical studies on the impact of heparin with DTIs on clinical outcomes are thus warranted.


Subject(s)
Antithrombins , Arginine/analogs & derivatives , Heparin , Pipecolic Acids , Sulfonamides , Humans , Animals , Mice , Heparin/pharmacology , Heparin/therapeutic use , Antithrombins/pharmacology , Antithrombins/therapeutic use , Anticoagulants/therapeutic use , Pneumonectomy , Disease Models, Animal , Mice, Inbred C57BL , Hirudins/pharmacology , Fibrinolytic Agents , Lung/metabolism , Peptide Fragments/pharmacology , Recombinant Proteins/pharmacology , Thrombin/pharmacology , Thrombin/metabolism
9.
J Cell Sci ; 135(7)2022 04 01.
Article in English | MEDLINE | ID: mdl-35274124

ABSTRACT

Cancer cells exploit a variety of migration modes to leave primary tumors and establish metastases, including amoeboid cell migration, which is typically reliant on bleb formation. Here we demonstrate that thrombin induces dynamic blebbing in the MDA-MB-231 breast cancer cell line and confirm that protease-activated receptor 1 (PAR1) activation is sufficient to induce this effect. Cell confinement has been implicated as a driving force in bleb-based migration. Unexpectedly, we found that gentle contact compression, exerted using a custom built 'cell press' to mechanically stimulate cells, reduced thrombin-induced blebbing. Thrombin-induced blebbing was similarly attenuated using the small molecule Yoda1, an agonist of the mechanosensitive Ca2+ channel Piezo1, and this attenuation was impaired in Piezo1-depleted cells. Additionally, Piezo1 activation suppressed thrombin-induced phosphorylation of ezrin, radixin and moesin (ERM) proteins, which are implicated in the blebbing process. Our results provide mechanistic insights into Piezo1 activation as a suppressor of dynamic blebbing, specifically that which is induced by thrombin.


Subject(s)
Breast Neoplasms , Ion Channels , Cell Movement , Female , Humans , Ion Channels/metabolism , Phosphorylation , Thrombin/metabolism , Thrombin/pharmacology
10.
Microvasc Res ; 151: 104613, 2024 01.
Article in English | MEDLINE | ID: mdl-37793562

ABSTRACT

Thrombosis can lead to significant mortality and morbidity. Both platelets and vascular endothelial cells play significant roles in thrombosis. Platelets' response to blood flow-induced shear stress can vary greatly depending on shear stress magnitude, pattern and shear exposure time. Endothelial cells are also sensitive to the biomechanical environment. Endothelial cell activation and dysfunction can occur under low oscillatory shear stress and low tensile strain. Platelet and endothelial cell interaction can also be affected by mechanical conditions. The goal of this study was to investigate how blood flow-induced shear stress, vascular wall tensile strain, platelet-endothelial cell stress history, and platelet-endothelial cell interaction affect platelet thrombogenicity. Platelets and human coronary artery endothelial cells were pretreated with physiological and pathological shear stress and/or tensile strain separately. The pretreated cells were then put together and exposed to pulsatile shear stress and cyclic tensile strain simultaneously in a shearing-stretching device. Following treatment, platelet thrombin generation rate, platelet and endothelial cell activation, and platelet adhesion to endothelial cells was measured. The results demonstrated that shear stress pretreatment of endothelial cells and platelets caused a significant increase in platelet thrombin generation rate, cell surface phosphatidylserine expression, and adhesion to endothelial cells. Shear stress pretreatment of platelets and endothelial cells attenuated endothelial cell ICAM-1 expression under stenosis conditions, as well as vWF expression under recirculation conditions. These results indicate that platelets are sensitized by prior shearing, while in comparison, the interaction with shear stress-pretreated platelets may reduce endothelial cell sensitivity to pathological shear stress and tensile strain.


Subject(s)
Endothelial Cells , Thrombosis , Humans , Endothelial Cells/metabolism , Thrombin/metabolism , Thrombin/pharmacology , Blood Platelets/metabolism , Platelet Adhesiveness , Thrombosis/etiology , Stress, Mechanical , Platelet Activation
11.
Brain Behav Immun ; 116: 85-100, 2024 02.
Article in English | MEDLINE | ID: mdl-38042209

ABSTRACT

Acute spinal cord injury (SCI) always results in sustainable recruitment of inflammatory cells driven by sequentially generated chemokines, thereby eliciting excessive neuroinflammation. However, the underlying mechanism of temporally produced chemokines remains elusive. Reactive astrocytes are known to be the main sources of chemokines at the lesion site, which can be immediately activated by thrombin following SCI. In the present study, SCI was shown to induce a sequential production of chemokines CCL2 and CCL5 from astrocytes, which were associated with a persistent infiltration of macrophages/microglia. The rapidly induced CCL2 and later induced CCL5 from astrocytes were regulated by thrombin at the damaged tissues. Investigation of the regulatory mechanism revealed that thrombin facilitated astrocytic CCL2 production through activation of ERK/JNK/NFκB pathway, whereas promoted CCL5 production through PLCß3/NFκB and ERK/JNK/NFκB signal pathway. Inhibition of thrombin activity significantly decreased production of astrocytic CCL2 and CCL5, and reduced the accumulation of macrophages/microglia at the lesion site. Accordingly, the locomotor function of rats was remarkably improved. The present study has provided a new regulatory mechanism on thrombin-mediated sequential production of astrocytic chemokines, which might be beneficial for clinical therapy of CNS neuroinflammation.


Subject(s)
Astrocytes , Spinal Cord Injuries , Rats , Animals , Astrocytes/metabolism , Thrombin/pharmacology , Neuroinflammatory Diseases , Chemokines/metabolism , Spinal Cord/metabolism
12.
Neurochem Res ; 49(3): 597-616, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37978153

ABSTRACT

Neuroinflammation is assumed as the critical pathophysiologic mechanism of white matter lesions (WMLs), and infiltrated peripheral monocyte-derived macrophages are implicated in the development of neuroinflammation. This study sought to explore the blood molecules that promote the migration of peripheral monocytes to the sites of WMLs. The serum protein expression profiles of patients and Sprague-Dawley rat models with WMLs were detected by data-independent acquisition (DIA) proteomics technique. Compared with corresponding control groups, we acquired 62 and 41 differentially expressed proteins (DEPs) in the serum of patients and model rats with WMLs respectively. Bioinformatics investigations demonstrated that these DEPs were linked to various Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and Gene Ontology (GO) terms involved in neuroinflammation. Afterward, we identified thrombin-activatable fibrinolysis inhibitor (TAFI) as a shared and overexpressed protein in clinical and animal serum samples, which was further verified by enzyme-linked immunosorbent assay. Additionally, an upregulation of TAFI was also observed in the white matter of rat models, and the inhibition of TAFI impeded the migration of peripheral monocytes to the area of WMLs. In vitro experiments suggested that TAFI could enhance the migration ability of RAW264.7 cells and increase the expression of Ccr2. Our study demonstrates that neuroinflammatory signals can be detected in the peripheral blood of WMLs patients and model rats. TAFI may serve as a potential protein that promotes the migration of peripheral monocytes to WMLs regions, thereby providing a novel molecular target for further investigation into the interaction between the central and peripheral immune systems.


Subject(s)
Carboxypeptidase B2 , White Matter , Humans , Rats , Animals , Fibrinolysis/physiology , Carboxypeptidase B2/genetics , Carboxypeptidase B2/metabolism , Neuroinflammatory Diseases , Monocytes/metabolism , Proteomics , White Matter/metabolism , Rats, Sprague-Dawley , Thrombin/metabolism , Thrombin/pharmacology
13.
Arterioscler Thromb Vasc Biol ; 43(8): 1441-1454, 2023 08.
Article in English | MEDLINE | ID: mdl-37317855

ABSTRACT

BACKGROUND: Endothelial CLICs (chloride intracellular channel proteins) CLIC1 and CLIC4 are required for the GPCRs (G-protein-coupled receptors) S1PR1 (sphingosine-1-phosphate receptor 1) and S1PR3 to activate the small GTPases Rac1 (Ras-related C3 botulinum toxin substrate 1) and RhoA (Ras homolog family member A). To determine whether CLIC1 and CLIC4 function in additional endothelial GPCR pathways, we evaluated CLIC function in thrombin signaling via the thrombin-regulated PAR1 (protease-activated receptor 1) and downstream effector RhoA. METHODS: We assessed the ability of CLIC1 and CLIC4 to relocalize to cell membranes in response to thrombin in human umbilical vein endothelial cells (HUVEC). We examined CLIC1 and CLIC4 function in HUVEC by knocking down expression of each CLIC protein and compared thrombin-mediated RhoA or Rac1 activation, ERM (ezrin/radixin/moesin) phosphorylation, and endothelial barrier modulation in control and CLIC knockdown HUVEC. We generated a conditional murine allele of Clic4 and examined PAR1-mediated lung microvascular permeability and retinal angiogenesis in mice with endothelial-specific loss of Clic4. RESULTS: Thrombin promoted relocalization of CLIC4, but not CLIC1, to HUVEC membranes. Knockdown of CLIC4 in HUVEC reduced thrombin-mediated RhoA activation, ERM phosphorylation, and endothelial barrier disruption. Knockdown of CLIC1 did not reduce thrombin-mediated RhoA activity but prolonged the RhoA and endothelial barrier response to thrombin. Endothelial-specific deletion of Clic4 in mice reduced lung edema and microvascular permeability induced by PAR1 activating peptide. CONCLUSIONS: CLIC4 is a critical effector of endothelial PAR1 signaling and is required to regulate RhoA-mediated endothelial barrier disruption in cultured endothelial cells and murine lung endothelium. CLIC1 was not critical for thrombin-mediated barrier disruption but contributed to the barrier recovery phase after thrombin treatment.


Subject(s)
Receptor, PAR-1 , rhoA GTP-Binding Protein , Humans , Mice , Animals , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism , rhoA GTP-Binding Protein/metabolism , Thrombin/pharmacology , Thrombin/metabolism , Endothelium/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Cells, Cultured , Chloride Channels/genetics , Chloride Channels/metabolism , Mitochondrial Proteins/metabolism
14.
Int Wound J ; 21(4): e14622, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38158884

ABSTRACT

This study aims to evaluate the clinical effects of different blood derivatives on wound healing using network meta-analysis. PubMed, Embase, OVID, Web of Science, SCOPUS and Cochrane Central were searched to obtain studies about blood derivatives on wound healing until October 2023. R 4.2.0 and Stata 15.0 softwares were used for data analysis. Forty-four studies comprising 5164 patients were included. The results of network meta-analysis showed that the healing area from high to low was GF + ORCCB, ORCCB, GF, PRF, Unnas paste dressing, APG, PRP injection, PRP, PRP + thrombin gel, PPP, HPL, CT. The healing time from low to high was PRP + thrombin gel, GF, PRP, PC + K, PC, APG, PRF, CT, Silver sulfadiazine ointment. The number of patients cured from high to low was APG, PRP injection, PRP, Aurix, PRF, Leucopatch, HPL, Antimicrobial Ointment Dressing, CT, 60 µg/cm2 repifermin, 120 µg/cm2 repifermin, AFG, PPP. The order of analgesic effect from high to low was AFG, Aminogam gel, PRF, PRP, Oxidised oil, APG, GF, CT. The order of the number of wound infection cases from low to high is APG, 20 µg/cm2 repifermin, 60 µg/cm2 repifermin, PRP, LeucoPatch, CT, PPP, Antiseptic ointment dressing. Healing area: GF + ORCCB had the best effect; Healing time: PRP + thrombin gel took the shortest time. The number of cured patients and the reduction of wound infection: APG has the best effect. Analgesic effect: AFG has the best effect. More studies with large sample sizes are needed to confirm the above findings.


Subject(s)
Platelet-Rich Plasma , Wound Infection , Humans , Network Meta-Analysis , Thrombin/pharmacology , Ointments , Fibroblast Growth Factor 10/pharmacology , Wound Healing , Treatment Outcome , Analgesics
15.
Infect Immun ; 91(3): e0055622, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36853027

ABSTRACT

Platelets are known for essential activities in hemostasis and for their important contribution to protection against infectious pathogens. Klebsiella pneumoniae is an opportunistic pathogen widely known to cause nosocomial infections. Recently, hypervirulent strains of K. pneumoniae have been emerging, which can cause severe infections in immunocompetent individuals. Combined with the increase in antibiotic resistance, it is important to understand how K. pneumoniae affects components of the immune system. We studied the interactions of human platelets with several K. pneumoniae strains (the wild type encapsulated strain, and a nonencapsulated mutant). Thrombin-stimulated whole human and mouse blood significantly inhibited bacterial growth compared to unstimulated whole blood. Furthermore, we investigated the effect of K. pneumoniae on platelet activation. Both strains induced significant increase in activation of both unstimulated and thrombin-stimulated human platelets. Additionally, only the nonencapsulated mutant increased aggregation of platelets in response to ADP. K. pneumoniae killing assays were then performed with washed platelets in the presence or absence of thrombin. Surprisingly, washed platelets failed to exhibit any effects on the growth of K. pneumoniae. We further explored the impact of platelets on monocyte-mediated killing of K. pneumoniae. Importantly, we found that activated platelets significantly enhanced monocyte-mediated killing of K. pneumoniae. This effect was likely due to the formation of platelet-monocyte aggregates in blood upon thrombin stimulation. Overall, this study highlights the role of platelets in mediating a protective response against K. pneumoniae and reinforces the importance of platelets in modulating leukocyte behavior.


Subject(s)
Blood Platelets , Klebsiella Infections , Animals , Mice , Humans , Klebsiella pneumoniae , Monocytes , Thrombin/pharmacology , Platelet Activation , Klebsiella Infections/microbiology , Anti-Bacterial Agents
16.
Cell Mol Biol (Noisy-le-grand) ; 69(4): 46-52, 2023 Apr 30.
Article in English | MEDLINE | ID: mdl-37329549

ABSTRACT

Hyperglycemia, insulin resistance, and endothelium dysfunction are related to platelet hyperactivity in type 2 diabetes mellitus (T2D) patients. Glucosamine (GlcN) has inhibitory effects on platelets of animals and healthy donors, but this role in platelets from T2D patients is unknown. The aim of this study was to evaluate the GlcN in vitro effects on platelet aggregation in T2D patients and healthy donors. Donors´ and T2D patients' samples were analyzed through flow cytometry, Western blot, and platelet aggregometry. Platelet aggregation was induced using ADP and thrombin, with or without GlcN, N-Acetyl-glucosamine, galactose, or fucose. GlcN inhibited ADP and thrombin-induced platelet aggregation, while the other carbohydrates did not. GlcN suppressed the second wave of ADP-induced platelet aggregation. No differences in the percent of inhibition of ADP-induced platelet aggregation by GlcN were found between donors and T2D patients, but this effect was significantly higher in healthy donors using thrombin as an agonist. In addition, GlcN increased protein O-GlcNAcylation (O-GlcNAc) in the platelets from T2D patients but not in healthy donors. In conclusion, GlcN inhibited the platelet aggregation induced by ADP and thrombin for both study groups and increased O-GlcNAc in platelets from T2D patients. Further studies are required to evaluate the possible use of GlcN as an antiplatelet agent.


Subject(s)
Diabetes Mellitus, Type 2 , Platelet Aggregation , Animals , Glucosamine/pharmacology , Glucosamine/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Thrombin/metabolism , Thrombin/pharmacology , Blood Platelets/metabolism , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/therapeutic use , Platelet Aggregation Inhibitors/metabolism
17.
J Biochem Mol Toxicol ; 37(11): e23476, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37466159

ABSTRACT

Daboxin P, reported earlier from the venom of Daboia russellii, disturbs the blood coagulation cascade by targeting factor X and factor Xa. The present study exhibits that Daboxin P also inhibits platelet aggregation induced by various agonists. The thrombin-induced platelet aggregation was inhibited maximum whereas inhibition of collagen-induced platelet aggregation was found to be 50% and no inhibition of adenosine diphosphate (ADP) and arachidonic acid-induced aggregation was observed. Daboxin P dose-dependently inhibited the thrombin-induced platelet aggregation with Anti-Aggregation 50 (AD50 ) dose of 55.166 nM and also reduced the thrombin-mediated calcium influx. In-silico interaction studies suggested that Daboxin P binds to thrombin and blocks its interaction with its receptor on the platelet surface. Quenching of thrombin's emission spectrum by Daboxin P and electrophoretic profiles of pull-down assay further reveals the binding between Daboxin P and thrombin. Thus, the present study demonstrates that Daboxin P inhibits thrombin-induced platelet aggregation by binding to thrombin.


Subject(s)
Platelet Aggregation , Thrombin , Thrombin/pharmacology , Phospholipases A2/pharmacology , Blood Coagulation , Blood Platelets , Viper Venoms/pharmacology
18.
BMC Cardiovasc Disord ; 23(1): 191, 2023 04 12.
Article in English | MEDLINE | ID: mdl-37046189

ABSTRACT

We explored the effect of thrombin on human aortic smooth muscle cells (HASMCs) and further analyzed its role in the pathogenesis of atherosclerosis (AS). Thrombin-induced differentially expressed genes (DEGs) in HASMCs were identified by analyzing expression profiles from the GEO. Subsequently, enrichment analysis, GSEA, PPI network, and gene-microRNAs networks were interrogated to identify hub genes and associated pathways. Enrichment analysis results indicated that thrombin causes HASMCs to secrete various pro-inflammatory cytokines and chemokines, exacerbating local inflammatory response in AS. Moreover, we identified 9 HUB genes in the PPI network, which are closely related to the inflammatory response and the promotion of the cell cycle. Additionally, we found that thrombin inhibits lipid metabolism and autophagy of HASMCs, potentially contributing to smooth muscle-derived foam cell formation. Our study deepens a mechanistic understanding of the effect of thrombin on HASMCs and provides new insight into treating AS.


Subject(s)
Atherosclerosis , Transcriptome , Humans , Thrombin/genetics , Thrombin/metabolism , Thrombin/pharmacology , Atherosclerosis/pathology , Myocytes, Smooth Muscle/metabolism , Muscle, Smooth/metabolism , Muscle, Smooth/pathology
19.
BMC Cardiovasc Disord ; 23(1): 97, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36809978

ABSTRACT

BACKGROUND AND OBJECTIVE: Protease-activated receptor 1 (PAR1) is crucial in individuals with acute myocardial infarction (AMI). The continuous and prompt PAR1 activation mainly dependent on PAR1 trafficking is essential for the role of PAR1 during AMI in which cardiomyocytes are in hypoxia. However, the PAR1 trafficking in cardiomyocytes specially during the hypoxia is still unclear. METHODS AND RESULT: A rat AMI model was created. PAR1 activation with thrombin-receptor activated peptide (TRAP) had a transient effect on cardiac function in normal rats but persistent improvement in rats with AMI. Cardiomyocytes from neonatal rats were cultured in a normal CO2 incubator and a hypoxic modular incubator chamber. The cells were then subjected to western blot for the total protein expression and staining with fluorescent reagent and antibody for PAR1 localization. No change in total PAR1 expression following TRAP stimulation was observed; however, it led to increased PAR1 expression in the early endosomes in normoxic cells and decreased expression in the early endosomes in hypoxic cells. Under hypoxic conditions, TRAP restored the PAR1 expression on both cell and endosomal surfaces within an hour by decreasing Rab11A (8.5-fold; 179.93 ± 9.82% of the normoxic control group, n = 5) and increasing Rab11B (15.5-fold) expression after 4 h of hypoxia. Similarly, Rab11A knockdown upregulated PAR1 expression under normoxia, and Rab11B knockdown downregulated PAR1 expression under both normoxic and hypoxic conditions. Cardiomyocytes knocked out of both Rab11A, and Rad11B lost the TRAP-induced PAR1 expression but still exhibited the early endosomal TRAP-induced PAR1 expression under hypoxia. CONCLUSIONS: TRAP-mediated activation of PAR1 in cardiomyocytes did not alter the total PAR1 expression under normoxic conditions. Instead, it triggers a redistribution of PAR1 levels under normoxic and hypoxic conditions. TRAP reverses the hypoxia-inhibited PAR1 expression in cardiomyocytes by downregulating Rab11A expression and upregulating Rab11B expression.


Subject(s)
Myocardial Infarction , Receptor, PAR-1 , Animals , Rats , Hypoxia/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Peptides/metabolism , Peptides/pharmacology , Receptor, PAR-1/metabolism , Receptors, Thrombin/metabolism , Thrombin/metabolism , Thrombin/pharmacology
20.
Platelets ; 34(1): 2151995, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36529884

ABSTRACT

Ethanol and resveratrol have been shown to inhibit platelet aggregation. The aim of this study was to determine whether resveratrol has an additional effect on ethanol-induced inhibition of platelet aggregation. Ca2+ entry and subsequent aggregation of human platelets were measured by the fluorescence method and light transmittance method, respectively. Thromboxane B2 concentrations in media containing platelets were measured by using the enzyme-linked immunosorbent assay. Platelet aggregation induced by thrombin (0.025 U/ml) was significantly inhibited by preincubation of platelets with ethanol (0.5%). Preincubation with resveratrol (3.125 µM), which did not affect thrombin-induced platelet aggregation, significantly augmented the inhibitory effect of ethanol on platelet aggregation. Similar synergic effects of ethanol and resveratrol were found on aggregatory responses to collagen (2 µg/ml) and arachidonic acid (0.25 mM). On the other hand, the thrombin-induced increase in intracellular Ca2+ concentration ([Ca2+]i) was not affected by ethanol alone, resveratrol alone or both ethanol and resveratrol together. In nominally Ca2+-free medium, arachidonic acid (0.75 mM) caused a potent platelet aggregation, which was not affected by the presence of ethanol alone, resveratrol alone, or both of them together. Thromboxane B2 formation induced by thrombin was significantly inhibited by ethanol (0.5%) alone and resveratrol (3.125 µM) alone, and these inhibitory effects were significantly augmented in the presence of both ethanol and resveratrol together. Resveratrol shows an additive effect on ethanol-induced inhibition of platelet aggregation. This effect by resveratrol is partly explained by its inhibitory action on thromboxane A2 production in platelets. In addition, both ethanol and resveratrol attenuate platelet aggregation through acting on the Ca2+-dependent intra-platelet pathway after an increase in [Ca2+]i induced by thrombin.


Subject(s)
Platelet Aggregation , Thrombin , Humans , Resveratrol/pharmacology , Resveratrol/metabolism , Thrombin/pharmacology , Thrombin/metabolism , Arachidonic Acid/pharmacology , Arachidonic Acid/metabolism , Ethanol/pharmacology , Ethanol/metabolism , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/metabolism , Blood Platelets/metabolism , Thromboxane B2
SELECTION OF CITATIONS
SEARCH DETAIL