Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 384
Filter
1.
Chem Biol Drug Des ; 103(5): e14533, 2024 May.
Article in English | MEDLINE | ID: mdl-38684373

ABSTRACT

Hirudin is one of the specific inhibitors of thrombin, which has been confirmed to have strong bioactivities, including inhibiting tumors. However, the function and mechanism of hirudin and protease-activated receptor 1 (PAR-1) in diffuse large B-cell lymphoma (DLBCL) have not been clear. Detecting the expression PAR-1 in DLBCL tissues and cells by RT-qPCR and IHC. Transfected sh-NC, sh-PAR-1, or pcDNA3.1-PAR-1 in DLBCL cells or processed DLBCL cells through added thrombin, Vorapaxar, Recombinant hirudin (RH), or Na2S2O4 and co-culture with EA.hy926. And built DLBCL mice observed tumor growth. Detecting the expression of related genes by RT-qPCR, Western blot, IHC, and immunofluorescence, measured the cellular hypoxia with Hypoxyprobe-1 Kit, and estimated the cell inflammatory factors, proliferation, migration, invasion, and apoptosis by ELISA, CCK-8, flow cytometry, wound-healing and Transwell. Co-immunoprecipitation and pull-down measurement were used to verify the relationship. PAR-1 was highly expressed in DLBCL tissues and cells, especially in SUDHL2. Na2S2O4 induced SUDHL2 hypoxia, and PAR-1 did not influence thrombin-activated hypoxia. PAR-1 could promote SUDHL2 proliferation, migration, and invasion, and it was unrelated to cellular hypoxia. PAR-1 promoted proliferation, migration, and angiogenesis of EA.hy926 or SUDHL2 through up-regulation vascular endothelial growth factor (VEGF). RH inhibited tumor growth, cell proliferation, and migration, promoted apoptosis of DLBCL, and inhibited angiogenesis by down-regulating PAR-1-VEGF. RH inhibits proliferation, migration, and angiogenesis of DLBCL cells by down-regulating PAR-1-VEGF.


Subject(s)
Apoptosis , Cell Proliferation , Hirudins , Lymphoma, Large B-Cell, Diffuse , Neovascularization, Pathologic , Receptor, PAR-1 , Recombinant Proteins , Vascular Endothelial Growth Factor A , Humans , Hirudins/pharmacology , Receptor, PAR-1/metabolism , Receptor, PAR-1/antagonists & inhibitors , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Animals , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Mice , Cell Line, Tumor , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Apoptosis/drug effects , Recombinant Proteins/pharmacology , Recombinant Proteins/metabolism , Cell Proliferation/drug effects , Cell Movement/drug effects , Angiogenesis
2.
Int J Mol Sci ; 24(3)2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36768341

ABSTRACT

Diabetic encephalopathy (DE) is an inflammation-associated diabetes mellitus (DM) complication. Inflammation and coagulation are linked and are both potentially modulated by inhibiting the thrombin cellular protease-activated receptor 1 (PAR1). Our aim was to study whether coagulation pathway modulation affects DE. Diabetic C57BL/6 mice were treated with PARIN5, a novel PAR1 modulator. Behavioral changes in the open field and novel object recognition tests, serum neurofilament (NfL) levels and thrombin activity in central and peripheral nervous system tissue (CNS and PNS, respectively), brain mRNA expression of tumor necrosis factor α (TNF-α), Factor X (FX), prothrombin, and PAR1 were assessed. Subtle behavioral changes were detected in diabetic mice. These were accompanied by an increase in serum NfL, an increase in central and peripheral neural tissue thrombin activity, and TNF-α, FX, and prothrombin brain intrinsic mRNA expression. Systemic treatment with PARIN5 prevented the appearance of behavioral changes, normalized serum NfL and prevented the increase in peripheral but not central thrombin activity. PARIN5 treatment prevented the elevation of both TNF-α and FX but significantly elevated prothrombin expression. PARIN5 treatment prevents behavioral and neural damage in the DE model, suggesting it for future clinical research.


Subject(s)
Diabetes Mellitus, Experimental , Receptor, PAR-1 , Thrombin , Animals , Mice , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Disease Models, Animal , Inflammation/metabolism , Mice, Inbred C57BL , Prothrombin/metabolism , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-1/metabolism , Receptors, Thrombin/metabolism , RNA, Messenger/metabolism , Streptozocin , Thrombin/metabolism , Tumor Necrosis Factor-alpha/metabolism
3.
Cells ; 10(12)2021 12 13.
Article in English | MEDLINE | ID: mdl-34944024

ABSTRACT

BACKGROUND: Protease-activated receptor 1 (PAR1) and toll-like receptors (TLRs) are inflammatory mediators contributing to atherogenesis and atherothrombosis. Vorapaxar, which selectively antagonizes PAR1-signaling, is an approved, add-on antiplatelet therapy for secondary prevention. The non-hemostatic, platelet-independent, pleiotropic effects of vorapaxar have not yet been studied. METHODS AND RESULTS: Cellular targets of PAR1 signaling in the vasculature were identified in three patient cohorts with atherosclerotic disease. Evaluation of plasma biomarkers (n = 190) and gene expression in endomyocardial biopsies (EMBs) (n = 12) revealed that PAR1 expression correlated with endothelial activation and vascular inflammation. PAR1 colocalized with TLR2/4 in human carotid plaques and was associated with TLR2/4 gene transcription in EMBs. In addition, vorapaxar reduced atherosclerotic lesion size in apolipoprotein E-knock out (ApoEko) mice. This reduction was associated with reduced expression of vascular adhesion molecules and TLR2/4 presence, both in isolated murine endothelial cells and the aorta. Thrombin-induced uptake of oxLDL was augmented by additional TLR2/4 stimulation and abrogated by vorapaxar. Plaque-infiltrating pro-inflammatory cells were reduced in vorapaxar-treated ApoEko mice. A shift toward M2 macrophages paralleled a decreased transcription of pro-inflammatory cytokines and chemokines. CONCLUSIONS: PAR1 inhibition with vorapaxar may be effective in reducing residual thrombo-inflammatory event risk in patients with atherosclerosis independent of its effect on platelets.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/drug therapy , Lactones/administration & dosage , Pyridines/administration & dosage , Receptor, PAR-1/genetics , Vascular Diseases/drug therapy , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Female , Humans , Inflammation/drug therapy , Inflammation/genetics , Inflammation/pathology , Intercellular Adhesion Molecule-1/genetics , Lactones/adverse effects , Male , Mice , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Platelet Aggregation/drug effects , Pyridines/adverse effects , Receptor, PAR-1/antagonists & inhibitors , Thrombin/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Vascular Cell Adhesion Molecule-1/genetics , Vascular Diseases/genetics , Vascular Diseases/pathology
4.
Bioorg Med Chem ; 51: 116498, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34794000

ABSTRACT

Heptapeptide SFLLRNP is a receptor-tethered ligand of protease-activated receptor 1 (PAR-1), and its Phe at position 2 is essential for the aggregation of human platelets. To validate the structural elements of the Phe-phenyl group in receptor activation, we have synthesized a complete set of S/Phe/LLRNP peptides comprising different series of fluorophenylalanine isomers (Fn)Phe, where n = 1, 2, 3, and 5. Phe-2-phenyl was strongly suggested to be involved in the edge-to-face CH/π interaction with the receptor aromatic group. In the present study, to prove this receptor interaction definitively, we synthesized another series of peptide analogs containing (F4)Phe-isomers, with the phenyl group of each isomer possessing only one hydrogen atom at the ortho, meta, or para position. When the peptides were assayed for their platelet aggregation activity, S/(2,3,4,6-F4)Phe/LLRNP and S/(2,3,4,5-F4)Phe/LLRNP exhibited noticeable activity (34% and 6% intensities of the native peptide, respectively), whereas S/(2,3,5,6-F4)Phe/LLRNP was completely inactive. The results indicated that, at the ortho and meta positions but not at the para position, benzene-hydrogen atoms are required for the CH/π interaction to activate the receptor. The results provided a decisive evidence of the molecular recognition property of Phe, the phenyl benzene-hydrogen atom of which participates directly in the interaction with the receptor aromatic π plane.


Subject(s)
Peptide Fragments/pharmacology , Phenylalanine/pharmacology , Receptor, PAR-1/antagonists & inhibitors , Dose-Response Relationship, Drug , Healthy Volunteers , Humans , Ligands , Molecular Structure , Peptide Fragments/chemistry , Phenylalanine/chemistry , Platelet Aggregation/drug effects , Receptor, PAR-1/metabolism , Structure-Activity Relationship
5.
Life Sci ; 286: 120045, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34653426

ABSTRACT

OBJECTIVE: Endothelial dysfunction occurs as an early event in cardiovascular disease. Previously, vorapaxar, a proteinase-activated receptor-1 antagonist, was shown to cause endothelial damage in a cell culture study. Therefore, our study aimed to compare the effects of vorapaxar and parmodulin-2, proteinase-activated receptor-1 biased agonist, on human left internal mammary artery endothelial function in vitro. METHOD: Isolated arteries were hung in the organ baths. Acetylcholine responses (10-11-10-6 M) were obtained in endothelium-intact tissues the following incubation with vorapaxar/parmodulin-2 (10-6 M) to determine the effects of these molecules on the endothelium-dependent relaxation. Subsequently, endothelium-dependent relaxation responses of tissues were investigated in the presence of L-NAME (10-4 M), L-arginine (10-5 M), indomethacin (10-5 M), and charybdotoxin-apamin (10-7 M) in addition to vorapaxar/parmodulin-2 incubation. Besides, the effect of these molecules on endothelium-independent relaxation response was evaluated with sodium nitroprusside (10-11-10-6 M). Finally, the sections of human arteries were imaged using a transmission electron microscope, and the integrity of the endothelial layer was evaluated. RESULTS: We found that vorapaxar caused significant endothelial dysfunction by disrupting nitric oxide and endothelium-derived hyperpolarizing factor-dependent relaxation mechanisms. Parmodulin-2 did not cause endothelial damage. Neither vorapaxar nor parmodulin-2 disrupted endothelium-independent relaxation responses. The effect of vorapaxar on the endothelial layer was supported by the transmission electron microscope images. CONCLUSION: Parmodulin-2 may be a better option than vorapaxar in treating cardiovascular diseases since it can inhibit PAR-1 without caused endothelial dysfunction.


Subject(s)
Benzamides/pharmacology , Lactones/pharmacology , Pyridines/pharmacology , Receptor, PAR-1/antagonists & inhibitors , Acetylcholine/pharmacology , Adult , Apamin/pharmacology , Biological Factors/metabolism , Charybdotoxin/pharmacology , Endothelium, Vascular/drug effects , Humans , Male , Mammary Arteries/drug effects , Middle Aged , Muscle Relaxation/drug effects , Muscle, Smooth, Vascular/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitroprusside/pharmacology , Potassium Channel Blockers/pharmacology , Receptor, PAR-1/agonists , Receptor, PAR-1/metabolism , Vasodilation/drug effects
6.
Sci Rep ; 11(1): 16170, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34373558

ABSTRACT

Proteinase-activated receptor-1 (PAR1), triggered by thrombin and other serine proteinases such as tissue kallikrein-4 (KLK4), is a key driver of inflammation, tumor invasiveness and tumor metastasis. The PAR1 transmembrane G-protein-coupled receptor therefore represents an attractive target for therapeutic inhibitors. We thus used a computational design to develop a new PAR1 antagonist, namely, a catalytically inactive human KLK4 that acts as a proteinase substrate-capture reagent, preventing receptor cleavage (and hence activation) by binding to and occluding the extracellular R41-S42 canonical PAR1 proteolytic activation site. On the basis of in silico site-saturation mutagenesis, we then generated KLK4S207A,L185D, a first-of-a-kind 'decoy' PAR1 inhibitor, by mutating the S207A and L185D residues in wild-type KLK4, which strongly binds to PAR1. KLK4S207A,L185D markedly inhibited PAR1 cleavage, and PAR1-mediated MAPK/ERK activation as well as the migration and invasiveness of melanoma cells. This 'substrate-capturing' KLK4 variant, engineered to bind to PAR1, illustrates proof of principle for the utility of a KLK4 'proteinase substrate capture' approach to regulate proteinase-mediated PAR1 signaling.


Subject(s)
Kallikreins/metabolism , Receptor, PAR-1/antagonists & inhibitors , Amino Acid Substitution , Binding Sites , Cell Line, Tumor , Computer Simulation , Drug Design , Humans , Kallikreins/chemistry , Kallikreins/genetics , Kinetics , MCF-7 Cells , Mutagenesis, Site-Directed , Neoplasm Invasiveness/prevention & control , Protein Engineering , Protein Interaction Domains and Motifs , Proteolysis , Receptor, PAR-1/chemistry , Receptor, PAR-1/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Substrate Specificity , Thrombin/metabolism
7.
Int J Mol Sci ; 22(10)2021 May 12.
Article in English | MEDLINE | ID: mdl-34066284

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly fatal disease with a 5-year survival rate of less than 10% following diagnosis. The aggressive and invasive properties of pancreatic cancer tumors coupled with poor diagnostic options contribute to the high mortality rate since most patients present with late-stage disease. Accordingly, PDAC is linked to the highest rate of cancer-associated venous thromboembolic disease of all solid tumor malignancies. However, in addition to promoting clot formation, recent studies suggest that the coagulation system in PDAC mediates a reciprocal relationship, whereby coagulation proteases and receptors promote PDAC tumor progression and dissemination. Here, upregulation of tissue factor (TF) by tumor cells can drive local generation of the central coagulation protease thrombin that promotes cell signaling activity through protease-activated receptors (PARs) expressed by both tumor cells and multiple stromal cell subsets. Moreover, the TF-thrombin-PAR1 signaling axis appears to be a major mechanism of cancer progression in general and PDAC in particular. Here, we summarize the current literature regarding the role of PAR1 in PDAC and review possibilities for pharmacologically targeting PAR1 as a PDAC therapeutic approach.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Molecular Targeted Therapy , Pancreatic Neoplasms/drug therapy , Receptor, PAR-1/antagonists & inhibitors , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Humans , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology
8.
Psychopharmacology (Berl) ; 238(6): 1645-1656, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33624157

ABSTRACT

INTRODUCTION: Alzheimer's disease (AD) is a progressive brain disorder accompanied with synaptic failures and decline in cognitive and learning processes. Protease-activated receptor 1 (PAR1) is the major thrombin receptor in the brain that is implicated in synaptic plasticity and memory formation. In the current study, we hypothesized that inhibition of PAR1 would theoretically prevent amyloid beta (Aß) accumulation in the brain and then contribute to reduce risk of AD. The aim of the present study was to evaluate the effect of PAR1 inhibition by using SCH (as an inhibitor of PAR1) on spatial learning, memory, and synaptic plasticity in the CA1 region of the hippocampus in rat model of Alzheimer's disease. METHODS: For the induction of Alzheimer's disease, amyloid beta (Aß) 1-42 was injected in the CA1 region of the hippocampus. The rats were divided into four groups: group I (surgical sham); group II rat mode of Alzheimer's disease (AD); group III (SCH) (25 µg/kg) intraperitoneally (i.p.), and group IV (AD + SCH). After 14 days of protocol, the rats in group III received SCH and 30 min after injection behavioral and electrophysiological tests were performed. Learning and memory ability was assessed by Morris water maze and novel object recognition tests. Extracellular evoked field excitatory postsynaptic potentials (fEPSP) were recorded in the stratum radiatum of the CA1 area. RESULTS: Our results showed that AD rats showed impairments in learning and memory, and long-term potentiation (LTP) was not induced in these rats. However, injection of SCH overcame the AD-induced impairment in LTP generation in the CA1 area of the hippocampus and improved learning and memory impairment.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Memory Disorders/drug therapy , Receptor, PAR-1/antagonists & inhibitors , Alzheimer Disease/physiopathology , Animals , Cognition/drug effects , Disease Models, Animal , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Male , Maze Learning/drug effects , Neuronal Plasticity/drug effects , Rats , Rats, Wistar , Spatial Learning/drug effects , Synaptic Transmission/drug effects
9.
Exp Biol Med (Maywood) ; 246(6): 688-694, 2021 03.
Article in English | MEDLINE | ID: mdl-33302737

ABSTRACT

Acute respiratory disease caused by a novel coronavirus (SARS-CoV-2) has spread all over the world, since its discovery in 2019, Wuhan, China. This disease is called COVID-19 and already killed over 1 million people worldwide. The clinical symptoms include fever, dry cough, dyspnea, headache, dizziness, generalized weakness, vomiting, and diarrhea. Unfortunately, so far, there is no validated vaccine, and its management consists mainly of supportive care. Venous thrombosis and pulmonary embolism are highly prevalent in patients suffering from severe COVID-19. In fact, a prothrombotic state seems to be present in most fatal cases of the disease. SARS-CoV-2 leads to the production of proinflammatory cytokines, causing immune-mediated tissue damage, disruption of the endothelial barrier, and uncontrolled thrombogenesis. Thrombin is the key regulator of coagulation and fibrin formation. In severe COVID-19, a dysfunctional of physiological anticoagulant mechanisms leads to a progressive increase of thrombin activity, which is associated with acute respiratory distress syndrome development and a poor prognosis. Protease-activated receptor type 1 (PAR1) is the main thrombin receptor and may represent an essential link between coagulation and inflammation in the pathophysiology of COVID-19. In this review, we discuss the potential role of PAR1 inhibition and regulation in COVID-19 treatment.


Subject(s)
Blood Coagulation/physiology , COVID-19/pathology , Disseminated Intravascular Coagulation/pathology , Receptor, PAR-1/metabolism , Thrombin/metabolism , Anticoagulants/therapeutic use , Blood Coagulation/drug effects , Blood Coagulation Factors/metabolism , Disseminated Intravascular Coagulation/drug therapy , Humans , Pulmonary Embolism/pathology , Pulmonary Embolism/prevention & control , Receptor, PAR-1/antagonists & inhibitors , Receptors, Cell Surface/metabolism , SARS-CoV-2 , Venous Thrombosis/pathology , Venous Thrombosis/prevention & control , COVID-19 Drug Treatment
10.
Eur J Pharmacol ; 893: 173838, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33359646

ABSTRACT

Chronic kidney disease (CKD) involves interstitial fibrosis as an influential underlying pathological process associated with compromised renal function regardless of etiological cause of the injury. The tubulointerstitial fibrosis is found to be well correlated with declining renal function and its subsequent culmination into renal failure. Given the prominent role of thrombin in multiple diseases, it was tempting for us to investigate the outcome of a direct thrombin inhibitor in renal injury. We investigated the involvement of thrombin in renal injury and fibrosis by using an FDA approved orally active, direct thrombin inhibitor, dabigatran etexilate (DB). We used a robust experimental model of unilateral ureteral obstruction (UUO)-induced renal injury which shows progressive tubulointerstitial fibrosis (TIF) along with tubular injury and inflammation. The obstructed kidney showed severe TIF as compared to control kidneys. The administration of DB significantly inhibited UUO-induced collagen-1 and TIF by inhibition of thrombin activated protease activated receptor (PAR)-1 expression in fibrotic kidney. In addition, DB administration improved histoarchitecture of obstructed kidney, inhibited TGF-ß and SNAI2-induced epithelial-mesenchymal transition (EMT) program. Our study highlights the importance of thrombin signalling in TIF and provides strong evidences to support the notion that a direct thrombin inhibitor ameliorates TIF by PAR-1 mediated mechanism.


Subject(s)
Antithrombins/pharmacology , Dabigatran/pharmacology , Kidney Diseases/prevention & control , Kidney/drug effects , Receptor, PAR-1/antagonists & inhibitors , Animals , Collagen Type I/metabolism , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Fibrosis , Kidney/metabolism , Kidney/pathology , Kidney Diseases/etiology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Mice , Oxidative Stress/drug effects , Receptor, PAR-1/metabolism , Signal Transduction , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Ureteral Obstruction/complications
11.
Biomolecules ; 10(11)2020 11 13.
Article in English | MEDLINE | ID: mdl-33203057

ABSTRACT

Diabetic peripheral neuropathy (DPN) is a disabling common complication of diabetes mellitus (DM). Thrombin, a coagulation factor, is increased in DM and affects nerve function via its G-protein coupled protease activated receptor 1 (PAR1). METHODS: A novel PAR1 modulator (PARIN5) was designed based on the thrombin PAR1 recognition site. Coagulation, motor and sensory function and small fiber loss were evaluated by employing the murine streptozotocin diabetes model. RESULTS: PARIN5 showed a safe coagulation profile and showed no significant effect on weight or glucose levels. Diabetic mice spent shorter time on the rotarod (p <0.001), and had hypoalgesia (p <0.05), slow conduction velocity (p <0.0001) and reduced skin innervation (p <0.0001). Treatment with PARIN5 significantly improved rotarod performance (p <0.05), normalized hypoalgesia (p <0.05), attenuated slowing of nerve conduction velocity (p <0.05) and improved skin innervation (p <0.0001). CONCLUSION: PARIN5 is a novel pharmacological approach for prevention of DPN development, via PAR1 pathway modulation.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetic Neuropathies/drug therapy , Drug Delivery Systems/methods , Peptide Fragments/administration & dosage , Receptor, PAR-1/antagonists & inhibitors , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetic Neuropathies/metabolism , Male , Mice , Mice, Inbred C57BL , Receptor, PAR-1/metabolism
12.
Clin Sci (Lond) ; 134(21): 2873-2891, 2020 11 13.
Article in English | MEDLINE | ID: mdl-33078834

ABSTRACT

Protease-activated receptor (PAR)-1 has emerged as a key profibrotic player in various organs including kidney. PAR-1 activation leads to deposition of extracellular matrix (ECM) proteins in the tubulointerstitium and induction of epithelial-mesenchymal transition (EMT) during renal fibrosis. We tested the anti-fibrotic potential of vorapaxar, a clinically approved PAR-1 antagonist for cardiovascular protection, in an experimental kidney fibrosis model of unilateral ureteral obstruction (UUO) and an AKI-to-chronic kidney disease (CKD) transition model of unilateral ischemia-reperfusion injury (UIRI), and dissected the underlying renoprotective mechanisms using rat tubular epithelial cells. PAR-1 is activated mostly in the renal tubules in both the UUO and UIRI models of renal fibrosis. Vorapaxar significantly reduced kidney injury and ameliorated morphologic changes in both models. Amelioration of kidney fibrosis was evident from down-regulation of fibronectin (Fn), collagen and α-smooth muscle actin (αSMA) in the injured kidney. Mechanistically, inhibition of PAR-1 inhibited MAPK ERK1/2 and transforming growth factor-ß (TGF-ß)-mediated Smad signaling, and suppressed oxidative stress, overexpression of pro-inflammatory cytokines and macrophage infiltration into the kidney. These beneficial effects were recapitulated in cultured tubular epithelial cells in which vorapaxar ameliorated thrombin- and hypoxia-induced TGF-ß expression and ECM accumulation. In addition, vorapaxar mitigated capillary loss and the expression of adhesion molecules on the vascular endothelium during AKI-to-CKD transition. The PAR-1 antagonist vorapaxar protects against kidney fibrosis during UUO and UIRI. Its efficacy in human CKD in addition to CV protection warrants further investigation.


Subject(s)
Kidney/injuries , Lactones/pharmacology , Pyridines/pharmacology , Receptor, PAR-1/antagonists & inhibitors , Animals , Biomarkers/metabolism , Cell Hypoxia/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Extracellular Matrix Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibrosis , Inflammation/pathology , Kidney/drug effects , Kidney/pathology , Kidney Tubules/drug effects , Kidney Tubules/pathology , Macrophages/drug effects , Macrophages/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Oxidative Stress/drug effects , Rats , Reactive Oxygen Species/metabolism , Receptor, PAR-1/metabolism , Reperfusion Injury/complications , Smad3 Protein/metabolism , Thrombin/pharmacology , Transforming Growth Factor beta/metabolism , Up-Regulation/drug effects , Ureteral Obstruction/complications , Ureteral Obstruction/pathology
13.
Med Hypotheses ; 143: 110150, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32763660

ABSTRACT

COVID-19 due to the SARS-CoV-2 infection is a multi-systemic immune syndrome affecting mainly the lungs, oropharyngeal region, and other vascular endothelial beds. There are tremendous ongoing efforts for the aim of developing drugs against the COVID-19 syndrome-associated inflammation. However, currently no specific medicine is present for the absolute pharmacological cure of COVID-19 mucositis. The re-purposing/re-positioning of already existing drugs is a very important strategy for the management of ongoing pandemy since the development of a new drug needs decades. Apart from altering angiotensin signaling pathways, novel drug candidates for re-purposing comprise medications shall target COVID-19 pathobiology, including pharmaceutical formulations that antagonize proteinase-activated receptors (PARs), mainly PAR-1. Activation of the PAR-1, mediators and hormones impact on the hemostasis, endothelial activation, alveolar epithelial cells and mucosal inflammatory responses which are the essentials of the COVID-19 pathophysiology. In this context, Ankaferd hemostat (Ankaferd Blood Stopper, ABS) which is an already approved hemostatic agent affecting via vital erythroid aggregation and fibrinogen gamma could be a potential topical remedy for the mucosal management of COVID-19. ABS is a clinically safe and effective topical hemostatic agent of plant origin capable of exerting pleiotropic effects on the endothelial cells, angiogenesis, cell proliferation and vascular dynamics. ABS had been approved as a topically applied hemostatic agent for the management of post-surgical/dental bleedings and healing of infected inflammatory mucosal wounds. The anti-inflammatory and proteinase-activated receptor axis properties of ABS with a considerable amount of oestrogenic hormone presence highlight this unique topical hemostatic drug regarding the clinical re-positioning for COVID-19-associated mucositis. Topical ABS as a biological response modifier may lessen SARS-CoV-2 associated microthrombosis, endothelial dysfunction, oropharyngeal inflammation and mucosal lung damage. Moreover, PAR-1 inhibition ability of ABS might be helpful for reducing the initial virus propagation and mocasal spread of COVID-19.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Betacoronavirus , Coronavirus Infections/complications , Estrogens/physiology , Hemostatics/therapeutic use , Mucositis/drug therapy , Pandemics , Phytoestrogens/therapeutic use , Phytotherapy , Plant Extracts/therapeutic use , Pneumonia, Viral/complications , Receptor, PAR-1/antagonists & inhibitors , Administration, Topical , Age Distribution , Anti-Inflammatory Agents/administration & dosage , COVID-19 , Coronavirus Infections/blood , Coronavirus Infections/drug therapy , Coronavirus Infections/epidemiology , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/physiopathology , Drug Repositioning , Endothelium, Vascular/drug effects , Estrogens/agonists , Hemostatics/administration & dosage , Humans , Mucositis/etiology , Phytoestrogens/administration & dosage , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Pneumonia, Viral/blood , Pneumonia, Viral/drug therapy , Pneumonia, Viral/epidemiology , Receptor, PAR-1/physiology , SARS-CoV-2 , Stomatitis/drug therapy , Stomatitis/etiology , Thrombophilia/blood , Thrombophilia/etiology , COVID-19 Drug Treatment
14.
Br J Pharmacol ; 177(21): 4971-4974, 2020 11.
Article in English | MEDLINE | ID: mdl-32639031

ABSTRACT

In the search to rapidly identify effective therapies that will mitigate the morbidity and mortality of COVID-19, attention has been directed towards the repurposing of existing drugs. Candidates for repurposing include drugs that target COVID-19 pathobiology, including agents that alter angiotensin signalling. Recent data indicate that key findings in COVID-19 patients include thrombosis and endotheliitis. Activation of proteinase-activated receptor 1 (PAR1), in particular by the serine protease thrombin, is a critical element in platelet aggregation and coagulation. PAR1 activation also impacts on the actions of other cell types involved in COVID-19 pathobiology, including endothelial cells, fibroblasts and pulmonary alveolar epithelial cells. Vorapaxar is an approved inhibitor of PAR1, used for treatment of patients with myocardial infarction or peripheral arterial disease. We discuss evidence for a possible beneficial role for vorapaxar in the treatment of COVID-19 patients and other as-yet non-approved antagonists of PAR1 and proteinase-activated receptor 4 (PAR4). LINKED ARTICLES: This article is part of a themed issue on The Pharmacology of COVID-19. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.21/issuetoc.


Subject(s)
Coronavirus Infections/drug therapy , Lactones/administration & dosage , Pneumonia, Viral/drug therapy , Pyridines/administration & dosage , Receptor, PAR-1/antagonists & inhibitors , Animals , Betacoronavirus/isolation & purification , COVID-19 , Coronavirus Infections/virology , Drug Repositioning , Humans , Lactones/pharmacology , Pandemics , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/pharmacology , Pneumonia, Viral/virology , Pyridines/pharmacology , Receptor, PAR-1/metabolism , Receptors, Thrombin/antagonists & inhibitors , Receptors, Thrombin/metabolism , SARS-CoV-2 , COVID-19 Drug Treatment
15.
Arterioscler Thromb Vasc Biol ; 40(8): 1891-1904, 2020 08.
Article in English | MEDLINE | ID: mdl-32493172

ABSTRACT

OBJECTIVE: Platelets are critical to the formation of a hemostatic plug and the pathogenesis of atherothrombosis. Preclinical animal models, especially the mouse, provide an important platform to assess the efficacy and safety of antiplatelet drugs. However, these studies are limited by inherent differences between human and mouse platelets and the species-selectivity of many drugs. To circumvent these limitations, we developed a new protocol for the adoptive transfer of human platelets into thrombocytopenic nonobese diabetic/severe combined immune deficiency mice, that is, a model where all endogenous platelets are replaced by human platelets in mice accepting xenogeneic tissues. Approach and Results: To demonstrate the power of this new model, we visualized and quantified hemostatic plug formation and stability by intravital spinning disk confocal microscopy following laser ablation injury to the saphenous vein. Integrin αIIbß3-dependent hemostatic platelet plug formation was achieved within ≈30 seconds after laser ablation injury in humanized platelet mice. Pretreatment of mice with standard dual antiplatelet therapy (Aspirin+Ticagrelor) or PAR1 inhibitor, L-003959712 (an analog of vorapaxar), mildly prolonged the bleeding time and significantly reduced platelet adhesion to the site of injury. Consistent with findings from clinical trials, inhibition of PAR1 in combination with dual antiplatelet therapy markedly prolonged bleeding time in humanized platelet mice. CONCLUSIONS: We propose that this novel mouse model will provide a robust platform to test and predict the safety and efficacy of experimental antiplatelet drugs and to characterize the hemostatic function of synthetic, stored and patient platelets.


Subject(s)
Blood Platelets/physiology , Hemostasis/drug effects , Adoptive Transfer , Animals , Benzofurans/pharmacology , Carbamates/pharmacology , Dual Anti-Platelet Therapy/adverse effects , Humans , Male , Mice , Models, Animal , Receptor, PAR-1/antagonists & inhibitors
16.
Arterioscler Thromb Vasc Biol ; 40(8): 1905-1917, 2020 08.
Article in English | MEDLINE | ID: mdl-32580633

ABSTRACT

OBJECTIVE: Remodeling of the extracellular matrix plays a vital role in cardiovascular diseases. Using a mouse model of postnatal ascending aortic aneurysms (termed Fbln4SMKO), we have reported that abnormal mechanosensing led to aneurysm formation in Fbln4SMKO with an upregulation of the mechanosensitive transcription factor, Egr1 (Early growth response 1). However, the role of Egr1 and its upstream regulator(s) in the initiation of aneurysm development and their relationship to an aneurysmal microenvironment are unknown. Approach and Results: To investigate the contribution of Egr1 in the aneurysm development, we deleted Egr1 in Fbln4SMKO mice and generated double knockout mice (DKO, Fbln4SMKO; Egr1-/-). Aneurysms were prevented in DKO mice (42.8%) and Fbln4SMKO; Egr1+/- mice (26%). Ingenuity Pathway Analysis identified PAR1 (protease-activated receptor 1) as a potential Egr1 upstream gene. Protein and transcript levels of PAR1 were highly increased in Fbln4SMKO aortas at postnatal day 1 before aneurysm formed, together with active thrombin and MMP (matrix metalloproteinase)-9, both of which serve as a PAR1 activator. Concordantly, protein levels of PAR1, Egr1, and thrombin were significantly increased in human thoracic aortic aneurysms. In vitro cyclic stretch assays (1.0 Hz, 20% strain, 8 hours) using mouse primary vascular smooth muscle cells induced marked expression of PAR1 and secretion of prothrombin in response to mechanical stretch. Thrombin was sufficient to induce Egr1 expression in a PAR1-dependent manner. CONCLUSIONS: We propose that thrombin, MMP-9, and mechanical stimuli in the Fbln4SMKO aorta activate PAR1, leading to the upregulation of Egr1 and initiation of ascending aortic aneurysms.


Subject(s)
Aortic Aneurysm, Thoracic/etiology , Early Growth Response Protein 1/physiology , Extracellular Matrix Proteins/physiology , Receptor, PAR-1/physiology , Aged , Aged, 80 and over , Animals , Extracellular Matrix Proteins/deficiency , Female , Humans , Male , Matrix Metalloproteinase 9/physiology , Mice , Middle Aged , Receptor, PAR-1/antagonists & inhibitors , Stress, Mechanical , Thrombin/pharmacology
17.
J Am Heart Assoc ; 9(12): e015616, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32495720

ABSTRACT

Background Although PAR-1 (protease-activated receptor-1) exerts important functions in the pathophysiology of the cardiovascular system, the role of PAR-1 signaling in heart failure development remains largely unknown. We tested the hypothesis that PAR-1 signaling inhibition has protective effects on the progression of cardiac remodeling induced by chronic renin-angiotensin system activation using renin-overexpressing hypertensive (Ren-Tg) mice. Methods and Results We treated 12- to 16-week-old male wild-type (WT) mice and Ren-Tg mice with continuous subcutaneous infusion of the PAR-1 antagonist SCH79797 or vehicle for 4 weeks. The thicknesses of interventricular septum and the left ventricular posterior wall were greater in Ren-Tg mice than in WT mice, and SCH79797 treatment significantly decreased these thicknesses in Ren-Tg mice. The cardiac fibrosis area and monocyte/macrophage deposition were greater in Ren-Tg mice than in WT mice, and both conditions were attenuated by SCH79797 treatment. Cardiac mRNA expression levels of PAR-1, TNF-α (tumor necrosis factor-α), TGF-ß1 (transforming growth factor-ß1), and COL3A1 (collagen type 3 α1 chain) and the ratio of ß-myosin heavy chain (ß-MHC) to α-MHC were all greater in Ren-Tg mice than in WT mice; SCH79797 treatment attenuated these increases in Ren-Tg mice. Prothrombin fragment 1+2 concentration and factor Xa in plasma were greater in Ren-Tg mice than in WT mice, and both conditions were unaffected by SCH79797 treatment. In isolated cardiac fibroblasts, both thrombin and factor Xa enhanced ERK1/2 (extracellular signal-regulated kinase 1/2) phosphorylation, and SCH79797 pretreatment abolished this enhancement. Furthermore, gene expression of PAR-1, TGF-ß1, and COL3A1 were enhanced by factor Xa, and all were inhibited by SCH79797. Conclusions The results indicate that PAR-1 signaling is involved in cardiac remodeling induced by renin-angiotensin system activation, which may provide a novel therapeutic target for heart failure.


Subject(s)
Hypertension/drug therapy , Hypertrophy, Left Ventricular/prevention & control , Myocardium/metabolism , Pyrroles/pharmacology , Quinazolines/pharmacology , Receptor, PAR-1/antagonists & inhibitors , Renin/metabolism , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Animals , Collagen Type III/genetics , Collagen Type III/metabolism , Cytokines/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosis , HEK293 Cells , Humans , Hypertension/genetics , Hypertension/metabolism , Hypertension/physiopathology , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Inflammation Mediators/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Myocardium/pathology , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism , Renin/genetics , Signal Transduction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Up-Regulation
18.
Am J Physiol Renal Physiol ; 318(5): F1067-F1073, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32200667

ABSTRACT

Protease-activated receptors (PARs) are coagulation protease targets, and they increase expression of inflammatory cytokines and chemokines in various diseases. Of all PARs, previous reports have shown that PAR1 or PAR2 inhibition is protective against diabetic glomerular injury. However, how PAR1 and PAR2 cooperatively contribute to diabetic kidney disease (DKD) pathogenesis and whether dual blockade of PARs is more effective in DKD remain elusive. To address this issue, male type I diabetic Akita mice heterozygous for endothelial nitric oxide synthase were used as a model of DKD. Mice (4 mo old) were divided into four treatment groups and administered vehicle, PAR1 antagonist (E5555, 60 mg·kg-1·day-1), PAR2 antagonist (FSLLRY, 3 mg·kg-1·day-1), or E5555 + FSLLRY for 4 wk. The results showed that the urinary albumin creatinine ratio was significantly reduced when both PAR1 and PAR2 were blocked with E5555 + FSLLRY compared with the vehicle-treated group. Dual blockade of PAR1 and PAR2 by E5555 + FSLLRY additively ameliorated histological injury, including mesangial expansion, glomerular macrophage infiltration, and collagen type IV deposition. Marked reduction of inflammation- and fibrosis-related gene expression in the kidney was also observed. In vitro, PAR1 and PAR2 agonists additively increased mRNA expression of macrophage chemoattractant protein 1 or plasminogen activator inhibitor-1 in human endothelial cells. Changes induced by the PAR1 agonist were blocked by a NF-κB inhibitor, whereas those of the PAR2 agonist were blocked by MAPK and/or NF-κB inhibitors. These findings suggest that PAR1 and PAR2 additively contribute to DKD pathogenesis and that dual blockade of both could be a novel therapeutic option for treatment of patients with DKD.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Diabetic Nephropathies/prevention & control , Imines/pharmacology , Kidney/drug effects , Oligopeptides/pharmacology , Pyridines/pharmacology , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-2/antagonists & inhibitors , Albuminuria/genetics , Albuminuria/metabolism , Albuminuria/prevention & control , Animals , Cell Line , Cell Proliferation/drug effects , Collagen Type IV/metabolism , Cytokines/metabolism , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Disease Models, Animal , Drug Therapy, Combination , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibrosis , Humans , Inflammation Mediators/metabolism , Kidney/metabolism , Kidney/pathology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics , Receptor, PAR-1/metabolism , Receptor, PAR-2/metabolism , Signal Transduction
19.
Bioorg Med Chem Lett ; 30(8): 127046, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32122739

ABSTRACT

A series of novel vorapaxar analogues with different amino substitutes at the C-7, C-9a and aromatic substitutes at the C-4 position were designed, synthesized, and evaluated for their inhibitory activity to PAR-1. Several compounds showed good potency in antagonist activity based on the intracellular calcium mobilization assay and excellent pharmacokinetics profile in rats. Among these analogues, 3d exhibited excellent PAR-1 inhibitory activity (IC50 = 0.18 µM) and the lower ability to cross the blood-brain barrier compared with vorapaxar (IC50 = 0.25 µM). Compound 3d has the potential to be developed as a new generation of PAR-1 antagonists with a better therapeutic window.


Subject(s)
Drug Design , Lactones/pharmacology , Pyridines/pharmacology , Receptor, PAR-1/antagonists & inhibitors , Dose-Response Relationship, Drug , Humans , Lactones/chemical synthesis , Lactones/chemistry , Models, Molecular , Molecular Structure , Pyridines/chemical synthesis , Pyridines/chemistry , Receptor, PAR-1/metabolism , Structure-Activity Relationship
20.
Breast Cancer Res Treat ; 180(2): 379-384, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32034579

ABSTRACT

PURPOSE: Protease-activated receptor 1 (PAR1) is a signaling protein ubiquitously present on the surface of tumor cells, and its homologous protein fragment, PAR1-activating peptide (P1AP), can inhibit protein signal transduction of PAR1/G in tumor cells. pH (Low) insertion peptide (pHLIP) can target the acidic tumor microenvironment (TME) and can be used as an excellent carrier to deliver P1AP to tumor cells for therapeutic purposes. METHODS: PAR1 expression on the surface of MDA-MB-231 cells and human MCF10A mammary epithelial cells was observed. The binding between fluorescent-labeled pHLIP(Var7)-P1AP and MDA-MB-231 cells under different pH values was analyzed. The effect of pHLIP(Var7)-P1AP on the proliferation of MDA-MB-231 cells was analyzed under the conditions of pH 7.4 and 6.0. RESULTS: PAR1 was highly expressed on the surface of MDA-MB-231 cells. In an acidic environment (pH 6.0 and 5.0), fluorescent-labeled pHLIP(Var7)-P1AP and MDA-MB-231 cells had a high binding ability, and the binding ability increased with the decrease in pH. In an acidic environment (pH 6.0), pHLIP(Var7)-P1AP significantly inhibited MDA-MB-231 cell proliferation. With 0.5 µg, 1 µg, 2 µg, 4 µg, and 8 µg of pHLIP(Var7)-P1AP, the cell proliferation inhibition rates were 3.39%, 5.27%, 14.29%, 22.14%, and 35.69%, respectively. CONCLUSION: PAR1 was highly expressed on the surface of MDA-MB-231 cells. pHLIP(Var7)-P1AP can effectively target MDA-MB-231 cells in an acidic environment and inhibit the growth of MDA-MB-231 cells by inhibiting the signal transduction of PAR1/G protein.


Subject(s)
Drug Delivery Systems/methods , Membrane Proteins/pharmacology , Oligopeptides/pharmacology , Receptor, PAR-1/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation , Female , Humans , Hydrogen-Ion Concentration , Signal Transduction , Triple Negative Breast Neoplasms/chemistry , Triple Negative Breast Neoplasms/metabolism , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...