Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Circ Heart Fail ; 16(10): e010621, 2023 10.
Article in English | MEDLINE | ID: mdl-37477012

ABSTRACT

BACKGROUND: PAR1 (protease-activated receptor-1) contributes to acute thrombosis, but it is not clear whether the receptor is involved in deleterious inflammatory and profibrotic processes in heart failure. Here, we employ the pepducin technology to determine the effects of targeting PAR1 in a mouse heart failure with reduced ejection fraction model. METHODS: After undergoing transverse aortic constriction pressure overload or sham surgery, C57BL/6J mice were randomized to daily sc PZ-128 pepducin or vehicle, and cardiac function, inflammation, fibrosis, and molecular analyses conducted at 7 weeks RESULTS: After 7 weeks of transverse aortic constriction, vehicle mice had marked increases in macrophage/monocyte infiltration and fibrosis of the left ventricle as compared with Sham mice. PZ-128 treatment significantly suppressed the inflammatory cell infiltration and cardiac fibrosis. Despite no effect on myocyte cell hypertrophy, PZ-128 afforded a significant reduction in overall left ventricle weight and completely protected against the transverse aortic constriction-induced impairments in left ventricle ejection fraction. PZ-128 significantly suppressed transverse aortic constriction-induced increases in an array of genes involved in myocardial stress, fibrosis, and inflammation. CONCLUSIONS: The PZ-128 pepducin is highly effective in protecting against cardiac inflammation, fibrosis, and loss of left ventricle function in a mouse model.


Subject(s)
Heart Failure , Mice , Animals , Receptor, PAR-1 , Mice, Inbred C57BL , Myocardium/pathology , Fibrosis , Inflammation/pathology , Disease Models, Animal
2.
Hepatology ; 77(5): E111-E112, 2023 May 01.
Article in English | MEDLINE | ID: mdl-36869845
3.
Hepatology ; 76(6): 1778-1793, 2022 12.
Article in English | MEDLINE | ID: mdl-35603482

ABSTRACT

BACKGROUND AND AIMS: Insulin resistance and poor glycemic control are key drivers of the development of NAFLD and have recently been shown to be associated with fibrosis progression in NASH. However, the underlying mechanisms involving dysfunctional glucose metabolism and relationship with NAFLD/NASH progression remain poorly understood. We set out to determine whether protease-activated receptor 2 (PAR2), a sensor of extracellular inflammatory and coagulation proteases, links NAFLD and NASH with liver glucose metabolism. APPROACH AND RESULTS: Here, we demonstrate that hepatic expression of PAR2 increases in patients and mice with diabetes and NAFLD/NASH. Mechanistic studies using whole-body and liver-specific PAR2-knockout mice reveal that hepatic PAR2 plays an unexpected role in suppressing glucose internalization, glycogen storage, and insulin signaling through a bifurcating Gq -dependent mechanism. PAR2 activation downregulates the major glucose transporter of liver, GLUT2, through Gq -MAPK-FoxA3 and inhibits insulin-Akt signaling through Gq -calcium-CaMKK2 pathways. Therapeutic dosing with a liver-homing pepducin, PZ-235, blocked PAR2-Gq signaling and afforded significant improvements in glycemic indices and HbA1c levels in severely diabetic mice. CONCLUSIONS: This work provides evidence that PAR2 is a major regulator of liver glucose homeostasis and a potential target for the treatment of diabetes and NASH.


Subject(s)
Diabetes Mellitus, Experimental , Insulin Resistance , Non-alcoholic Fatty Liver Disease , Mice , Animals , Non-alcoholic Fatty Liver Disease/metabolism , Receptor, PAR-2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Liver/metabolism , Insulin/metabolism , Glucose/metabolism , Mice, Knockout
5.
Methods Mol Biol ; 2383: 307-333, 2022.
Article in English | MEDLINE | ID: mdl-34766299

ABSTRACT

Pepducins are lipidated peptides that target the intracellular loops of G protein-coupled receptors (GPCRs) in order to modulate transmembrane signaling to internally located effectors. With a wide array of potential activities ranging from partial, biased, or full agonism to antagonism, pepducins represent a versatile class of compounds that can be used to potentially treat diverse human diseases or be employed as novel tools to probe complex mechanisms of receptor activation and signaling in cells and in animals. Here, we describe a number of different pepducins including an advanced compound, PZ-128, that has successfully progressed through phase 2 clinical trials in cardiac patients demonstrating safety and efficacy in suppressing myonecrosis and arterial thrombosis.


Subject(s)
Lipopeptides/therapeutic use , Animals , Humans , Receptors, G-Protein-Coupled , Signal Transduction
6.
Mol Cancer Res ; 20(3): 373-386, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34753803

ABSTRACT

MALT1 is the effector protein of the CARMA/Bcl10/MALT1 (CBM) signalosome, a multiprotein complex that drives pro-inflammatory signaling pathways downstream of a diverse set of receptors. Although CBM activity is best known for its role in immune cells, emerging evidence suggests that it plays a key role in the pathogenesis of solid tumors, where it can be activated by selected G protein-coupled receptors (GPCR). Here, we demonstrated that overexpression of GPCRs implicated in breast cancer pathogenesis, specifically the receptors for Angiotensin II and thrombin (AT1R and PAR1), drove a strong epithelial-to-mesenchymal transition (EMT) program in breast cancer cells that is characteristic of claudin-low, triple-negative breast cancer (TNBC). In concert, MALT1 was activated in these cells and contributed to the dramatic EMT phenotypic changes through regulation of master EMT transcription factors including Snail and ZEB1. Importantly, blocking MALT1 signaling, through either siRNA-mediated depletion of MALT1 protein or pharmacologic inhibition of its activity, was effective at partially reversing the molecular and phenotypic indicators of EMT. Treatment of mice with mepazine, a pharmacologic MALT1 inhibitor, reduced growth of PAR1+, MDA-MB-231 xenografts and had an even more dramatic effect in reducing the burden of metastatic disease. These findings highlight MALT1 as an attractive therapeutic target for claudin-low TNBCs harboring overexpression of one or more selected GPCRs. IMPLICATIONS: This study nominates a GPCR/MALT1 signaling axis as a pathway that can be pharmaceutically targeted to abrogate EMT and metastatic progression in TNBC, an aggressive form of breast cancer that currently lacks targeted therapies.


Subject(s)
Triple Negative Breast Neoplasms , Animals , Cell Line, Tumor , Cell Movement , Claudins/pharmacology , Claudins/therapeutic use , Epithelial-Mesenchymal Transition , Humans , Mice , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Receptor, PAR-1/therapeutic use , Triple Negative Breast Neoplasms/metabolism
8.
Arterioscler Thromb Vasc Biol ; 41(1): e33-e45, 2021 01.
Article in English | MEDLINE | ID: mdl-33267659

ABSTRACT

OBJECTIVE: 12-LOX (12-lipoxygenase) produces a number of bioactive lipids including 12(S)-HETE that are involved in inflammation and platelet reactivity. The GPR31 (G-protein-coupled receptor 31) is the proposed receptor of 12(S)-HETE; however, it is not known whether the 12(S)-HETE-GPR31 signaling axis serves to enhance or inhibit platelet activity. Approach and Results: Using pepducin technology and biochemical approaches, we provide evidence that 12(S)-HETE-GPR31 signals through Gi to enhance PAR (protease-activated receptor)-4-mediated platelet activation and arterial thrombosis using both human platelets and mouse carotid artery injury models. 12(S)-HETE suppressed AC (adenylyl cyclase) activity through GPR31 and resulted in Rap1 (Ras-related protein 1) and p38 activation and low but detectable calcium flux but did not induce platelet aggregation. A GPR31 third intracellular (i3) loop-derived pepducin, GPR310 (G-protein-coupled receptor 310), significantly inhibited platelet aggregation in response to thrombin, collagen, and PAR4 agonist, AYPGKF, in human and mouse platelets but relative sparing of PAR1 agonist SFLLRN in human platelets. GPR310 treatment gave a highly significant 80% protection (P=0.0018) against ferric chloride-induced carotid artery injury in mice by extending occlusion time, without any effect on tail bleeding. PAR4-mediated dense granule secretion and calcium flux were both attenuated by GPR310. Consistent with these results, GPR310 inhibited 12(S)-HETE-mediated and PAR4-mediated Rap1-GTP and RASA3 translocation to the plasma membrane and attenuated PAR4-Akt and ERK activation. GPR310 caused a right shift in thrombin-mediated human platelet aggregation, comparable to the effects of inhibition of the Gi-coupled P2Y12 receptor. Co-immunoprecipitation studies revealed that GPR31 and PAR4 form a heterodimeric complex in recombinant systems. CONCLUSIONS: The 12-LOX product 12(S)-HETE stimulates GPR31-Gi-signaling pathways, which enhance thrombin-PAR4 platelet activation and arterial thrombosis in human platelets and mouse models. Suppression of this bioactive lipid pathway, as exemplified by a GPR31 pepducin antagonist, may provide beneficial protective effects against platelet aggregation and arterial thrombosis with minimal effect on hemostasis.


Subject(s)
Blood Platelets/metabolism , Carotid Artery Thrombosis/blood , Hemostasis , Platelet Aggregation , Receptors, G-Protein-Coupled/blood , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/blood , Animals , CHO Cells , Carotid Artery Thrombosis/prevention & control , Cricetulus , Disease Models, Animal , Female , Fibrinolytic Agents/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/blood , Humans , Male , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Thrombin/blood , Signal Transduction , Thrombin/metabolism
9.
Arterioscler Thromb Vasc Biol ; 40(12): 2990-3003, 2020 12.
Article in English | MEDLINE | ID: mdl-33028101

ABSTRACT

OBJECTIVE: Arterial thrombosis leading to ischemic injury worsens the prognosis of many patients with cardiovascular disease. PZ-128 is a first-in-class pepducin that reversibly inhibits PAR1 (protease-activated receptor 1) on platelets and other vascular cells by targeting the intracellular surface of the receptor. The TRIP-PCI (Thrombin Receptor Inhibitory Pepducin in Percutaneous Coronary Intervention) trial was conducted to assess the safety and efficacy of PZ-128 in patients undergoing cardiac catheterization with intent to perform percutaneous coronary intervention. Approach and Results: In this randomized, double-blind, placebo-controlled, phase 2 trial, 100 patients were randomly assigned (2:1) to receive PZ-128 (0.3 or 0.5 mg/kg), or placebo in a 2-hour infusion initiated just before the start of cardiac catheterization, on top of standard oral antiplatelet therapy. Rates of the primary end point of bleeding were not different between the combined PZ-128 doses (1.6%, 1/62) and placebo group (0%, 0/35). The secondary end points of major adverse coronary events at 30 and 90 days did not significantly differ but were numerically lower in the PZ-128 groups (0% and 2% in the PZ-128 groups, 6% and 6% with placebo, p=0.13, p=0.29, respectively). In the subgroup of patients with elevated baseline cardiac troponin I, the exploratory end point of 30-day major adverse coronary events + myocardial injury showed 83% events in the placebo group versus 31% events in the combined PZ-128 drug groups, an adjusted relative risk of 0.14 (95% CI, 0.02-0.75); P=0.02. CONCLUSIONS: In this first-in-patient experience, PZ-128 added to standard antiplatelet therapy appeared to be safe, well tolerated, and potentially reduced periprocedural myonecrosis, thus providing the basis for further clinical trials. Registration: URL: https://www.clinicaltrials.gov. Unique identifier: NCT02561000.


Subject(s)
Acute Coronary Syndrome/therapy , Blood Platelets/drug effects , Cardiac Catheterization , Cell-Penetrating Peptides/administration & dosage , Coronary Artery Disease/therapy , Lipopeptides/administration & dosage , Myocardium/pathology , Percutaneous Coronary Intervention , Platelet Aggregation Inhibitors/administration & dosage , Receptor, PAR-1/agonists , Thrombosis/prevention & control , Acute Coronary Syndrome/diagnostic imaging , Aged , Blood Platelets/metabolism , Cardiac Catheterization/adverse effects , Cardiac Catheterization/instrumentation , Cell-Penetrating Peptides/adverse effects , Cell-Penetrating Peptides/pharmacokinetics , Coronary Artery Disease/diagnostic imaging , Double-Blind Method , Female , Humans , Infusions, Intravenous , Lipopeptides/adverse effects , Lipopeptides/pharmacokinetics , Male , Middle Aged , Necrosis , Percutaneous Coronary Intervention/adverse effects , Percutaneous Coronary Intervention/instrumentation , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/pharmacokinetics , Proof of Concept Study , Prospective Studies , Receptor, PAR-1/metabolism , Recurrence , Stents , Thrombosis/blood , Thrombosis/etiology , Time Factors , Treatment Outcome , United States
10.
Mol Metab ; 29: 99-113, 2019 11.
Article in English | MEDLINE | ID: mdl-31668396

ABSTRACT

OBJECTIVE: Increases in hepatic and plasma cholesterol occur in patients with nonalcoholic fatty liver disease (NAFLD), although the reason for this is not well understood. We investigated whether Protease-Activated Receptor 2 (PAR2) plays a role in cholesterol and lipid homeostasis in NAFLD. METHODS: Human liver biopsies (n = 108) were quantified for PAR2 expression from NAFLD cases randomly selected and stratified by liver fibrosis stage, the primary predictor for clinical outcomes, while controlling for age, gender, and BMI between fibrosis groups. Demographic data and laboratory studies on plasma samples were obtained within 6 months of liver biopsy. Wild-type and PAR2-KO (C57BL/6 F2rl1-/-) mice were fed either normal or high fat diet for 16 weeks and plasma and liver assayed for lipids and soluble metabolites. RESULTS: Severity of NAFLD and plasma cholesterol levels significantly correlated with hepatocyte PAR2 expression in NAFLD patients. Conversely, PAR2 deficiency in mice resulted in reduced expression of key hepatic genes involved in cholesterol synthesis, a 50% drop in plasma and total liver cholesterol, and induced a reverse cholesterol transport system that culminated in 25% higher fecal bile acid output. PAR2-deficient mice exhibited enhanced fatty acid ß-oxidation with a ketogenic shift and an unexpected increase in liver glycogenesis. Mechanistic studies identified Gi-Jnk1/2 as key downstream effectors of protease-activated PAR2 in the regulation of lipid and cholesterol homeostasis in liver. CONCLUSIONS: These data indicate that PAR2 may be a new target for the suppression of plasma cholesterol and hepatic fat accumulation in NAFLD and related metabolic conditions.


Subject(s)
Cholesterol/metabolism , Lipid Metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Receptor, PAR-2/metabolism , Adult , Aged , Animals , Cholesterol/blood , Diet, High-Fat , Disease Progression , Female , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Lipid Peroxidation , Liver/pathology , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Receptor, PAR-2/deficiency , Receptor, PAR-2/genetics
11.
Oncogene ; 38(49): 7384-7398, 2019 12.
Article in English | MEDLINE | ID: mdl-31420608

ABSTRACT

Protease-activated receptor 1 (PAR1), a thrombin-responsive G protein-coupled receptor (GPCR), is implicated in promoting metastasis in multiple tumor types, including both sarcomas and carcinomas, but the molecular mechanisms responsible remain largely unknown. We previously discovered that PAR1 stimulation in endothelial cells leads to activation of NF-κB, mediated by a protein complex comprised of CARMA3, Bcl10, and the MALT1 effector protein (CBM complex). Given the strong association between NF-κB and metastasis, we hypothesized that this CBM complex could play a critical role in the PAR1-driven metastatic progression of specific solid tumors. In support of our hypothesis, we demonstrate that PAR1 stimulation results in NF-κB activation in both osteosarcoma and breast cancer, which is suppressed by siRNA-mediated MALT1 knockdown, suggesting that an intact CBM complex is required for the response in both tumor cell types. We identify several metastasis-associated genes that are upregulated in a MALT1-dependent manner after PAR1 stimulation in cancer cells, including those encoding the matrix remodeling protein, MMP9, and the cytokines, IL-1ß and IL-8. Further, exogenous expression of PAR1 in MCF7 breast cancer cells confers highly invasive and metastatic behavior which can be blocked by CRISPR/Cas9-mediated MALT1 knockout. Importantly, we find that PAR1 stimulation induces MALT1 protease activity in both osteosarcoma and breast cancer cells, an activity that is mechanistically linked to NF-κB activation and potentially other responses associated with aggressive phenotype. Several small molecule MALT1 protease inhibitors have recently been described that could therefore represent promising new therapeutics for the prevention and/or treatment of PAR1-driven tumor metastasis.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , NF-kappa B/metabolism , Osteosarcoma/pathology , Receptor, PAR-1/metabolism , Animals , Apoptosis , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Movement , Cell Proliferation , Female , Humans , Mice , Mice, Nude , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , NF-kappa B/genetics , Osteosarcoma/genetics , Osteosarcoma/metabolism , Receptor, PAR-1/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Thromb Res ; 177: 59-69, 2019 May.
Article in English | MEDLINE | ID: mdl-30851630

ABSTRACT

ACS patients undergoing percutaneous coronary intervention (PCI) when treated with bivalirudin and clopidogrel had increased frequency of early stent thrombosis. 24 patients referred for intervention with planned bivalirudin therapy, not previously treated with a P2Y12 inhibitor and not receiving heparins or αIIbß3 inhibitors were randomized to treatment with either clopidogrel (600 mg) or prasugrel (60 mg). Platelet aggregation (PA) was measured by light transmission aggregometry (LTA) of platelet-rich plasma in response to ADP, PAR1/PAR4 thrombin receptor agonists and collagen at baseline and at 1, 2, 4 and 16 h following the cessation of bivalirudin infusion. Prasugrel-mediated inhibition of PA was significantly greater than that of clopidogrel at all time points for ADP as well as PAR1. There was an unanticipated, significantly greater protection of PAR4-mediated platelet aggregation only detected with prasugrel and not observed with clopidogrel. We further examined the effect of the hyperreactive PAR4 Thr120 variant in the protease-activated receptor 4 (PAR4), single nucleotide polymorphism (SNP) rs773902 on aggregation protection. The PAR4 protective effect with prasugrel was lost in individuals carrying the PAR4 Thr120 variant, and not in Ala120 homozygote. PAR1, ADP and collagen inhibition was not significantly affected in the hyperreactive PAR4 Thr120 variant. We documented that the P2Y12 ADP receptor-mediated regulation of the strength of the high-affinity conformation of αIIbß3 as detected by PAC-1 ab, and in control of platelet adhesiveness through Rap1 GTPase protein activation. Importantly, the PAR4 Thr120 variant resulted in the increased rate and magnitude of Rap1 activation. Human platelet PAR4 mediated-activation of αIIbß3 was phospholipase C beta (PLCß)-dependent and unlike mouse platelet PI3K-independent. These data identify a PAR4-dependent inhibitory mechanism for the prasugrel-mediated platelet inhibition, not seen with clopidogrel that could explain the reduction in stent thrombosis documented in clinical trials with prasugrel.


Subject(s)
Antithrombins/therapeutic use , Peptide Fragments/therapeutic use , Percutaneous Coronary Intervention , Platelet Aggregation Inhibitors/therapeutic use , Prasugrel Hydrochloride/therapeutic use , Thrombosis/prevention & control , Aged , Female , Hirudins , Humans , Male , Middle Aged , Percutaneous Coronary Intervention/adverse effects , Receptors, Purinergic P2Y12/metabolism , Receptors, Thrombin/metabolism , Recombinant Proteins/therapeutic use , Stents/adverse effects , Thrombosis/etiology , Thrombosis/metabolism
13.
J Biol Chem ; 294(12): 4401-4411, 2019 03 22.
Article in English | MEDLINE | ID: mdl-30670596

ABSTRACT

Pancreatic ß-cell failure in type 2 diabetes mellitus is a serious challenge that results in an inability of the pancreas to produce sufficient insulin to properly regulate blood glucose levels. Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor expressed by ß-cells that has recently been proposed as a potential target for improving glycemic control and suppressing binge eating behaviors. We discovered that TAAR1 is coupled to Gαs-signaling pathways in insulin-secreting ß-cells to cause protein kinase A (PKA)/exchange protein activated by cAMP (Epac)-dependent release of insulin, activation of RAF proto-oncogene, Ser/Thr kinase (Raf)-mitogen-activated protein kinase (MAPK) signaling, induction of cAMP response element-binding protein (CREB)-insulin receptor substrate 2 (Irs-2), and increased ß-cell proliferation. Interestingly, TAAR1 triggered cAMP-mediated calcium influx and release from internal stores, both of which were required for activation of a MAPK cascade utilizing calmodulin-dependent protein kinase II (CaMKII), Raf, and MAPK/ERK kinase 1/2 (MEK1/2). Together, these data identify TAAR1/Gαs-mediated signaling pathways that promote insulin secretion, improved ß-cell function and proliferation, and highlight TAAR1 as a promising new target for improving ß-cell health in type 2 diabetes mellitus.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Receptors, G-Protein-Coupled/physiology , Signal Transduction/physiology , Adenylyl Cyclases/metabolism , Animals , Calcium/metabolism , Cell Line , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Glucose/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/enzymology , Mice , Phosphorylation , Rats , Receptors, G-Protein-Coupled/agonists
14.
J Invest Dermatol ; 139(2): 412-421, 2019 02.
Article in English | MEDLINE | ID: mdl-30287285

ABSTRACT

PAR2 has been proposed to contribute to lesion formation and intense itch in atopic dermatitis. Here, we tested the ability of a cell-penetrating pepducin, PZ-235, to mitigate the potentially deleterious effects of PAR2 in models of atopic dermatitis. PZ-235 significantly inhibited PAR2-mediated expression of inflammatory factors NF-κB, TSLP, TNF-α, and differentiation marker K10 by 94%-98% (P < 0.001) in human keratinocytes and suppressed IL-4 and IL-13 by 68%-83% (P < 0.05) in mast cells. In delayed pepducin treatment models of oxazolone- and DNFB-induced dermatitis, PZ-235 significantly attenuated skin thickening by 43%-100% (P < 0.01) and leukocyte crusting by 57% (P < 0.05), and it inhibited ex vivo chemotaxis of leukocytes toward PAR2 agonists. Daily PZ-235 treatment of filaggrin-deficient mice exposed to dust mite allergens for 8 weeks significantly suppressed total leukocyte and T-cell infiltration by 50%-68%; epidermal thickness by 60%-77%; and skin thickening, scaling, excoriation, and total lesion severity score by 46%-56%. PZ-235 significantly reduced itching caused by wasp venom peptide degranulation of mast cells in mice by 51% (P < 0.05), which was comparable to the protective effects conferred by PAR2 deficiency. Taken together, these results suggest that a PAR2 pepducin may confer broad therapeutic benefits as a disease-modifying treatment for atopic dermatitis and itch.


Subject(s)
Cell-Penetrating Peptides/pharmacology , Dermatitis, Atopic/drug therapy , Pruritus/drug therapy , Receptor, PAR-2/antagonists & inhibitors , Animals , Cell-Penetrating Peptides/therapeutic use , Dermatitis, Atopic/etiology , Dermatitis, Atopic/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Filaggrin Proteins , Humans , Keratinocytes , Male , Mice , Pruritus/etiology , Pruritus/pathology , Receptor, PAR-2/immunology , Receptor, PAR-2/metabolism
15.
Int J Mol Sci ; 19(8)2018 Jul 31.
Article in English | MEDLINE | ID: mdl-30065181

ABSTRACT

The G-protein coupled receptors (GPCRs) belong to a large family of diverse receptors that are well recognized as pharmacological targets. However, very few of these receptors have been pursued as oncology drug targets. The Protease-activated receptor 1 (PAR1), which is a G-protein coupled receptor, has been shown to act as an oncogene and is an emerging anti-cancer drug target. In this paper, we provide an overview of PAR1's biased signaling role in metastatic cancers of the breast, lungs, and ovaries and describe the development of PAR1 inhibitors that are currently in clinical use to treat acute coronary syndromes. PAR1 inhibitor PZ-128 is in a Phase II clinical trial and is being developed to prevent ischemic and thrombotic complication of patients undergoing cardiac catheterization. PZ-128 belongs to a new class of cell-penetrating, membrane-tethered peptides named pepducins that are based on the intracellular loops of receptors targeting the receptor G-protein interface. Application of PZ-128 as an anti-metastatic and anti-angiogenic therapeutic agent in breast, lung, and ovarian cancer is being reviewed.


Subject(s)
Breast Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Receptor, PAR-1/metabolism , Animals , Breast Neoplasms/metabolism , Cell-Penetrating Peptides/therapeutic use , Female , Humans , Lipopeptides/therapeutic use , Lung Neoplasms/metabolism , Ovarian Neoplasms/metabolism , Receptor, PAR-1/antagonists & inhibitors , Signal Transduction/drug effects
16.
Arterioscler Thromb Vasc Biol ; 38(6): 1368-1380, 2018 06.
Article in English | MEDLINE | ID: mdl-29622563

ABSTRACT

OBJECTIVE: Protease-activated receptor-1 (PAR1) is classically activated by thrombin and is critical in controlling the balance of hemostasis and thrombosis. More recently, it has been shown that noncanonical activation of PAR1 by matrix metalloprotease-1 (MMP1) contributes to arterial thrombosis. However, the role of PAR1 in long-term development of atherosclerosis is unknown, regardless of the protease agonist. APPROACH AND RESULTS: We found that plasma MMP1 was significantly correlated (R=0.33; P=0.0015) with coronary atherosclerotic burden as determined by angiography in 91 patients with coronary artery disease and acute coronary syndrome undergoing cardiac catheterization or percutaneous coronary intervention. A cell-penetrating PAR1 pepducin, PZ-128, currently being tested as an antithrombotic agent in the acute setting in the TRIP-PCI study (Thrombin Receptor Inhibitory Pepducin-Percutaneous Coronary Intervention), caused a significant decrease in total atherosclerotic burden by 58% to 70% (P<0.05) and reduced plaque macrophage content by 54% (P<0.05) in apolipoprotein E-deficient mice. An MMP1 inhibitor gave similar beneficial effects, in contrast to the thrombin inhibitor bivalirudin that gave no improvement on atherosclerosis end points. Mechanistic studies revealed that inflammatory signaling mediated by MMP1-PAR1 plays a critical role in amplifying tumor necrosis factor α signaling in endothelial cells. CONCLUSIONS: These data suggest that targeting the MMP1-PAR1 system may be effective in tamping down chronic inflammatory signaling in plaques and halting the progression of atherosclerosis.


Subject(s)
Aortic Diseases/enzymology , Atherosclerosis/enzymology , Carotid Artery Diseases/enzymology , Coronary Artery Disease/enzymology , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 1/metabolism , Receptor, PAR-1/metabolism , Adult , Aged , Aged, 80 and over , Animals , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Biomarkers/blood , Carotid Artery Diseases/pathology , Carotid Artery Diseases/prevention & control , Cell Line , Cell-Penetrating Peptides/pharmacology , Clinical Trials, Phase II as Topic , Coronary Artery Disease/blood , Coronary Artery Disease/pathology , Disease Models, Animal , Disease Progression , Female , Fibrinolytic Agents/pharmacology , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Hydroxamic Acids/pharmacology , Lipopeptides/pharmacology , Male , Matrix Metalloproteinase 1/blood , Matrix Metalloproteinase Inhibitors/pharmacology , Mice, Inbred C57BL , Mice, Knockout, ApoE , Middle Aged , Multicenter Studies as Topic , Oligopeptides/pharmacology , Plaque, Atherosclerotic , Randomized Controlled Trials as Topic , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-1/blood , Signal Transduction , Tumor Necrosis Factor-alpha/blood , United States
17.
J Biol Chem ; 291(44): 23188-23198, 2016 10 28.
Article in English | MEDLINE | ID: mdl-27613872

ABSTRACT

Chronic liver inflammation and fibrosis in nonalcoholic steatohepatitis can lead to cirrhosis and liver failure for which there are currently no approved treatments. Protease-activated receptor-2 (PAR2) is an emerging new target expressed on liver stellate cells and hepatocytes that regulates the response to liver injury and inflammation. Here, we identified a pepducin to block the deleterious actions of PAR2 in promoting liver fibrosis. Non-alcoholic fatty liver disease and early fibrosis were induced by the methionine-choline-deficient diet in mice. Fibrotic liver disease was induced by administering carbon tetrachloride for 8 weeks. Mice were treated with the pepducin PZ-235 either from onset of the experiment or after fibrosis was established. Hepatic fibrosis, collagen content, inflammatory cytokines, steatosis, triglycerides, and NAFLD activity score were assessed as primary outcome parameters depending on the model. The activity of the PAR2 pepducin on cultured stellate cell activation and hepatocyte reactive oxygen species production was evaluated. PZ-235 significantly suppressed liver fibrosis, collagen deposition, inflammatory cytokines, NAFLD activity score, steatosis, triglycerides, aspartate transaminase, alanine transaminase, and stellate cell proliferation by up to 50-100%. The PAR2 inhibitor afforded significant protective effects against hepatocellular necrosis and attenuated PAR2-mediated reactive oxygen species production in hepatocytes. PZ-235 was distributed to liver and other mouse tissues and was found to form a well structured α-helix that closely resembles the juxtamembrane helical region of the analogous TM6 and third intracellular region of the intact receptor that is critical for coupling to internal G proteins. The ability of PZ-235 to effectively suppress fibrosis, hepatocellular necrosis, reactive oxygen species production, steatosis, and inflammation indicates the potential for PAR2 pepducin inhibitors to be broadly efficacious in the treatment of liver fibrosis.


Subject(s)
Lipopeptides/administration & dosage , Liver Cirrhosis/prevention & control , Receptor, PAR-2/metabolism , Animals , Hepatocytes , Humans , Liver/drug effects , Liver/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Receptor, PAR-2/antagonists & inhibitors , Receptor, PAR-2/genetics
18.
Arterioscler Thromb Vasc Biol ; 36(1): 189-97, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26681756

ABSTRACT

OBJECTIVE: Pepducins are membrane-tethered, cell-penetrating lipopeptides that target the cytoplasmic surface of their cognate receptor. Here, we report the first human use of a protease-activated receptor-1-based pepducin, which is intended as an antiplatelet agent to prevent ischemic complications of percutaneous coronary interventions. APPROACH AND RESULTS: PZ-128 was administered by 1 to 2 hours continuous intravenous infusion (0.01-2 mg/kg) to 31 subjects with coronary artery disease or multiple coronary artery disease risk factors. Safety, antiplatelet efficacy, and pharmacokinetics were assessed at baseline and 0.5, 1, 2, 6, 24 hours, and 7 to 10 days postdosing. The inhibitory effects of PZ-128 on platelet aggregation stimulated by the protease-activated receptor-1 agonist SFLLRN (8 µmol/L) at 30 minutes to 6 hours were dose dependent with 20% to 40% inhibition at 0.3 mg/kg, 40% to 60% at 0.5 mg/kg, and ≥ 80% to 100% at 1 to 2 mg/kg. The subgroup receiving aspirin in the 0.5 and 1-mg/kg dose cohorts had 65% to 100% inhibition of final aggregation to SFLLRN at 30 minutes to 2 hours and 95% to 100% inhibition by 6 hours. The inhibitory effects of 0.5 mg/kg PZ-128 were reversible with 50% recovery of aggregation to SFLLRN by 24 hours. There were no significant effects of PZ-128 on aggregation induced by AYPGKF, ADP, or collagen, indicating that the observed effects were specific to protease-activated receptor-1. The plasma half-life was 1.3 to 1.8 hours, and PZ-128 was nondetectable in urine. There were no effects on bleeding, coagulation, clinical chemistry, or ECG parameters. CONCLUSIONS: PZ-128 is a promising antiplatelet agent that provides rapid, specific, dose dependent, and reversible inhibition of platelet protease-activated receptor-1 through a novel intracellular mechanism. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT01806077.


Subject(s)
Blood Platelets/drug effects , Cell-Penetrating Peptides/administration & dosage , Coronary Artery Disease/therapy , Lipopeptides/administration & dosage , Percutaneous Coronary Intervention , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation/drug effects , Receptor, PAR-1/antagonists & inhibitors , Adult , Aged , Blood Platelets/metabolism , Cell-Penetrating Peptides/adverse effects , Cell-Penetrating Peptides/pharmacokinetics , Coronary Artery Disease/blood , Coronary Artery Disease/diagnosis , Dose-Response Relationship, Drug , Female , Half-Life , Humans , Infusions, Intravenous , Lipopeptides/adverse effects , Lipopeptides/pharmacokinetics , Male , Middle Aged , Percutaneous Coronary Intervention/adverse effects , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Function Tests , Receptor, PAR-1/metabolism , Treatment Outcome
19.
Methods Mol Biol ; 1324: 191-203, 2015.
Article in English | MEDLINE | ID: mdl-26202271

ABSTRACT

Lipopeptides based on the intracellular loops of cell-surface receptors, known as "Pepducins," represent a promising new class of compounds used for the study of membrane proteins and as potential therapeutics in a variety of diseases. Detailed knowledge of the three-dimensional structure of G-protein-coupled receptors (GPCRs) and delineation of the mechanisms of pepducin activation and biased G-protein signaling has facilitated the development of even more potent pepducin allosteric modulators.


Subject(s)
Lipopeptides/metabolism , Lipopeptides/therapeutic use , Receptors, G-Protein-Coupled/metabolism , Allosteric Regulation , Amino Acid Sequence , Animals , Humans , Lipopeptides/chemistry , Lipopeptides/pharmacology , Molecular Sequence Data , Protein Conformation , Receptors, G-Protein-Coupled/chemistry , Signal Transduction
20.
J Biol Chem ; 290(25): 15785-15798, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25934391

ABSTRACT

G protein-coupled receptors (GPCRs) are remarkably versatile signaling systems that are activated by a large number of different agonists on the outside of the cell. However, the inside surface of the receptors that couple to G proteins has not yet been effectively modulated for activity or treatment of diseases. Pepducins are cell-penetrating lipopeptides that have enabled chemical and physical access to the intracellular face of GPCRs. The structure of a third intracellular (i3) loop agonist, pepducin, based on protease-activated receptor-1 (PAR1) was solved by NMR and found to closely resemble the i3 loop structure predicted for the intact receptor in the on-state. Mechanistic studies revealed that the pepducin directly interacts with the intracellular H8 helix region of PAR1 and allosterically activates the receptor through the adjacent (D/N)PXXYYY motif through a dimer-like mechanism. The i3 pepducin enhances PAR1/Gα subunit interactions and induces a conformational change in fluorescently labeled PAR1 in a very similar manner to that induced by thrombin. As pepducins can potentially be made to target any GPCR, these data provide insight into the identification of allosteric modulators to this major drug target class.


Subject(s)
Biomimetic Materials/chemistry , Cell-Penetrating Peptides/chemistry , GTP-Binding Protein alpha Subunits/chemistry , Receptor, PAR-1/chemistry , Allosteric Regulation/genetics , Amino Acid Motifs , Animals , COS Cells , Chlorocebus aethiops , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , HEK293 Cells , Humans , Nuclear Magnetic Resonance, Biomolecular , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...