Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 63
Filtrer
1.
Circ Heart Fail ; 16(10): e010621, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37477012

RÉSUMÉ

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.


Sujet(s)
Défaillance cardiaque , Souris , Animaux , Récepteur de type PAR-1 , Souris de lignée C57BL , Myocarde/anatomopathologie , Fibrose , Inflammation/anatomopathologie , Modèles animaux de maladie humaine
2.
Hepatology ; 77(5): E111-E112, 2023 May 01.
Article de Anglais | MEDLINE | ID: mdl-36869845
3.
Hepatology ; 76(6): 1778-1793, 2022 12.
Article de Anglais | MEDLINE | ID: mdl-35603482

RÉSUMÉ

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.


Sujet(s)
Diabète expérimental , Insulinorésistance , Stéatose hépatique non alcoolique , Souris , Animaux , Stéatose hépatique non alcoolique/métabolisme , Récepteur de type PAR-2/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Foie/métabolisme , Insuline/métabolisme , Glucose/métabolisme , Souris knockout
4.
Methods Mol Biol ; 2383: 307-333, 2022.
Article de Anglais | MEDLINE | ID: mdl-34766299

RÉSUMÉ

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.


Sujet(s)
Lipopeptides/usage thérapeutique , Animaux , Humains , Récepteurs couplés aux protéines G , Transduction du signal
5.
Arterioscler Thromb Vasc Biol ; 41(5): e265-e279, 2021 05 05.
Article de Anglais | MEDLINE | ID: mdl-33761760
6.
Arterioscler Thromb Vasc Biol ; 41(1): e33-e45, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-33267659

RÉSUMÉ

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.


Sujet(s)
Plaquettes/métabolisme , Thrombose carotidienne/sang , Hémostase , Agrégation plaquettaire , Récepteurs couplés aux protéines G/sang , Acide éicosatétraénoïque-5,8,10,14 hydroxy-12/sang , Animaux , Cellules CHO , Thrombose carotidienne/prévention et contrôle , Cricetulus , Modèles animaux de maladie humaine , Femelle , Fibrinolytiques/pharmacologie , Sous-unités alpha Gi-Go des protéines G/sang , Humains , Mâle , Agrégation plaquettaire/effets des médicaments et des substances chimiques , Antiagrégants plaquettaires/pharmacologie , Récepteurs couplés aux protéines G/antagonistes et inhibiteurs , Récepteurs à la thrombine/sang , Transduction du signal , Thrombine/métabolisme
7.
Arterioscler Thromb Vasc Biol ; 40(12): 2990-3003, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-33028101

RÉSUMÉ

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.


Sujet(s)
Syndrome coronarien aigu/thérapie , Plaquettes/effets des médicaments et des substances chimiques , Cathétérisme cardiaque , Peptides de pénétration cellulaire/administration et posologie , Maladie des artères coronaires/thérapie , Lipopeptides/administration et posologie , Myocarde/anatomopathologie , Intervention coronarienne percutanée , Antiagrégants plaquettaires/administration et posologie , Récepteur de type PAR-1/agonistes , Thrombose/prévention et contrôle , Syndrome coronarien aigu/imagerie diagnostique , Sujet âgé , Plaquettes/métabolisme , Cathétérisme cardiaque/effets indésirables , Cathétérisme cardiaque/instrumentation , Peptides de pénétration cellulaire/effets indésirables , Peptides de pénétration cellulaire/pharmacocinétique , Maladie des artères coronaires/imagerie diagnostique , Méthode en double aveugle , Femelle , Humains , Perfusions veineuses , Lipopeptides/effets indésirables , Lipopeptides/pharmacocinétique , Mâle , Adulte d'âge moyen , Nécrose , Intervention coronarienne percutanée/effets indésirables , Intervention coronarienne percutanée/instrumentation , Antiagrégants plaquettaires/effets indésirables , Antiagrégants plaquettaires/pharmacocinétique , Étude de validation de principe , Études prospectives , Récepteur de type PAR-1/métabolisme , Récidive , Endoprothèses , Thrombose/sang , Thrombose/étiologie , Facteurs temps , Résultat thérapeutique , États-Unis
8.
J Thromb Haemost ; 18(1): 23-35, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31444884

RÉSUMÉ

BACKGROUND: Vorapaxar is indicated with standard antiplatelet therapy (APT) in patients with a history of myocardial infarction (MI) or peripheral arterial disease (PAD). OBJECTIVES: To evaluate the comparative effects of vorapaxar on platelet-fibrin clot characteristics (PFCC), coagulation, inflammation, and platelet and endothelial function during treatment with daily 81 mg aspirin (A), 75 mg clopidogrel (C), both (C + A), or neither. METHODS: Thrombelastography, conventional platelet aggregation (PA), ex vivo endothelial function by ENDOPAT, coagulation, platelet activation/inflammation marked by urinary 11-dehydrothromboxane B2 (UTxB2 ) and safety were determined in patients who were APT naïve (n = 21), on C (n = 8), on A (n = 29), and on A + C (n = 23) during 1 month of vorapaxar therapy and 1 month of offset. RESULTS: Vorapaxar had no effect on PFCC, ADP- or collagen-induced PA, thrombin time, fibrinogen, PT, PTT, von Willebrand factor (vWF), D-dimer, or endothelial function (P > .05 in all groups). Inhibition of SFLLRN (PAR-1 activating peptide)-stimulated PA by vorapaxar was accelerated by A + C at 2 hours (P < .05 versus other groups) with nearly complete inhibition by 30 days that persisted through 30 days after discontinuation in all groups (P < .001). SFLLRN-induced PA during offset was lower in APT patients versus APT-naïve patients (P < .05). Inhibition of UTxB2 was observed in APT-naive patients treated with vorapaxar (P < .05). Minor bleeding was only observed in C-treated patients. CONCLUSION: Vorapaxar had no influence on PFCC measured by thrombelastography, coagulation, or endothelial function irrespective of APT. Inhibition of protease activated receptor (PAR)-1 mediated platelet aggregation by vorapaxar was accelerated by A + C and offset was prolonged by concomitant APT. Vorapaxar-induced anti-inflammatory effects were observed in non-aspirin-treated patients.


Sujet(s)
Lactones , Antiagrégants plaquettaires , Humains , Inflammation/traitement médicamenteux , Pyridines
9.
Mol Metab ; 29: 99-113, 2019 11.
Article de Anglais | MEDLINE | ID: mdl-31668396

RÉSUMÉ

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.


Sujet(s)
Cholestérol/métabolisme , Métabolisme lipidique , Foie/métabolisme , Stéatose hépatique non alcoolique/anatomopathologie , Récepteur de type PAR-2/métabolisme , Adulte , Sujet âgé , Animaux , Cholestérol/sang , Alimentation riche en graisse , Évolution de la maladie , Femelle , Sous-unités alpha Gi-Go des protéines G/métabolisme , Humains , Peroxydation lipidique , Foie/anatomopathologie , Cirrhose du foie/anatomopathologie , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Adulte d'âge moyen , Mitogen-Activated Protein Kinase 8/métabolisme , Mitogen-Activated Protein Kinase 9/métabolisme , Stéatose hépatique non alcoolique/métabolisme , Récepteur de type PAR-2/déficit , Récepteur de type PAR-2/génétique
10.
Thromb Res ; 177: 59-69, 2019 May.
Article de Anglais | MEDLINE | ID: mdl-30851630

RÉSUMÉ

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.


Sujet(s)
Antithrombiniques/usage thérapeutique , Fragments peptidiques/usage thérapeutique , Intervention coronarienne percutanée , Antiagrégants plaquettaires/usage thérapeutique , Chlorhydrate de prasugrel/usage thérapeutique , Thrombose/prévention et contrôle , Sujet âgé , Femelle , Hirudines , Humains , Mâle , Adulte d'âge moyen , Intervention coronarienne percutanée/effets indésirables , Récepteurs purinergiques P2Y12/métabolisme , Récepteurs à la thrombine/métabolisme , Protéines recombinantes/usage thérapeutique , Endoprothèses/effets indésirables , Thrombose/étiologie , Thrombose/métabolisme
11.
J Nutr ; 149(3): 416-421, 2019 03 01.
Article de Anglais | MEDLINE | ID: mdl-30753659

RÉSUMÉ

BACKGROUND: Menaquinone-4 (MK4), a vitamin K metabolite, is converted from phylloquinone through a process that requires intermediates of endogenous cholesterol production. Recent evidence suggests that MK4 is involved in kidney function. OBJECTIVE: The purpose of this study was to determine the effect of atorvastatin treatment on MK4 formation in young and old male mice. METHODS: C57BL/6 male mice (4-mo-old and 20-mo-old) were randomly assigned to either a diet containing 300 mg atorvastatin/kg with 3 mg phylloquinone/kg or a control diet containing 3 mg phylloquinone/kg for 8 wk. During week 8, all mice received deuterium-labeled phylloquinone in the diet. Labeled and unlabeled phylloquinone and MK4 in liver, kidney, brain, and intestine were measured by atmospheric pressure chemical ionization LC/MS. 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase gene expression was quantified by reverse transcriptase-PCR. Tissue MK4 and phylloquinone concentrations were compared between atorvastatin treatment groups with use of general linear models. RESULTS: There was no age-treatment interaction on MK4 tissue concentrations. In atorvastatin-treated mice, total MK4 and percentage of deuterium-labeled MK4 in kidney were both approximately 45% lower compared to values in mice not given atorvastatin (all P < 0.05). MK4 concentrations did not differ between groups in any other tissue measured. CONCLUSION: In male mice, atorvastatin reduced endogenous MK4 formation in the kidney, but not other organs. These observations are consistent with our hypothesis that cholesterol metabolism is involved in the generation of MK4. Further research is needed to understand potential regulatory mechanisms and the unique functions of MK4 in the kidney.


Sujet(s)
Atorvastatine/pharmacologie , Vitamine K2/analogues et dérivés , Animaux , Cholestérol/sang , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Mâle , Souris , Souris de lignée C57BL , Triglycéride/sang , Vitamine K2/métabolisme
12.
J Biol Chem ; 294(12): 4401-4411, 2019 03 22.
Article de Anglais | MEDLINE | ID: mdl-30670596

RÉSUMÉ

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.


Sujet(s)
Diabète de type 2/métabolisme , Cellules à insuline/métabolisme , Récepteurs couplés aux protéines G/physiologie , Transduction du signal/physiologie , Adenylate Cyclase/métabolisme , Animaux , Calcium/métabolisme , Lignée cellulaire , AMP cyclique/métabolisme , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Cyclic AMP-Dependent Protein Kinases/métabolisme , Glucose/pharmacologie , Facteurs d'échange de nucléotides guanyliques/métabolisme , Substrats du récepteur à l'insuline/génétique , Cellules à insuline/effets des médicaments et des substances chimiques , Cellules à insuline/enzymologie , Souris , Phosphorylation , Rats , Récepteurs couplés aux protéines G/agonistes
13.
J Invest Dermatol ; 139(2): 412-421, 2019 02.
Article de Anglais | MEDLINE | ID: mdl-30287285

RÉSUMÉ

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.


Sujet(s)
Peptides de pénétration cellulaire/pharmacologie , Eczéma atopique/traitement médicamenteux , Prurit/traitement médicamenteux , Récepteur de type PAR-2/antagonistes et inhibiteurs , Animaux , Peptides de pénétration cellulaire/usage thérapeutique , Eczéma atopique/étiologie , Eczéma atopique/anatomopathologie , Modèles animaux de maladie humaine , Évaluation préclinique de médicament , Protéines filaggrine , Humains , Kératinocytes , Mâle , Souris , Prurit/étiologie , Prurit/anatomopathologie , Récepteur de type PAR-2/immunologie , Récepteur de type PAR-2/métabolisme
14.
Int J Mol Sci ; 19(8)2018 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-30065181

RÉSUMÉ

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.


Sujet(s)
Tumeurs du sein/traitement médicamenteux , Tumeurs du poumon/traitement médicamenteux , Tumeurs de l'ovaire/traitement médicamenteux , Récepteur de type PAR-1/métabolisme , Animaux , Tumeurs du sein/métabolisme , Peptides de pénétration cellulaire/usage thérapeutique , Femelle , Humains , Lipopeptides/usage thérapeutique , Tumeurs du poumon/métabolisme , Tumeurs de l'ovaire/métabolisme , Récepteur de type PAR-1/antagonistes et inhibiteurs , Transduction du signal/effets des médicaments et des substances chimiques
15.
Arterioscler Thromb Vasc Biol ; 38(6): 1368-1380, 2018 06.
Article de Anglais | MEDLINE | ID: mdl-29622563

RÉSUMÉ

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.


Sujet(s)
Maladies de l'aorte/enzymologie , Athérosclérose/enzymologie , Artériopathies carotidiennes/enzymologie , Maladie des artères coronaires/enzymologie , Matrix Metalloproteinase 13/métabolisme , Matrix metalloproteinase 1/métabolisme , Récepteur de type PAR-1/métabolisme , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Animaux , Maladies de l'aorte/anatomopathologie , Maladies de l'aorte/prévention et contrôle , Athérosclérose/anatomopathologie , Athérosclérose/prévention et contrôle , Marqueurs biologiques/sang , Artériopathies carotidiennes/anatomopathologie , Artériopathies carotidiennes/prévention et contrôle , Lignée cellulaire , Peptides de pénétration cellulaire/pharmacologie , Essais cliniques de phase II comme sujet , Maladie des artères coronaires/sang , Maladie des artères coronaires/anatomopathologie , Modèles animaux de maladie humaine , Évolution de la maladie , Femelle , Fibrinolytiques/pharmacologie , Cellules endothéliales de la veine ombilicale humaine/enzymologie , Humains , Acides hydroxamiques/pharmacologie , Lipopeptides/pharmacologie , Mâle , Matrix metalloproteinase 1/sang , Inhibiteurs de métalloprotéinases matricielles/pharmacologie , Souris de lignée C57BL , Souris invalidées pour les gènes ApoE , Adulte d'âge moyen , Études multicentriques comme sujet , Oligopeptides/pharmacologie , Plaque d'athérosclérose , Essais contrôlés randomisés comme sujet , Récepteur de type PAR-1/antagonistes et inhibiteurs , Récepteur de type PAR-1/sang , Transduction du signal , Facteur de nécrose tumorale alpha/sang , États-Unis
16.
Gut ; 66(5): 930-938, 2017 05.
Article de Anglais | MEDLINE | ID: mdl-26858343

RÉSUMÉ

OBJECTIVE: Alcoholic steatohepatitis is a life-threatening condition with short-term mortality up to 40%. It features hepatic neutrophil infiltration and blood neutrophilia, and may evolve from ethanol-induced breakdown of the enteric barrier and consequent bacteraemia. Signalling through CXCR1/2 G-protein-coupled-receptors (GPCRs), the interleukin (IL)-8 receptors, is critical for the recruitment and activation of neutrophils. We have developed short lipopeptides (pepducins), which inhibit post-ligand GPCR activation precisely targeting individual GPCRs. DESIGN: Experimental alcoholic liver disease was induced by administering alcohol and a Lieber-DeCarli high-fat diet. CXCR1/2 GPCRs were blocked via pepducins either from onset of the experiment or after disease was fully established. Hepatic inflammatory infiltration, hepatocyte lipid accumulation and overall survival were assessed as primary outcome parameters. Neutrophil activation was assessed by myeloperoxidase activity and liver cell damage by aspartate aminotransferase and alanine aminotransferase plasma levels. Chemotaxis assays were performed to identify chemoattractant signals derived from alcohol-exposed hepatocytes. RESULTS: Here, we show that experimental alcoholic liver disease is driven by CXCR1/2-dependent activation of neutrophils. CXCR1/2-specific pepducins not only protected mice from liver inflammation, weight loss and mortality associated with experimental alcoholic liver disease, but therapeutic administration cured disease and prevented further mortality in fully established disease. Hepatic neutrophil infiltration and triglyceride accumulation was abrogated by CXCR1/2 blockade. Moreover, CXCL-1 plasma levels were decreased with the pepducin therapy as was the transcription of hepatic IL-1ß mRNA. CONCLUSIONS: We propose that high circulating IL-8 in human alcoholic hepatitis may cause pathogenic overzealous neutrophil activation, and therapeutic blockade via pepducins merits clinical study.


Sujet(s)
Stéatose hépatique alcoolique/traitement médicamenteux , Lipopeptides/pharmacologie , Récepteurs à l'interleukine-8A/antagonistes et inhibiteurs , Récepteurs à l'interleukine-8B/antagonistes et inhibiteurs , Transduction du signal/effets des médicaments et des substances chimiques , Alanine transaminase/sang , Animaux , Aspartate aminotransferases/sang , Caspase-1/métabolisme , Chimiokine CXCL1/génétique , Chimiotaxie/effets des médicaments et des substances chimiques , Stéatose hépatique alcoolique/complications , Stéatose hépatique alcoolique/métabolisme , Stéatose hépatique alcoolique/anatomopathologie , Femelle , Cellules HepG2 , Hépatite/étiologie , Hépatite/prévention et contrôle , Hépatocytes/métabolisme , Humains , Interleukine-1 bêta/génétique , Métabolisme lipidique/effets des médicaments et des substances chimiques , Lipopeptides/usage thérapeutique , Souris , Souris de lignée C57BL , Granulocytes neutrophiles/physiologie , Myeloperoxidase/métabolisme , Récepteurs à l'interleukine-8A/sang , Taux de survie , Transcription génétique/effets des médicaments et des substances chimiques , Triglycéride/métabolisme , Facteur de nécrose tumorale alpha/génétique , Perte de poids/effets des médicaments et des substances chimiques
17.
J Biol Chem ; 291(44): 23188-23198, 2016 10 28.
Article de Anglais | MEDLINE | ID: mdl-27613872

RÉSUMÉ

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.


Sujet(s)
Lipopeptides/administration et posologie , Cirrhose du foie/prévention et contrôle , Récepteur de type PAR-2/métabolisme , Animaux , Hépatocytes , Humains , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Cirrhose du foie/génétique , Cirrhose du foie/métabolisme , Mâle , Souris , Souris de lignée C57BL , Espèces réactives de l'oxygène/métabolisme , Récepteur de type PAR-2/antagonistes et inhibiteurs , Récepteur de type PAR-2/génétique
18.
J Nutr ; 146(8): 1521-7, 2016 08.
Article de Anglais | MEDLINE | ID: mdl-27385762

RÉSUMÉ

BACKGROUND: There has been limited characterization of biological variables that impact vitamin K metabolism. This gap in knowledge can limit the translation of data obtained from preclinical animal studies to future human studies. OBJECTIVE: The purpose of this study was to determine the effects of diet, sex, and housing on serum, tissue, and fecal vitamin K concentrations and gene expression in C57BL6 mice during dietary vitamin K manipulation. METHODS: C57BL6 4-mo-old male and female mice were randomly assigned to conventional or suspended-wire cages and fed control [1400 ± 80 µg phylloquinone (PK)/kg] or deficient (31 ± 0.45 µg PK/kg) diets for 28 d in a factorial design. PK and menaquinone (MK) 4 plasma and tissue concentrations were measured by HPLC. Long-chain MKs were measured in all matrices by LC-atmospheric pressure chemical ionization-mass spectrometry. Gene expression was quantified by reverse transcriptase-polymerase chain reaction in the liver, brain, kidney, pancreas, and adipose tissue. RESULTS: Male and female mice responded differently to dietary manipulation in a tissue-dependent manner. In mice fed the control diet, females had ∼3-fold more MK4 in the brain and mesenteric adipose tissue than did males and 100% greater PK concentrations in the liver, kidney, and mesenteric adipose tissue than did males. In mice fed the deficient diet, kidney MK4 concentrations were ∼4-fold greater in females than in males, and there were no differences in other tissues. Males and females differed in the expression of vitamin K expoxide reductase complex 1 (Vkorc1) in mesenteric adipose tissue and the pancreas and ubiA domain-containing protein 1 (Ubiad1) in the kidney and brain. There was no effect of housing on serum, tissue, or fecal concentrations of any vitamin K form. CONCLUSIONS: Vitamin K concentrations and expression of key metabolic enzymes differ between male and female mice and in response to the dietary PK concentration. Identifying factors that may impact study design and outcomes of interest is critical to optimize study parameters examining vitamin K metabolism in animal models.


Sujet(s)
Tissu adipeux/métabolisme , Encéphale/métabolisme , Régime alimentaire , Rein/métabolisme , Foie/métabolisme , Pancréas/métabolisme , Vitamine K/métabolisme , Tissu adipeux/enzymologie , Animaux , Dimethylallyltransferase/métabolisme , Femelle , Logement , Hébergement animal , Mâle , Protéines membranaires/métabolisme , Mésentère/enzymologie , Mésentère/métabolisme , Souris de lignée C57BL , Pancréas/enzymologie , Facteurs sexuels , Distribution tissulaire , Vitamine K/administration et posologie , Phytoménadione/administration et posologie , Phytoménadione/métabolisme , Vitamine K2/métabolisme , Carence en vitamine K/enzymologie , Carence en vitamine K/métabolisme , Vitamin K epoxide reductases/métabolisme
19.
Arterioscler Thromb Vasc Biol ; 36(1): 189-97, 2016 Jan.
Article de Anglais | MEDLINE | ID: mdl-26681756

RÉSUMÉ

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.


Sujet(s)
Plaquettes/effets des médicaments et des substances chimiques , Peptides de pénétration cellulaire/administration et posologie , Maladie des artères coronaires/thérapie , Lipopeptides/administration et posologie , Intervention coronarienne percutanée , Antiagrégants plaquettaires/administration et posologie , Agrégation plaquettaire/effets des médicaments et des substances chimiques , Récepteur de type PAR-1/antagonistes et inhibiteurs , Adulte , Sujet âgé , Plaquettes/métabolisme , Peptides de pénétration cellulaire/effets indésirables , Peptides de pénétration cellulaire/pharmacocinétique , Maladie des artères coronaires/sang , Maladie des artères coronaires/diagnostic , Relation dose-effet des médicaments , Femelle , Période , Humains , Perfusions veineuses , Lipopeptides/effets indésirables , Lipopeptides/pharmacocinétique , Mâle , Adulte d'âge moyen , Intervention coronarienne percutanée/effets indésirables , Antiagrégants plaquettaires/effets indésirables , Antiagrégants plaquettaires/pharmacocinétique , Tests fonctionnels plaquettaires , Récepteur de type PAR-1/métabolisme , Résultat thérapeutique
20.
Methods Mol Biol ; 1324: 191-203, 2015.
Article de Anglais | MEDLINE | ID: mdl-26202271

RÉSUMÉ

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.


Sujet(s)
Lipopeptides/métabolisme , Lipopeptides/usage thérapeutique , Récepteurs couplés aux protéines G/métabolisme , Régulation allostérique , Séquence d'acides aminés , Animaux , Humains , Lipopeptides/composition chimique , Lipopeptides/pharmacologie , Données de séquences moléculaires , Conformation des protéines , Récepteurs couplés aux protéines G/composition chimique , Transduction du signal
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE