Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Cell ; 186(24): 5375-5393.e25, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37995657

ABSTRACT

Itch is an unpleasant sensation that evokes a desire to scratch. The skin barrier is constantly exposed to microbes and their products. However, the role of microbes in itch generation is unknown. Here, we show that Staphylococcus aureus, a bacterial pathogen associated with itchy skin diseases, directly activates pruriceptor sensory neurons to drive itch. Epicutaneous S. aureus exposure causes robust itch and scratch-induced damage. By testing multiple isogenic bacterial mutants for virulence factors, we identify the S. aureus serine protease V8 as a critical mediator in evoking spontaneous itch and alloknesis. V8 cleaves proteinase-activated receptor 1 (PAR1) on mouse and human sensory neurons. Targeting PAR1 through genetic deficiency, small interfering RNA (siRNA) knockdown, or pharmacological blockade decreases itch and skin damage caused by V8 and S. aureus exposure. Thus, we identify a mechanism of action for a pruritogenic bacterial factor and demonstrate the potential of inhibiting V8-PAR1 signaling to treat itch.


Subject(s)
Peptide Hydrolases , Pruritus , Receptor, PAR-1 , Staphylococcal Infections , Staphylococcus aureus , Animals , Humans , Mice , Peptide Hydrolases/metabolism , Pruritus/microbiology , Receptor, PAR-1/metabolism , Staphylococcus aureus/enzymology , Staphylococcus aureus/pathogenicity , Staphylococcus aureus/physiology , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology
2.
Osteoarthritis Cartilage ; 32(7): 881-894, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38604493

ABSTRACT

OBJECTIVE: Transient receptor potential vanilloid 4 (TRPV4) is a multi-modally activated cation channel that mediates mechanotransduction pathways by which musculoskeletal tissues respond to mechanical load and regulate tissue health. Using conditional Trpv4 knockout mice, we investigated the role of Trpv4 in regulating intervertebral disc (IVD) health and injury-induced IVD degeneration. METHODS: Col2-Cre;Trpv4fl/f (Trpv4 KO) mice were used to knockout Trpv4 in all type 2 collagen-expressing cells. Effects of gene targeting alone was assessed in lumbar spines, using vertebral bone length measurement, histological, immunohistochemistry and gene expression analyses, and mechanical testing. Disc puncture was performed on caudal IVDs of wild-type (WT) and Trpv4 KO mice at 2.5- and 6.5-months-of-age. Six weeks after puncture (4- and 8-months-of-age at sacrifice), caudal spines were assessed using histological analyses. RESULTS: While loss of Trpv4 did not significantly alter vertebral bone length and tissue histomorphology compared to age-matched WT mice, Trpv4 KO mice showed decreased proteoglycan and PRG4 staining in the annulus fibrosus compared to WT. At the gene level, Trpv4 KO mice showed significantly increased expression of Acan, Bgn, and Prg4 compared to WT. Functionally, loss of Trpv4 was associated with significantly increased neutral zone length in lumbar IVDs. Following puncture, both Trpv4 KO and WT mice showed similar signs of degeneration at the site of injury. Interestingly, loss of Trpv4 prevented mechanically-induced degeneration in IVDs adjacent to sites of injury. CONCLUSION: These studies suggest a role for Trpv4 in regulating extracellular matrix synthesis and mediating the response of IVD tissues to mechanical stress.


Subject(s)
Disease Models, Animal , Extracellular Matrix , Intervertebral Disc Degeneration , Mice, Knockout , TRPV Cation Channels , Animals , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Degeneration/pathology , Mice , Extracellular Matrix/metabolism , Intervertebral Disc/metabolism , Intervertebral Disc/pathology , Lumbar Vertebrae , Weight-Bearing/physiology , Collagen Type II/metabolism , Mechanotransduction, Cellular/physiology , Aggrecans/metabolism , Stress, Mechanical , Proteoglycans/metabolism , Proteoglycans/genetics
3.
Arterioscler Thromb Vasc Biol ; 43(11): 2165-2178, 2023 11.
Article in English | MEDLINE | ID: mdl-37675637

ABSTRACT

BACKGROUND: SR-B1 (scavenger receptor class B type 1)/LDLR (low-density lipoprotein receptor) double knockout mice fed a high-fat, high-cholesterol diet containing cholate exhibit coronary artery disease characterized by occlusive coronary artery atherosclerosis, platelet accumulation in coronary arteries, and myocardial fibrosis. Platelets are involved in atherosclerosis development, and PAR (protease-activated receptor) 4 has a prominent role in platelet function in mice. However, the role of PAR4 on coronary artery disease in mice has not been tested. METHODS: We tested the effects of a PAR4 inhibitory pepducin (RAG8) on diet-induced aortic sinus and coronary artery atherosclerosis, platelet accumulation in atherosclerotic coronary arteries, and myocardial fibrosis in SR-B1/LDLR double knockout mice. SR-B1/LDLR double knockout mice were fed a high-fat, high-cholesterol diet containing cholate and injected daily with 20 mg/kg of either the RAG8 pepducin or a control reverse-sequence pepducin (SRQ8) for 20 days. RESULTS: Platelets from the RAG8-treated mice exhibited reduced thrombin and PAR4 agonist peptide-mediated activation compared with those from control SRQ8-treated mice when tested ex vivo. Although aortic sinus atherosclerosis levels did not differ, RAG8-treated mice exhibited reduced coronary artery atherosclerosis, reduced platelet accumulation in atherosclerotic coronary arteries, and reduced myocardial fibrosis. These protective effects were not accompanied by changes in circulating lipids, inflammatory cytokines, or immune cells. However, RAG8-treated mice exhibited reduced VCAM-1 (vascular cell adhesion molecule 1) protein levels in nonatherosclerotic coronary artery cross sections and reduced leukocyte accumulation in atherosclerotic coronary artery cross sections compared with those from SRQ8-treated mice. CONCLUSIONS: The PAR4 inhibitory RAG8 pepducin reduced coronary artery atherosclerosis and myocardial fibrosis in SR-B1/LDLR double knockout mice fed a high-fat, high-cholesterol diet containing cholate. Furthermore, RAG8 reduced VCAM-1 in nonatherosclerotic coronary arteries and reduced leukocyte and platelet accumulation in atherosclerotic coronary arteries. These findings identify PAR4 as an attractive target in reducing coronary artery disease development, and the use of RAG8 may potentially be beneficial in cardiovascular disease.


Subject(s)
Atherosclerosis , Coronary Artery Disease , Animals , Mice , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Atherosclerosis/metabolism , Cholates , Cholesterol , Coronary Artery Disease/genetics , Coronary Artery Disease/prevention & control , Fibrosis , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/genetics , Receptors, LDL/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
4.
Am J Physiol Cell Physiol ; 325(1): C272-C285, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37273236

ABSTRACT

Proteinase-activated receptors (PARs) are G protein-coupled receptors (GPCRs) activated by limited n-terminal proteolysis. PARs are highly expressed in many cancer cells, including prostate cancer (PCa), and regulate various aspects of tumor growth and metastasis. Specific activators of PARs in different physiological and pathophysiological contexts remain poorly defined. In this study, we examined the androgen-independent human prostatic cancer cell line PC3 and find the functional expression of PAR1 and PAR2, but not PAR4. Using genetically encoded PAR cleavage biosensors, we showed that PC3 cells secrete proteolytic enzymes that cleave PARs and trigger autocrine signaling. CRISPR/Cas9 targeting of PAR1 and PAR2 combined with microarray analysis revealed genes that are regulated through this autocrine signaling mechanism. We found several genes that are known PCa prognostic factors or biomarker to be differentially expressed in the PAR1-knockout (KO) and PAR2-KO PC3 cells. We further examined PAR1 and PAR2 regulation of PCa cell proliferation and migration and found that absence of PAR1 promotes PC3 cell migration and suppresses cell proliferation, whereas PAR2 deficiency showed opposite effects. Overall, these results demonstrate that autocrine signaling through PARs is an important regulator of PCa cell function.


Subject(s)
Prostatic Neoplasms , Receptor, PAR-1 , Male , Humans , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism , PC-3 Cells , Autocrine Communication , Receptor, PAR-2/genetics , Receptor, PAR-2/metabolism , Prostatic Neoplasms/genetics
5.
Br J Dermatol ; 189(3): 279-291, 2023 08 24.
Article in English | MEDLINE | ID: mdl-36652225

ABSTRACT

BACKGROUND: Granzyme K (GzmK) is a serine protease with minimal presence in healthy tissues while abundant in inflamed tissues. Initially thought to play an exclusive role in immune-mediated cell death, extracellular GzmK can also promote inflammation. OBJECTIVES: To evaluate the role of GzmK in the pathogenesis of atopic dermatitis (AD), the most common inflammatory skin disease. METHODS: A panel of human AD and control samples was analysed to determine if GzmK is elevated. Next, to determine a pathological role for GzmK in AD-like skin inflammation, oxazolone-induced dermatitis was induced in GzmK-/- and wild-type (WT) mice. RESULTS: In human lesional AD samples, there was an increase in the number of GzmK+ cells compared with healthy controls. GzmK-/- mice exhibited reduced overall disease severity characterized by reductions in scaling, erosions and erythema. Surprisingly, the presence of GzmK did not notably increase the overall pro-inflammatory response or epidermal barrier permeability in WT mice; rather, GzmK impaired angiogenesis, increased microvascular damage and microhaemorrhage. Mechanistically, GzmK contributed to vessel damage through cleavage of syndecan-1, a key structural component of the glycocalyx, which coats the luminal surface of vascular endothelia. CONCLUSIONS: GzmK may provide a potential therapeutic target for skin conditions associated with persistent inflammation, vasculitis and pathological angiogenesis.


Subject(s)
Dermatitis, Atopic , Granzymes , Animals , Humans , Mice , Dermatitis, Atopic/pathology , Epidermis/metabolism , Granzymes/metabolism , Inflammation , Skin/pathology
6.
J Biol Chem ; 295(8): 2520-2540, 2020 02 21.
Article in English | MEDLINE | ID: mdl-31892516

ABSTRACT

Proteinase-activated receptor (PAR)-4 is a member of the proteolytically-activated PAR family of G-protein-coupled receptors (GPCR) that represents an important target in the development of anti-platelet therapeutics. PARs are activated by proteolytic cleavage of their receptor N terminus by enzymes such as thrombin, trypsin, and cathepsin-G. This reveals the receptor-activating motif, termed the tethered ligand that binds intramolecularly to the receptor and triggers signaling. However, PARs are also activated by exogenous application of synthetic peptides derived from the tethered-ligand sequence. To better understand the molecular basis for PAR4-dependent signaling, we examined PAR4-signaling responses to a peptide library derived from the canonical PAR4-agonist peptide, AYPGKF-NH2, and we monitored activation of the Gαq/11-coupled calcium-signaling pathway, ß-arrestin recruitment, and mitogen-activated protein kinase (MAPK) pathway activation. We identified peptides that are poor activators of PAR4-dependent calcium signaling but were fully competent in recruiting ß-arrestin-1 and -2. Peptides that were unable to stimulate PAR4-dependent calcium signaling could not trigger MAPK activation. Using in silico docking and site-directed mutagenesis, we identified Asp230 in the extracellular loop-2 as being critical for PAR4 activation by both agonist peptide and the tethered ligand. Probing the consequence of biased signaling on platelet activation, we found that a peptide that cannot activate calcium signaling fails to cause platelet aggregation, whereas a peptide that is able to stimulate calcium signaling and is more potent for ß-arrestin recruitment triggered greater levels of platelet aggregation compared with the canonical PAR4 agonist peptide. These findings uncover molecular determinants critical for agonist binding and biased signaling through PAR4.


Subject(s)
Receptors, Thrombin/metabolism , Signal Transduction , Thrombin/metabolism , Alanine/genetics , Amino Acid Substitution , Calcium/metabolism , Calcium Signaling , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , HEK293 Cells , Humans , Isomerism , MAP Kinase Signaling System , Methylation , Molecular Docking Simulation , Mutant Proteins/metabolism , Mutation/genetics , Peptides/metabolism , Phosphorylation , Platelet Aggregation , Receptors, Thrombin/agonists , Structural Homology, Protein , beta-Arrestins/metabolism
7.
Can J Physiol Pharmacol ; 99(3): 255-269, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32846106

ABSTRACT

Platelets are small megakaryocyte-derived, anucleate, disk-like structures that play an outsized role in human health and disease. Both a decrease in the number of platelets and a variety of platelet function disorders result in petechiae or bleeding that can be life threatening. Conversely, the inappropriate activation of platelets, within diseased blood vessels, remains the leading cause of death and morbidity by affecting heart attacks and stroke. The fine balance of the platelet state in healthy individuals is controlled by a number of receptor-mediated signaling pathways that allow the platelet to rapidly respond and maintain haemostasis. G-protein coupled receptors (GPCRs) are particularly important regulators of platelet function. Here we focus on the major platelet-expressed GPCRs and discuss the roles of downstream signaling pathways (e.g., different G-protein subtypes or ß-arrestin) in regulating the different phases of the platelet activation. Further, we consider the potential for selectively targeting signaling pathways that may contribute to platelet responses in disease through development of biased agonists. Such selective targeting of GPCR-mediated signaling pathways by drugs, often referred to as biased signaling, holds promise in delivering therapeutic interventions that do not present significant side effects, especially in finely balanced physiological systems such as platelet activation in haemostasis.


Subject(s)
Blood Platelets/physiology , Receptors, G-Protein-Coupled/physiology , Signal Transduction/physiology , Animals , Humans
8.
Mol Pharmacol ; 97(6): 365-376, 2020 06.
Article in English | MEDLINE | ID: mdl-32234808

ABSTRACT

Proteinase-activated receptors (PARs) are a four-member family of G-protein-coupled receptors that are activated via proteolysis. PAR4 is a member of this family that is cleaved and activated by serine proteinases such as thrombin, trypsin, and cathepsin-G. PAR4 is expressed in a variety of tissues and cell types, including platelets, vascular smooth muscle cells, and neuronal cells. In studying PAR4 signaling and trafficking, we observed dynamic changes in the cell membrane, with spherical membrane protrusions that resemble plasma membrane blebbing. Since nonapoptotic membrane blebbing is now recognized as an important regulator of cell migration, cancer cell invasion, and vesicular content release, we sought to elucidate the signaling pathway downstream of PAR4 activation that leads to such events. Using a combination of pharmacological inhibition and CRISPR/CRISPR-associated protein 9 (Cas9)-mediated gene editing approaches, we establish that PAR4-dependent membrane blebbing occurs independently of the Gα q/11- and Gα i-signaling pathways and is dependent on signaling via the ß-arrestin-1/2 and Ras homolog family member A (RhoA) signaling pathways. Together these studies provide further mechanistic insight into PAR4 regulation of cellular function. SIGNIFICANCE STATEMENT: We find that the thrombin receptor PAR4 triggers cell membrane blebbing in a RhoA-and ß-arrestin-dependent manner. In addition to identifying novel cellular responses mediated by PAR4, these data provide further evidence for biased signaling in PAR4 since membrane blebbing was dependent on some, but not all, signaling pathways activated by PAR4.


Subject(s)
Cell Membrane/metabolism , Cell Membrane/pathology , Receptors, Thrombin/metabolism , beta-Arrestins/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , CRISPR-Cas Systems , Cell Shape , Gene Knockout Techniques , HEK293 Cells , Humans , Muscle, Smooth, Vascular/metabolism , Rats , Rats, Inbred WKY , Receptors, G-Protein-Coupled/metabolism , Receptors, Thrombin/agonists , Signal Transduction
9.
FASEB J ; 32(3): 1236-1249, 2018 03.
Article in English | MEDLINE | ID: mdl-29114087

ABSTRACT

The synthetic supercooling drug, icilin, and its primary receptor target, the cation channel transient receptor potential (TRP) melastatin-8 (TRPM8), have been described as potent negative regulators of inflammation in the colon. The aim of this study was to determine whether the anti-inflammatory action of icilin could potentially be used to treat autoimmune neuroinflammatory disorders, such as multiple sclerosis (MS). During experimental autoimmune encephalomyelitis (EAE)-a CD4+ T cell-driven murine model of MS-we found that both wild-type (WT) and TRPM8-deficient EAE mice were protected from disease progression during icilin treatment, as evidenced by delays in clinical onset and reductions in neuroinflammation. In vitro, icilin potently inhibited the proliferation of murine and human CD4+ T cells, with the peripheral expansion of autoantigen-restricted T cells similarly diminished by the administration of icilin in mice. Attenuation of both TRPM8-/- and TRP ankyrin-1-/- T-cell proliferation by icilin was consistent with the WT phenotype, which suggests a mechanism that is independent of these channels. In addition, icilin treatment altered the expressional profile of activated CD4+ T cells to one that was indicative of restricted effector function and limited neuroinflammatory potential. These findings identify a potent anti-inflammatory role for icilin in lymphocyte-mediated neuroinflammation and highlight clear pleiotropic effects of the compound beyond classic TRP channel activation.-Ewanchuk, B. W., Allan, E. R. O., Warren, A. L., Ramachandran, R., Yates, R. M. The cooling compound icilin attenuates autoimmune neuroinflammation through modulation of the T-cell response.


Subject(s)
Calcium/metabolism , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Inflammation/prevention & control , Pyrimidinones/pharmacology , T-Lymphocytes/immunology , TRPA1 Cation Channel/physiology , TRPM Cation Channels/physiology , Animals , Calcium Channel Agonists/pharmacology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Inflammation/immunology , Inflammation/pathology , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/drug effects
10.
Pharmacol Rev ; 68(4): 1110-1142, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27677721

ABSTRACT

Given that over 2% of the human genome codes for proteolytic enzymes and their inhibitors, it is not surprising that proteinases serve many physiologic-pathophysiological roles. In this context, we provide an overview of proteolytic mechanisms regulating inflammation, with a focus on cell signaling stimulated by the generation of inflammatory peptides; activation of the proteinase-activated receptor (PAR) family of G protein-coupled receptors (GPCR), with a mechanism in common with adhesion-triggered GPCRs (ADGRs); and by proteolytic ion channel regulation. These mechanisms are considered in the much wider context that proteolytic mechanisms serve, including the processing of growth factors and their receptors, the regulation of matrix-integrin signaling, and the generation and release of membrane-tethered receptor ligands. These signaling mechanisms are relevant for inflammatory, neurodegenerative, and cardiovascular diseases as well as for cancer. We propose that the inflammation-triggering proteinases and their proteolytically generated substrates represent attractive therapeutic targets and we discuss appropriate targeting strategies.

11.
Pharmacol Res ; 133: 152-159, 2018 07.
Article in English | MEDLINE | ID: mdl-29787869

ABSTRACT

The aim of this study was to better understand the role of TRPV4 in the regulation of blood vessel dilatation by blood flow and activation of GPCRs. Using pressure myography, the dilator responses to the TRPV4 agonist GSK1016790A and to acetylcholine, were examined in rat cremaster arterioles exposed to either no shear stress or to 200 µl/min flow for 6 min. In control vessels GSK1016709A caused vasodilatation (pEC50 7.73 ±â€¯0.12 M, ΔDmax 97 ±â€¯3%) which was significantly attenuated by the TRPV4 antagonists GSK2193874 (100 nM) (pEC50 6.19 ±â€¯0.11 M, p < 0.05) and HC067047 (300 nM) (pEC50 6.44 ±â€¯0.12 M) and abolished by removal of the endothelium. Shear conditioned arterioles were significantly more sensitive to GSK1016790A (pEC50 8.34 ±â€¯0.11, p < 0.05). Acetylcholine-induced vasodilatation (pEC50 7.02 ±â€¯0.07 M, ΔDmax 93 ±â€¯2%) was not affected by shear forces (pEC50 7.08 ±â€¯0.07 M, ΔDmax 95 ±â€¯1%). The dilator response to acetylcholine was unaffected by the TRPV4 antagonist GSK2193874 in control arterioles (pEC50 7.24 ±â€¯0.07 M, ΔDmax 97 ±â€¯2%). However, in shear treated arterioles, the acetylcholine-response was significantly attenuated by GSK2193874 (pEC50 6.25 ±â€¯0.12 M, p < 0.05) indicating an induced interaction between TRPV4 and muscarinic receptors. TRPV4 antibodies localized TRPV4 to the endothelium and shear stress had no effect on its localisation. Finally, agonist activation of the M3 muscarinic receptor opened TRPV4 in HEK293 cells. We concluded that shear stress increases endothelial TRPV4 agonist sensitivity and links TRPV4 activation to muscarinic receptor mediated endothelium-dependent vasodilatation, providing strong evidence that blood flow modulates downstream signalling from at least one but not all GPCRs expressed in the endothelium.


Subject(s)
Abdominal Muscles/blood supply , Arterioles/physiology , TRPV Cation Channels/physiology , Vasodilation/physiology , Animals , Endothelium, Vascular/physiology , HEK293 Cells , Humans , Leucine/analogs & derivatives , Leucine/pharmacology , Male , Rats, Wistar , Receptor, Muscarinic M3/physiology , Stress, Mechanical , Sulfonamides/pharmacology , TRPV Cation Channels/agonists
12.
Mol Pharmacol ; 91(4): 287-295, 2017 04.
Article in English | MEDLINE | ID: mdl-28126849

ABSTRACT

Thrombin initiates human platelet aggregation by coordinately activating proteinase-activated receptors (PARs) 1 and 4. However, targeting PAR1 with an orthosteric-tethered ligand binding-site antagonist results in bleeding, possibly owing to the important role of PAR1 activation on cells other than platelets. Because of its more restricted tissue expression profile, we have therefore turned to PAR4 as an antiplatelet target. We have identified an intracellular PAR4 C-terminal motif that regulates calcium signaling and ß-arrestin interactions. By disrupting this PAR4 calcium/ß-arrestin signaling process with a novel cell-penetrating peptide, we were able to inhibit both thrombin-triggered platelet aggregation in vitro and clot consolidation in vivo. We suggest that targeting PAR4 represents an attractive alternative to blocking PAR1 for antiplatelet therapy in humans.


Subject(s)
Blood Platelets/metabolism , Receptors, Thrombin/chemistry , Receptors, Thrombin/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Blood Platelets/drug effects , Calcium Signaling/drug effects , Cell-Penetrating Peptides/pharmacology , HEK293 Cells , Humans , MAP Kinase Signaling System/drug effects , Mice , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Transport/drug effects , Structure-Activity Relationship , Thrombosis/pathology , beta-Arrestins/metabolism
13.
Mol Pharmacol ; 89(5): 606-14, 2016 May.
Article in English | MEDLINE | ID: mdl-26957205

ABSTRACT

Thrombin is known to signal to cells by cleaving/activating a G-protein-coupled family of proteinase-activated receptors (PARs). The signaling mechanism involves the proteolytic unmasking of an N-terminal receptor sequence that acts as a tethered receptor-activating ligand. To date, the recognized targets of thrombin cleavage and activation for signaling are PAR1 and PAR4, in which thrombin cleaves at a conserved target arginine to reveal a tethered ligand. PAR2, which like PAR1 is also cleaved at an N-terminal arginine to unmask its tethered ligand, is generally regarded as a target for trypsin but not for thrombin signaling. We now show that thrombin, at concentrations that can be achieved at sites of acute injury or in a tumor microenvironment, can directly activate PAR2 vasorelaxation and signaling, stimulating calcium and mitogen-activated protein kinase responses along with triggeringß-arrestin recruitment. Thus, PAR2 can be added alongside PAR1 and PAR4 to the targets, whereby thrombin can affect tissue function.


Subject(s)
Calcium Signaling , MAP Kinase Signaling System , Receptor, PAR-2/agonists , Thrombin/metabolism , Vasodilation , Amino Acid Substitution , Animals , Aorta , Arrestins/metabolism , Calcium Signaling/drug effects , Cell Line , Endothelium, Vascular/physiology , Humans , In Vitro Techniques , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mutation , Oligopeptides/pharmacology , Peptide Fragments/agonists , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Transport/drug effects , Proteolysis , Rabbits , Receptor, PAR-2/chemistry , Receptor, PAR-2/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Vasodilation/drug effects , beta-Arrestins
14.
Proc Natl Acad Sci U S A ; 110(18): 7476-81, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23596210

ABSTRACT

Transient Receptor Potential Melastatin-8 (TRPM8), a recently identified member of the transient receptor potential (TRP) family of ion channels, is activated by mild cooling and by chemical compounds such as the supercooling agent, icilin. Since cooling, possibly involving TRPM8 stimulation, diminishes injury-induced peripheral inflammation, we hypothesized that TRPM8 activation may also attenuate systemic inflammation. We thus studied the involvement of TRPM8 in regulating colonic inflammation using two mouse models of chemically induced colitis. TRPM8 expression, localized immunohistochemically in transgenic TRPM8(GFP) mouse colon, was up-regulated in both human- and murine-inflamed colon samples, as measured by real-time PCR. Wild-type mice (but not TRPM8-nulls) treated systemically with the TRPM8 agonist, icilin showed an attenuation of chemically induced colitis, as reflected by a decrease in macroscopic and microscopic damage scores, bowel thickness, and myeloperoxidase activity compared with untreated animals. Furthermore, icilin treatment reduced the 2,4,6-trinitrobenzenesulfonic acid-induced increase in levels of inflammatory cytokines and chemokines in the colon. In comparison with wild-type mice, Dextran Sodium Sulfate (DSS)-treated TRPM8 knockout mice showed elevated colonic levels of the inflammatory neuropeptide calcitonin-gene-related peptide, although inflammatory indices were equivalent for both groups. Further, TRPM8 activation by icilin blocked capsaicin-triggered calcitonin-gene-related peptide release from colon tissue ex vivo and blocked capsaicin-triggered calcium signaling in Transient Receptor Potential Vaniloid-1 (TRPV1) and TRPM8 transfected HEK cells. Our data document an anti-inflammatory role for TRPM8 activation, in part due to an inhibiton of neuropeptide release, pointing to a novel therapeutic target for colitis and other inflammatory diseases.


Subject(s)
Colitis/pathology , Colitis/physiopathology , Inflammation/pathology , Inflammation/physiopathology , Ion Channel Gating , TRPM Cation Channels/metabolism , Animals , Calcitonin Gene-Related Peptide/metabolism , Calcium Signaling , Chemokines/metabolism , Colitis/complications , Colitis/drug therapy , Colon/metabolism , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Humans , Inflammation/complications , Inflammation Mediators/metabolism , Mice , Mice, Knockout , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , TRPM Cation Channels/deficiency , TRPM Cation Channels/genetics , TRPV Cation Channels/metabolism , Trinitrobenzenesulfonic Acid
15.
J Biol Chem ; 289(49): 34366-77, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25331954

ABSTRACT

Mucosal biopsies from inflamed colon of inflammatory bowel disease patients exhibit elevated epithelial apoptosis compared with those from healthy individuals, disrupting mucosal homeostasis and perpetuating disease. Therapies that decrease intestinal epithelial apoptosis may, therefore, ameliorate inflammatory bowel disease, but treatments that specifically target apoptotic pathways are lacking. Proteinase-activated receptor-2 (PAR2), a G protein-coupled receptor activated by trypsin-like serine proteinases, is expressed on intestinal epithelial cells and stimulates mitogenic pathways upon activation. We sought to determine whether PAR2 activation and signaling could rescue colonic epithelial (HT-29) cells from apoptosis induced by proapoptotic cytokines that are increased during inflammatory bowel disease. The PAR2 agonists 2-furoyl-LIGRLO (2f-LI), SLIGKV and trypsin all significantly reduced cleavage of caspase-3, -8, and -9, poly(ADP-ribose) polymerase, and the externalization of phosphatidylserine after treatment of cells with IFN-γ and TNF-α. Knockdown of PAR2 with siRNA eliminated the anti-apoptotic effect of 2f-LI and increased the sensitivity of HT-29 cells to cytokine-induced apoptosis. Concurrent inhibition of both MEK1/2 and PI3K was necessary to inhibit PAR2-induced survival. 2f-LI was found to increase phosphorylation and inactivation of pro-apoptotic BAD at Ser(112) and Ser(136) by MEK1/2 and PI3K-dependent signaling, respectively. PAR2 activation also increased the expression of anti-apoptotic MCL-1. Simultaneous knockdown of both BAD and MCL-1 had minimal effects on PAR2-induced survival, whereas single knockdown had no effect. We conclude that PAR2 activation reduces cytokine-induced epithelial apoptosis via concurrent stimulation of MEK1/2 and PI3K but little involvement of MCL-1 and BAD. Our findings represent a novel mechanism whereby serine proteinases facilitate epithelial cell survival and may be important in the context of colonic healing.


Subject(s)
Apoptosis/genetics , Colon/metabolism , Epithelial Cells/metabolism , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Receptor, PAR-2/metabolism , Apoptosis/drug effects , Calcium Signaling , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Colon/cytology , Colon/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Gene Expression Regulation , Homeostasis/drug effects , Humans , Interferon-gamma/pharmacology , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/genetics , Oligopeptides/pharmacology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylserines , Phosphoinositide-3 Kinase Inhibitors , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor, PAR-2/antagonists & inhibitors , Receptor, PAR-2/genetics , Trypsin/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
16.
J Biol Chem ; 288(52): 37319-31, 2013 Dec 27.
Article in English | MEDLINE | ID: mdl-24253040

ABSTRACT

Chronic kidney diseases cause significant morbidity and mortality in the population. During renal injury, kidney-localized proteinases can signal by cleaving and activating proteinase-activated receptor-2 (PAR2), a G-protein-coupled receptor involved in inflammation and fibrosis that is highly expressed in renal tubular cells. Following unilateral ureteric obstruction, PAR2-deficient mice displayed reduced renal tubular injury, fibrosis, collagen synthesis, connective tissue growth factor (CTGF), and α-smooth muscle actin gene expression at 7 days, compared with wild-type controls. In human proximal tubular epithelial cells in vitro, PAR2 stimulation with PAR2-activating peptide (PAR2-AP) alone significantly up-regulated the expression of CTGF, a potent profibrotic cytokine. The induction of CTGF by PAR2-AP was synergistically increased when combined with transforming growth factor-ß (TGF-ß). Consistent with these findings, treating human proximal tubular epithelial cells with PAR2-AP induced Smad2/3 phosphorylation in the canonical TGF-ß signaling pathway. The Smad2 phosphorylation and CTGF induction required signaling via both the TGFß-receptor and EGF receptor suggesting that PAR2 utilizes transactivation mechanisms to initiate fibrogenic signaling. Taken together, our data support the hypothesis that PAR2 synergizes with the TGFß signaling pathway to contribute to renal injury and fibrosis.


Subject(s)
ErbB Receptors/biosynthesis , Kidney Diseases/metabolism , Receptor, PAR-2/metabolism , Receptors, Transforming Growth Factor beta/biosynthesis , Signal Transduction , Transcriptional Activation , Animals , Cells, Cultured , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , ErbB Receptors/genetics , Female , Fibrosis/metabolism , Fibrosis/pathology , Humans , Kidney Diseases/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Mice , Mice, Mutant Strains , Oligopeptides/pharmacology , Receptor, PAR-2/agonists , Receptor, PAR-2/genetics , Receptors, Transforming Growth Factor beta/genetics , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism
17.
J Biol Chem ; 288(46): 32979-90, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24052258

ABSTRACT

Neutrophil proteinases released at sites of inflammation can affect tissue function by either activating or disarming signal transduction mediated by proteinase-activated receptors (PARs). Because PAR1 is expressed at sites where abundant neutrophil infiltration occurs, we hypothesized that neutrophil-derived enzymes might also regulate PAR1 signaling. We report here that both neutrophil elastase and proteinase-3 cleave the human PAR1 N terminus at sites distinct from the thrombin cleavage site. This cleavage results in a disarming of thrombin-activated calcium signaling through PAR1. However, the distinct non-canonical tethered ligands unmasked by neutrophil elastase and proteinase-3, as well as synthetic peptides with sequences derived from these novel exposed tethered ligands, selectively stimulated PAR1-mediated mitogen-activated protein kinase activation. This signaling was blocked by pertussis toxin, implicating a Gαi-triggered signal pathway. We conclude that neutrophil proteinases trigger biased PAR1 signaling and we describe a novel set of tethered ligands that are distinct from the classical tethered ligand revealed by thrombin. We further demonstrate the function of this biased signaling in regulating endothelial cell barrier integrity.


Subject(s)
GTP-Binding Protein alpha Subunits/metabolism , Leukocyte Elastase/metabolism , MAP Kinase Signaling System/physiology , Myeloblastin/metabolism , Receptor, PAR-1/metabolism , Cell Line, Tumor , Enzyme Activation/physiology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Protein alpha Subunits/genetics , HEK293 Cells , Humans , Leukocyte Elastase/genetics , Myeloblastin/genetics , Receptor, PAR-1/genetics , Thrombin/genetics , Thrombin/metabolism
18.
RSC Med Chem ; 15(1): 254-266, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38283230

ABSTRACT

The growth hormone secretagogue receptor (GHSR) is a G protein-coupled receptor which regulates various important physiological and pathophysiological processes in the body such as energy homeostasis, growth hormone secretion and regulation of appetite. As a result, it has been postulated as a potential therapeutic target for the treatment of cancer cachexia and other metabolic disorders, as well as a potential imaging agent target for cancers and cardiovascular diseases. Ghrelin is the primary high affinity endogenous ligand for GHSR and has limited secondary structure in solution, which makes it proteolytically unstable. This inherent instability in ghrelin can be overcome by incorporating helix-inducing staples that stabilize its structure and improve affinity and activity. We present an analysis of different stapling methods at positions 12 and 16 of ghrelin(1-20) analogues with the goal of increasing proteolytic stability and to retain or improve affinity and activity towards the GHSR. Ghrelin(1-20) analogues were modified with a wide range of chemical staples, including a lactam staple, triazole staple, hydrocarbon staple, Glaser staple, and xylene-thioether staple. Once synthesized, the receptor affinity and α-helicity were measured using competitive binding assays and circular dichroism spectroscopy, respectively. Generally, an increase in alpha-helicity using a flexible staple linker led to improved affinity towards GHSR. Ghrelin(1-20) analogues with a lactam, triazole, and hydrocarbon staple resulted in helical analogues with stronger affinity towards GHSR than unstapled ghrelin(1-20), a compound that lacks helical character. Compounds were also investigated for their agonist activity through ß-arrestin 1 & 2 recruitment BRET assays and for their metabolic stability through serum stability analysis.

19.
ChemMedChem ; 19(16): e202400013, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-38648251

ABSTRACT

Metastasis is responsible for about 90 % of cancer deaths. Anti-metastatic drugs, termed as migrastatics, offer a distinctive therapeutic approach to address cancer migration and invasion. However, therapeutic exploitation of metastasis-specific targets remains limited, and the effective prevention and suppression of metastatic cancer continue to be elusive. Lysophosphatidic acid receptor 1 (LPA1) is activated by an endogenous lipid molecule LPA, leading to a diverse array of cellular activities. Previous studies have shown that the LPA/LPA1 axis supports the progression of metastasis for many types of cancer. In this study, we report the synthesis and biological evaluation of fluorine-containing triazole derivatives as potent LPA1 antagonists, offering potential as migrastatic drugs for triple negative breast cancer (TNBC). In particular, compound 12 f, the most potent and highly selective in this series with an IC50 value of 16.0 nM in the cAMP assay and 18.4 nM in the calcium mobilization assay, inhibited cell survival, migration, and invasion in the TNBC cell line. Interestingly, the compound did not induce apoptosis in TNBC cells and demonstrated no cytotoxic effects. These results highlight the potential of LPA1 as a migrastatic target. Consequently, the LPA1 antagonists developed in this study hold promise as potential migrastatic candidates for TNBC.


Subject(s)
Antineoplastic Agents , Cell Movement , Receptors, Lysophosphatidic Acid , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Receptors, Lysophosphatidic Acid/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Movement/drug effects , Structure-Activity Relationship , Molecular Structure , Drug Screening Assays, Antitumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Cell Line, Tumor , Female , Triazoles/chemistry , Triazoles/pharmacology , Triazoles/chemical synthesis
20.
Biochem Cell Biol ; 91(6): 487-97, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24219291

ABSTRACT

Implantation serine proteinase 2 (ISP2), a S1 family serine proteinase, is known for its role in the critical processes of embryo hatching and implantation in the mouse uterus. Native implantation serine proteinases (ISPs) are co-expressed and co-exist as heterodimers in uterine and blastocyst tissues. The ISP1-ISP2 enzyme complex shows trypsin-like substrate specificity. In contrast, we found that ISP2, isolated as a 34 kDa monomer from a Pichia pastoris expression system, exhibited a mixed serine proteolytic substrate specificity, as determined by a phage display peptide cleavage approach and verified by the in vitro cleavage of synthetic peptides. Based upon the peptide sequence substrate selectivity, a database search identified many potential ISP2 targets of physiological relevance, including the proteinase activated receptor 2 (PAR2). The in vitro cleavage studies with PAR2-derived peptides confirmed the mixed substrate specificity of ISP2. Treatment of cell lines expressing proteinase-activated receptors (PARs) 1, 2, and 4 with ISP2 prevented receptor activation by either thrombin (PARs 1 and 4) or trypsin (PAR2). The disarming and silencing of PARs by ISP2 may play a role in successful embryo implantation.


Subject(s)
Peptides/metabolism , Receptor, PAR-1/metabolism , Receptor, PAR-2/metabolism , Serine Endopeptidases/metabolism , Signal Transduction/genetics , Amino Acid Sequence , Animals , Embryo Implantation , Female , Gene Expression , Gene Expression Regulation , Humans , Mice , Molecular Sequence Data , Peptide Library , Peptides/chemistry , Pichia/genetics , Pichia/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proteolysis , Rats , Receptor, PAR-1/genetics , Receptor, PAR-2/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine Endopeptidases/genetics , Thrombin/metabolism , Trypsin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL