41 results on '"Peishen Zhao"'
Search Results
2. The roles of RGS proteins in cardiometabolic disease
- Author
-
Samantha M. McNeill and Peishen Zhao
- Subjects
Pharmacology - Published
- 2023
- Full Text
- View/download PDF
3. Understanding VPAC receptor family peptide binding and selectivity
- Author
-
Sarah J. Piper, Giuseppe Deganutti, Jessica Lu, Peishen Zhao, Yi-Lynn Liang, Yao Lu, Madeleine M. Fletcher, Mohammed Akhter Hossain, Arthur Christopoulos, Christopher A. Reynolds, Radostin Danev, Patrick M. Sexton, and Denise Wootten
- Subjects
Multidisciplinary ,Pituitary Adenylate Cyclase-Activating Polypeptide ,General Physics and Astronomy ,General Chemistry ,Molecular Dynamics Simulation ,General Biochemistry, Genetics and Molecular Biology ,Vasoactive Intestinal Peptide ,Protein Binding - Abstract
The vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide (PACAP) receptors are key regulators of neurological processes. Despite recent structural data, a comprehensive understanding of peptide binding and selectivity among different subfamily receptors is lacking. Here, we determine structures of active, Gs-coupled, VIP-VPAC1R, PACAP27-VPAC1R, and PACAP27-PAC1R complexes. Cryo-EM structural analyses and molecular dynamics simulations (MDSs) reveal fewer stable interactions between VPAC1R and VIP than for PACAP27, more extensive dynamics of VIP interaction with extracellular loop 3, and receptor-dependent differences in interactions of conserved N-terminal peptide residues with the receptor core. MD of VIP modelled into PAC1R predicts more transient VIP-PAC1R interactions in the receptor core, compared to VIP-VPAC1R, which may underlie the selectivity of VIP for VPAC1R over PAC1R. Collectively, our work improves molecular understanding of peptide engagement with the PAC1R and VPAC1R that may benefit the development of novel selective agonists.
- Published
- 2022
- Full Text
- View/download PDF
4. Targeting VIP and PACAP Receptor Signaling: New Insights into Designing Drugs for the PACAP Subfamily of Receptors
- Author
-
Jessica Lu, Sarah J. Piper, Peishen Zhao, Laurence J. Miller, Denise Wootten, and Patrick M. Sexton
- Subjects
Receptors, Vasoactive Intestinal Polypeptide, Type I ,Organic Chemistry ,General Medicine ,Catalysis ,Computer Science Applications ,Inorganic Chemistry ,Pituitary Adenylate Cyclase-Activating Polypeptide ,Receptors, Vasoactive Intestinal Peptide, Type II ,Amino Acid Sequence ,Physical and Theoretical Chemistry ,Molecular Biology ,Spectroscopy ,Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I ,Signal Transduction ,Vasoactive Intestinal Peptide - Abstract
Pituitary Adenylate Cyclase-Activating Peptide (PACAP) and Vasoactive Intestinal Peptide (VIP) are neuropeptides involved in a diverse array of physiological and pathological processes through activating the PACAP subfamily of class B1 G protein-coupled receptors (GPCRs): VIP receptor 1 (VPAC1R), VIP receptor 2 (VPAC2R), and PACAP type I receptor (PAC1R). VIP and PACAP share nearly 70% amino acid sequence identity, while their receptors PAC1R, VPAC1R, and VPAC2R share 60% homology in the transmembrane regions of the receptor. PACAP binds with high affinity to all three receptors, while VIP binds with high affinity to VPAC1R and VPAC2R, and has a thousand-fold lower affinity for PAC1R compared to PACAP. Due to the wide distribution of VIP and PACAP receptors in the body, potential therapeutic applications of drugs targeting these receptors, as well as expected undesired side effects, are numerous. Designing selective therapeutics targeting these receptors remains challenging due to their structural similarities. This review discusses recent discoveries on the molecular mechanisms involved in the selectivity and signaling of the PACAP subfamily of receptors, and future considerations for therapeutic targeting.
- Published
- 2022
5. Implications of ligand-receptor binding kinetics on GLP-1R signalling
- Author
-
Peishen Zhao, Tin T. Truong, Jon Merlin, Patrick M. Sexton, and Denise Wootten
- Subjects
Pharmacology ,Kinetics ,Diabetes Mellitus, Type 2 ,Glucagon-Like Peptide 1 ,Humans ,Ligands ,Peptides ,Biochemistry ,Glucagon-Like Peptide-1 Receptor ,Receptors, G-Protein-Coupled - Abstract
G protein-coupled receptors (GPCRs) are the largest class of membrane proteins and in recent years there has been a growing appreciation of the importance in understanding temporal aspects of GPCR behaviour, including the kinetics of ligand binding and downstream receptor mediated signalling. Class B1 GPCRs are activated by peptide agonists and are validated therapeutic targets for numerous diseases. However, the kinetics of ligand binding and how this is linked to downstream activation of signalling cascades is not routinely assessed in development of peptide agonists for this receptor class. The glucagon-like peptide-1 receptor (GLP-1R) is a prototypical class B1 GPCR and a validated target for treatment of global health burdens, including type 2 diabetes and obesity. In this study we examined the kinetics of different steps in GLP-1R activation and subsequent cAMP production mediated by a series of GLP-1R peptide agonists, including the ligand-receptor interaction, ligand-receptor-mediated G protein engagement and conformational change and cAMP production. Our results revealed GLP-1R peptide agonist dissociation kinetics (K
- Published
- 2022
6. Chemical Synthesis and Characterization of a Nonfibrillating Glycoglucagon
- Author
-
Briony E. Forbes, Chaitra Chandrashekar, Mengjie Liu, Mohammed Akhter Hossain, Yuji Nishiuchi, Yasuhiro Kajihara, Peishen Zhao, John D. Wade, Wenyi Li, Hemayet Uddin, Feng Lin, and Denise Wootten
- Subjects
Pharmacology ,Agonist ,chemistry.chemical_classification ,Chemistry ,medicine.drug_class ,Organic Chemistry ,Biomedical Engineering ,Pharmaceutical Science ,Bioengineering ,Glucagon ,Chemical synthesis ,Combinatorial chemistry ,Hypoglycemia ,Residue (chemistry) ,medicine ,Thiol ,Moiety ,Humans ,Cysteine ,Glucagon receptor ,Biotechnology - Abstract
The current commercially available glucagon formulations for the treatment of severe hypoglycemia must be reconstituted immediately prior to use, owing to the susceptibility of glucagon to fibrillation and aggregation in an aqueous solution. This results in the inconvenience of handling, misuse, and wastage of this drug. To address these issues, we synthesized a glycosylated glucagon analogue in which the 25th residue (Trp) was replaced with a cysteine (Cys) and a Br-disialyloligosaccharide was conjugated at the Cys thiol moiety. The resulting analogue, glycoglucagon, is a highly potent full agonist at the glucagon receptor. Importantly, glycoglucagon exhibits markedly reduced propensity for fibrillation and enhanced thermal and metabolic stability. This novel analogue is thus a valuable lead for producing stable liquid glucagon formulations that will improve patient compliance and minimize wastage.
- Published
- 2021
7. Granzyme K initiates IL-6 and IL-8 release from epithelial cells by activating protease-activated receptor 2
- Author
-
Dion Kaiserman, Peishen Zhao, Caitlin Lorraine Rowe, Andrea Leong, Nicholas Barlow, Lars Thomas Joeckel, Corinne Hitchen, Sarah Elizabeth Stewart, Morley D. Hollenberg, Nigel Bunnett, Andreas Suhrbier, and Phillip Ian Bird
- Subjects
Multidisciplinary ,Interleukin-6 ,Endopeptidases ,Interleukin-8 ,Endothelial Cells ,Receptor, PAR-2 ,Epithelial Cells ,Receptor, PAR-1 ,Granzymes - Abstract
Granzyme K (GzmK) is a tryptic member of the granzyme family of chymotrypsin-like serine proteases produced by cells of the immune system. Previous studies have indicated that GzmK activates protease-activated receptor 1 (PAR1) enhancing activation of monocytes and wound healing in endothelial cells. Here, we show using peptides and full length proteins that GzmK and, to a lesser extent the related protease GzmA, are capable of activating PAR1 and PAR2. These cleavage events occur at the canonical arginine P1 residue and involve exosite interactions between protease and receptor. Despite cleaving PAR2 at the same point as trypsin, GzmK does not induce a classical Ca2+flux but instead activates a distinct signalling cascade, involving recruitment of β-arrestin and phosphorylation of ERK. In epithelial A549 cells, PAR2 activation by GzmK results in the release of inflammatory cytokines IL-6 and IL-8. These data suggest that during an immune response GzmK acts as a pro-inflammatory regulator, rather than as a cytotoxin.
- Published
- 2022
- Full Text
- View/download PDF
8. RAMPs regulate signalling bias and internalisation of the GIPR
- Author
-
Kathleen M. Caron, Duncan I. Mackie, Asuka Inoue, Denise Wootten, John B. Pawlak, Mark Soave, Matthew Harris, Ho Yan Yeung, Suleiman Al-Sabah, Matthew T. Harper, Stephen J. Briddon, Graham Ladds, Peishen Zhao, Sabrina Carvalho, Dewi Safitri, Patrick M. Sexton, David R. Poyner, Sarah J Routledge, Stephen J. Hill, Bashaier Al-Zaid, and Tin T. Truong
- Subjects
Gastric inhibitory polypeptide ,medicine.anatomical_structure ,Chemistry ,G protein ,RAMP1 ,Cell ,Knockout mouse ,medicine ,Phosphorylation ,Receptor ,Cell biology ,G protein-coupled receptor - Abstract
Gastric inhibitory polypeptide (GIP) receptor is a class B1 GPCR, that responds to GIP and physiologically potentiates glucose-stimulated insulin secretion. Like most class B1 GPCRs, GIPR has been shown to interact with RAMPs, yet the effects of RAMPs on its signalling and trafficking remain poorly understood. We demonstrate that RAMPs modulate G protein activation and GIPR internalisation profiles. RAMP3 reduced GIPR Gsactivation and cAMP production but retained GIPR at the cell surface, and this was associated with prolonged ERK1/2 phosphorylation and β-arrestin association. By contrast, RAMP1/2 reduced Gq/11/15activation of the GIPR. Through knockout mice studies, we show that RAMP1 is important to the normal physiological functioning of GIPR to regulate blood glucose levels. Thus, RAMPs act on G protein/β-arrestin complexes, having both acute and chronic effects on GIPR function, while this study also raises the possibility of a more general role of RAMP3 to enhance GPCR plasma membrane localisation.
- Published
- 2021
- Full Text
- View/download PDF
9. EZH2 Abundance Regulated by UHRF1/UBE2L6/UBR4 Ubiquitin System is the Potential Therapeutic Target to Trigger Pigmented Phenotype in Melanoma
- Author
-
Ralf B. Schittenhelm, Pacman Szeto, Samar Masoumi Moghaddam, Nicholas C. Wong, Cheung J, Malaka Ameratunga, Gamze Kuser Abali, Peishen Zhao, Isobel Leece, Fumihito Noguchi, Mark Shackleton, Miles Andrews, Chun-Teng Huang, and Youfang Zhang
- Subjects
Melanin ,Ubiquitin ,biology ,Downregulation and upregulation ,Cell growth ,EZH2 ,DNA methylation ,biology.protein ,Gene silencing ,Repressor ,macromolecular substances ,Cell biology - Abstract
Cellular heterogeneity in cancer is linked to disease progression and therapy response, although the mechanisms regulating distinct cellular states within tumours are not well understood. To address this, we identified melanin pigment content as a major source of phenotypic and functional heterogeneity in melanoma and compared RNAseq data from high (HPC) and low pigmented melanoma cells (LPC), revealing the polycomb repressor complex protein, EZH2, as a master regulator of these states. EZH2 protein, but not RNA expression, was found to be upregulated in LPCs and inversely correlated with melanin in pigmented patient melanomas. Surprisingly, conventional EZH2 methyltransferase inhibitors, GSK126 and EPZ6438, had no effect on LPC survival, clonogenicity and pigmentation, despite fully inhibiting methyltransferase activity. In contrast, EZH2 silencing by siRNA strategy or DZNep, MS1943 that reduces EZH2 protein levels, significantly inhibited cell growth in LPCs by hampering ribosome biogenesis. In addition, decline in EZH2 protein level induces pigmented cell phenotype by inducing melanin biosynthesis. Proteasomal inhibitor, MG132 treatment induced EZH2 protein levels in HPCs prompted us to look for differentially regulated ubiquitin system proteins in HPC vs LPCs. UBE2L6, E2 conjugating enzyme has been shown to be downregulated significantly in LPCs by UHRF1-mediated CpG methylation. Both biochemical assays and animal studies demonstrated that UBE2L6 expression decline, in turn, promotes EZH2 protein stability due to lack of ubiquitination on K381 residue in LPCs. UBR4 cooperates with UBE2L6 to facilitate this ubiquitination process. Targeting UHRF1/UBE2L6/UBR4 axis can be a better treatment option to trigger HPC state in melanoma in which conventional EZH2 inhibitors are ineffective.
- Published
- 2021
- Full Text
- View/download PDF
10. Structural and Functional Diversity among Agonist-Bound States of the GLP-1 Receptor
- Author
-
Patrick M. Sexton, Peishen Zhao, Brian P. Cary, Samuel H. Gellman, Tin T. Truong, Sarah J. Piper, Radostin Danev, Matthew J. Belousoff, and Denise Wootten
- Subjects
chemistry.chemical_classification ,Agonist ,Gs alpha subunit ,chemistry ,medicine.drug_class ,Biophysics ,medicine ,Peptide ,Receptor ,Conformational isomerism ,Function (biology) ,Glucagon-like peptide 1 receptor ,G protein-coupled receptor - Abstract
Recent advances in G protein-coupled receptor (GPCR) structural elucidation have strengthened previous hypotheses that multi-dimensional signal propagation mediated by these receptors is, in part, dependent on their conformational mobility. However, the relationship between receptor function and static structures determined via crystallography or cryo-electron microscopy is not always clear. This study examines the contribution of peptide agonist conformational plasticity to activation of the glucagon-like peptide-1 receptor (GLP-1R), an important clinical target. We employ variants of the peptides GLP-1 and exendin-4 to explore the interplay between helical propensity near the agonist N-terminus and the ability to bind to and activate the receptor. Cryo-EM analysis of a complex involving an exendin-4 analogue, the GLP-1R and Gs protein revealed two receptor conformers with distinct modes of peptide-receptor engagement. Our functional and structural data suggest that receptor conformational dynamics associated with flexibility of the peptide N-terminal activation domain may be a key determinant of agonist efficacy.
- Published
- 2021
- Full Text
- View/download PDF
11. AM833 Is a Novel Agonist of Calcitonin Family G Protein-Coupled Receptors: Pharmacological Comparison with Six Selective and Nonselective Agonists
- Author
-
Thomas Kruse, Patrick M. Sexton, Trine R. Clausen, Grace Mennen, Tin T. Truong, Denise Wootten, Peter Keov, Peishen Zhao, Sebastian Gb Furness, Madeleine M. Fletcher, and Caroline A. Hick
- Subjects
0301 basic medicine ,Agonist ,Calcitonin ,Male ,medicine.drug_class ,Amylin ,Peptide ,Pharmacology ,03 medical and health sciences ,0302 clinical medicine ,Medicine ,Animals ,Calcitonin receptor ,Protein Precursors ,Receptor ,G protein-coupled receptor ,chemistry.chemical_classification ,business.industry ,Receptors, Calcitonin ,Pramlintide ,Rats ,030104 developmental biology ,chemistry ,Molecular Medicine ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Obesity and associated comorbidities are a major health burden, and novel therapeutics to help treat obesity are urgently needed. There is increasing evidence that targeting the amylin receptors (AMYRs), heterodimers of the calcitonin G protein-coupled receptor (CTR) and receptor activity-modifying proteins, improves weight control and has the potential to act additively with other treatments such as glucagon-like peptide-1 receptor agonists. Recent data indicate that AMYR agonists, which can also independently activate the CTR, may have improved efficacy for treating obesity, even though selective activation of CTRs is not efficacious. AM833 (cagrilintide) is a novel lipidated amylin analog that is undergoing clinical trials as a nonselective AMYR and CTR agonist. In the current study, we have investigated the pharmacology of AM833 across 25 endpoints and compared this peptide with AMYR selective and nonselective lipidated analogs (AM1213 and AM1784), and the clinically used peptide agonists pramlintide (AMYR selective) and salmon CT (nonselective). We also profiled human CT and rat amylin as prototypical selective agonists of CTR and AMYRs, respectively. Our results demonstrate that AM833 has a unique pharmacological profile across diverse measures of receptor binding, activation, and regulation. SIGNIFICANCE STATEMENT: AM833 is a novel nonselective agonist of calcitonin family receptors that has demonstrated efficacy for the treatment of obesity in phase 2 clinical trials. This study demonstrates that AM833 has a unique pharmacological profile across diverse measures of receptor binding, activation, and regulation when compared with other selective and nonselective calcitonin receptor and amylin receptor agonists. The present data provide mechanistic insight into the actions of AM833.
- Published
- 2021
12. Evaluation of biased agonism mediated by dual agonists of the GLP-1 and glucagon receptors
- Author
-
Patrick M. Sexton, Denise Wootten, Elita Yuliantie, Rulue Chang, Dehua Yang, Peishen Zhao, Ming-Wei Wang, Sanaz Darbalaei, and Antao Dai
- Subjects
0301 basic medicine ,Gs alpha subunit ,Drug Agonism ,Peptide ,Pharmacology ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Glucagon-Like Peptide 1 ,Diabetes mellitus ,Functional selectivity ,medicine ,Receptors, Glucagon ,Humans ,Amino Acid Sequence ,Receptor ,Binding selectivity ,G protein-coupled receptor ,chemistry.chemical_classification ,Dose-Response Relationship, Drug ,Chemistry ,medicine.disease ,Peptide Fragments ,030104 developmental biology ,HEK293 Cells ,Oxyntomodulin ,030220 oncology & carcinogenesis ,Peptides ,Glucagon receptor ,Protein Binding - Abstract
Metabolic diseases such as obesity, diabetes, and their comorbidities have converged as one of the most serious health concerns on a global scale. Selective glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) agonists are one of the major therapeutics for type 2 diabetes and obesity. Polypharmacological approaches that enable modulation of multiple metabolic targets in a single drug have emerged as a potential avenue to improve therapeutic outcomes. Among numerous peptides under development are those targeting the GLP-1R and either the glucagon receptor (GCGR), glucose-dependent insulinotropic peptide receptor (GIPR) or all 3 receptors, as dual- or tri- peptide agonists. Despite many of them entering into clinical trials, current development has been based on only a limited understanding of the spectrum of potential pharmacological properties of these ligands beyond binding selectivity. In the present study, we examined the potential for agonists that target both GLP-1R and GCGR to exhibit biased agonism, comparing activity across proximal activation of Gs protein, cAMP accumulation, pERK1/2 and β-arrestin recruitment. Three distinct dual agonists that have different relative cAMP production potency for GLP-1R versus GCGR, "peptide 15", MEDI0382 and SAR425899, and one triagonist of the GLP-1R, GCGR and GIPR were examined. We demonstrated that all novel peptides have distinct biased agonism profiles relative to either of the cognate agonists of the receptors, and to each other. This is an important feature of the pharmacology of this drug class that needs to be considered alongside selectivity, bioavailability and pharmacokinetics for rational optimization of new therapeutics.
- Published
- 2020
13. Pharmacological characterization of mono-, dual- and tri-peptidic agonists at GIP and GLP-1 receptors
- Author
-
Elita Yuliantie, Peishen Zhao, Patrick M. Sexton, Ming-Wei Wang, Antao Dai, Sanaz Darbalaei, Denise Wootten, and Dehua Yang
- Subjects
0301 basic medicine ,Agonist ,endocrine system ,medicine.drug_class ,Adipose tissue ,Endogeny ,Gastric Inhibitory Polypeptide ,Pharmacology ,Biochemistry ,Glucagon-Like Peptide-1 Receptor ,Receptors, Gastrointestinal Hormone ,Iodine Radioisotopes ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Functional selectivity ,Chromogranins ,Cyclic AMP ,GTP-Binding Protein alpha Subunits, Gs ,Humans ,Phosphorylation ,Receptor ,beta-Arrestins ,G protein-coupled receptor ,Mitogen-Activated Protein Kinase 1 ,Mitogen-Activated Protein Kinase 3 ,Chemistry ,digestive, oral, and skin physiology ,HEK 293 cells ,Peptide Fragments ,030104 developmental biology ,HEK293 Cells ,030220 oncology & carcinogenesis ,Signal transduction ,Peptides ,hormones, hormone substitutes, and hormone antagonists - Abstract
Glucose-dependent insulinotropic peptide (GIP) is an incretin hormone with physiological roles in adipose tissue, the central nervous system and bone metabolism. While selective ligands for GIP receptor (GIPR) have not been advanced for disease treatment, dual and triple agonists of GIPR, in conjunction with that of glucagon-like peptide-1 (GLP-1) and glucagon receptors, are currently in clinical trials, with an expectation of enhanced efficacy beyond that of GLP-1 receptor (GLP-1R) agonist monotherapy for diabetic patients. Consequently, it is important to understand the pharmacological behavior of such drugs. In this study, we have explored signaling pathway specificity and the potential for biased agonism of mono-, dual- and tri-agonists of GIPR using human embryonic kidney 293 (HEK293) cells recombinantly expressing human GIPR or GLP-1R. Compared to GIP(1-42), the GIPR mono-agonists Pro3GIP and Lys3GIP are biased towards ERK1/2 phosphorylation (pERK1/2) relative to cAMP accumulation at GIPR, whereas the triple agonist at GLP-1R/GCGR/GIPR is biased towards pERK1/2 relative to β-arrestin2 recruitment. Moreover, the dual GIPR/GLP-1R agonist, LY3298176, is biased towards pERK1/2 relative to cAMP accumulation at both GIPR and GLP-1R compared to their respective endogenous ligands. These data reveal novel pharmacological properties of potential therapeutic agents that may impact on diversity in clinical responses.
- Published
- 2020
14. Granzyme A in Chikungunya and other arboviral infections
- Author
-
Dion Kaiserman, Luigi Aurelio, Thuy T. Le, Natalie A. Prow, Andreas Suhrbier, Phillip I. Bird, Peishen Zhao, Kelly Grace Magalhães, Diego Damasco Barboza, Alessandra Soares Schanoski, Paul R. Young, Cliomar Alves dos Santos, Paolo Marinho de Andrade Zanotto, David A. Muller, and Caitlin L. Rowe
- Subjects
0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,chikungunya ,Immunology ,Inflammation ,Arbovirus Infections ,Biology ,Dengue virus ,medicine.disease_cause ,Granzymes ,Virus ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Interferon ,granzyme A ,medicine ,Animals ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,NK cell ,Chikungunya ,Original Research ,ENZIMAS PROTEOLÍTICAS ,virus diseases ,Virology ,3. Good health ,Killer Cells, Natural ,Mice, Inbred C57BL ,030104 developmental biology ,arbovirus ,arthritis ,Granzyme A ,Chikungunya Fever ,medicine.symptom ,lcsh:RC581-607 ,Viral load ,030215 immunology ,medicine.drug - Abstract
Granzyme A (GzmA) is secreted by cytotoxic lymphocytes and has traditionally been viewed as a mediator of cell death. However, a growing body of data suggests the physiological role of GzmA is promotion of inflammation. Here, we show that GzmA is significantly elevated in the sera of chikungunya virus (CHIKV) patients and that GzmA levels correlated with viral loads and disease scores in these patients. Serum GzmA levels were also elevated in CHIKV mouse models, with NK cells the likely source. Infection of mice deficient in type I interferon responses with CHIKV, Zika virus, or dengue virus resulted in high levels of circulating GzmA. We also show that subcutaneous injection of enzymically active recombinant mouse GzmA was able to mediate inflammation, both locally at the injection site as well as at a distant site. Protease activated receptors (PARs) may represent targets for GzmA, and we show that treatment with PAR antagonist ameliorated GzmA- and CHIKV-mediated inflammation.
- Published
- 2020
15. Glucagon-like peptide-1 receptor internalisation controls spatiotemporal signalling mediated by biased agonists
- Author
-
Madeleine M. Fletcher, Patrick M. Sexton, Lachlan Clydesdale, Peishen Zhao, Denise Wootten, Michelle L. Halls, and Arthur Christopoulos
- Subjects
Dynamins ,0301 basic medicine ,MAPK/ERK pathway ,Agonist ,medicine.drug_class ,Caveolin 1 ,CHO Cells ,Biochemistry ,Glucagon-Like Peptide-1 Receptor ,03 medical and health sciences ,chemistry.chemical_compound ,Cricetulus ,Caveolae ,Fluorescence Resonance Energy Transfer ,medicine ,Functional selectivity ,Animals ,Receptor ,beta-Arrestins ,Dynamin ,G protein-coupled receptor ,Pharmacology ,Chemistry ,Liraglutide ,Clathrin ,Cell biology ,Oxyntomodulin ,030104 developmental biology ,Gene Expression Regulation ,Luminescent Measurements ,Exenatide ,Plasmids ,Signal Transduction - Abstract
The glucagon-like peptide-1 receptor (GLP-1R) is a major therapeutic target in the treatment of type 2 diabetes due to its roles in regulating blood glucose and in promoting weight loss. Like many GPCRs, it is pleiotropically coupled, can be activated by multiple ligands and is subject to biased agonism. The GLP-1R undergoes agonist mediated receptor internalisation that may be associated with spatiotemporal control of signalling and biased agonism, although to date, this has not been extensively explored. Here, we investigate GLP-1R trafficking and its importance with regard to signalling, including the localisation of key signalling molecules, mediated by biased peptide agonists that are either endogenous GLP-1R ligands or are used clinically. Each of the agonists promoted receptor internalisation through a dynamin and caveolae dependent mechanism and traffic the receptor to both degradative and recycling pathways. This internalisation is important for signalling, with cAMP and ERK1/2 phoshorylation (pERK1/2) generated by both plasma membrane localised and internalised receptors. Further assessment of pERK1/2 revealed that all peptides induced nuclear ERK activity, but ligands, liraglutide and oxyntomodulin that are biased towards pERK1/2 relative to cAMP (when compared to GLP-1 and exendin-4), also stimulated pERK1/2 activity in the cytosol. This compartmentalisation of ERK1/2 signalling was reliant on receptor internalisation, with restriction of receptor localisation to the plasma membrane limiting ERK1/2 signalling to the cytosol. Thus, this study implicates a role of receptor internalisation in spatiotemporal control of ERK1/2 signalling that may contribute to GLP-1R biased agonism.
- Published
- 2018
- Full Text
- View/download PDF
16. Two distinct domains of the glucagon-like peptide-1 receptor control peptide-mediated biased agonism
- Author
-
Patrick M. Sexton, Antao Dai, Thomas Coudrat, Arthur Christopoulos, Lachlan Clydesdale, Yi Lynn Liang, Dehua Yang, Peishen Zhao, Denise Wootten, Saifei Lei, Cassandra Koole, Xiaoqing Cai, Yang Feng, and Ming-Wei Wang
- Subjects
Models, Molecular ,0301 basic medicine ,Cell signaling ,Protein Conformation ,CHO Cells ,Crystallography, X-Ray ,Ligands ,Biochemistry ,Glucagon-Like Peptide-1 Receptor ,03 medical and health sciences ,Cricetulus ,Protein Domains ,Drug Discovery ,Cyclic AMP ,Arrestin ,Functional selectivity ,Animals ,Humans ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Receptor ,Molecular Biology ,Glucagon-like peptide 1 receptor ,G protein-coupled receptor ,Chemistry ,Cell Biology ,Cell biology ,Transmembrane domain ,030104 developmental biology ,Mutagenesis ,Calcium ,Signal transduction ,Peptides ,Signal Transduction - Abstract
G protein–coupled receptors (GPCRs) can be differentially activated by ligands to generate multiple and distinct downstream signaling profiles, a phenomenon termed biased agonism. The glucagon-like peptide-1 receptor (GLP-1R) is a class B GPCR and a key drug target for managing metabolic disorders; however, its peptide agonists display biased signaling that affects their relative efficacies. In this study, we combined mutagenesis experiments and mapping of surface mutations onto recently described GLP-1R structures, which revealed two major domains in the GLP-1/GLP-1R/G(s) protein active structure that are differentially important for both receptor quiescence and ligand-specific initiation and propagation of biased agonism. Changes to the conformation of transmembrane helix (TM) 5 and TM 6 and reordering of extracellular loop 2 were essential for the propagation of signaling linked to cAMP formation and intracellular calcium mobilization, whereas ordering and packing of residues in TMs 1 and 7 were critical for extracellular signal–regulated kinase 1/2 (pERK) activity. On the basis of these findings, we propose a model of distinct peptide–receptor interactions that selectively control how these different signaling pathways are engaged. This work provides important structural insight into class B GPCR activation and biased agonism.
- Published
- 2018
- Full Text
- View/download PDF
17. Phase-plate cryo-EM structure of a biased agonist-bound human GLP-1 receptor–Gs complex
- Author
-
Denise Wootten, Tin T. Truong, Saifei Lei, Ming-Wei Wang, Arthur Christopoulos, Wolfgang Baumeister, Lachlan Clydesdale, Yi Lynn Liang, Cassandra Koole, Laurence J. Miller, David M. Thal, Patrick M. Sexton, Mazdak Radjainia, Alisa Glukhova, Peishen Zhao, Sebastian G.B. Furness, Maryam Khoshouei, and Radostin Danev
- Subjects
Models, Molecular ,0301 basic medicine ,Agonist ,endocrine system ,Gs alpha subunit ,Protein Conformation ,G protein ,medicine.drug_class ,Peptide ,Glucagon-Like Peptide-1 Receptor ,03 medical and health sciences ,0302 clinical medicine ,Glucagon-Like Peptide 1 ,GTP-Binding Protein alpha Subunits, Gs ,Functional selectivity ,medicine ,Humans ,Receptor ,Glucagon-like peptide 1 receptor ,chemistry.chemical_classification ,Binding Sites ,Multidisciplinary ,Cryoelectron Microscopy ,digestive, oral, and skin physiology ,Transmembrane protein ,030104 developmental biology ,chemistry ,Biophysics ,hormones, hormone substitutes, and hormone antagonists ,030217 neurology & neurosurgery - Abstract
The class B glucagon-like peptide-1 (GLP-1) G protein-coupled receptor is a major target for the treatment of type 2 diabetes and obesity. Endogenous and mimetic GLP-1 peptides exhibit biased agonism-a difference in functional selectivity-that may provide improved therapeutic outcomes. Here we describe the structure of the human GLP-1 receptor in complex with the G protein-biased peptide exendin-P5 and a Gαs heterotrimer, determined at a global resolution of 3.3 A. At the extracellular surface, the organization of extracellular loop 3 and proximal transmembrane segments differs between our exendin-P5-bound structure and previous GLP-1-bound GLP-1 receptor structure. At the intracellular face, there was a six-degree difference in the angle of the Gαs-α5 helix engagement between structures, which was propagated across the G protein heterotrimer. In addition, the structures differed in the rate and extent of conformational reorganization of the Gαs protein. Our structure provides insights into the molecular basis of biased agonism.
- Published
- 2018
- Full Text
- View/download PDF
18. Structural basis of G
- Author
-
Anna, Qiao, Shuo, Han, Xinmei, Li, Zhixin, Li, Peishen, Zhao, Antao, Dai, Rulve, Chang, Linhua, Tai, Qiuxiang, Tan, Xiaojing, Chu, Limin, Ma, Thor Seneca, Thorsen, Steffen, Reedtz-Runge, Dehua, Yang, Ming-Wei, Wang, Patrick M, Sexton, Denise, Wootten, Fei, Sun, Qiang, Zhao, and Beili, Wu
- Subjects
Models, Molecular ,Protein Conformation, alpha-Helical ,Binding Sites ,Protein Conformation ,Cryoelectron Microscopy ,GTP-Binding Protein alpha Subunits, Gs ,Receptors, Glucagon ,Humans ,GTP-Binding Protein alpha Subunits, Gi-Go ,Glucagon ,Protein Binding ,Signal Transduction - Abstract
Class B G protein-coupled receptors, an important class of therapeutic targets, signal mainly through the G
- Published
- 2019
19. Activation of the GLP-1 receptor by a non-peptidic agonist
- Author
-
Arthur Christopoulos, Patrick M. Sexton, Lachlan Clydesdale, Asuka Inoue, Yi Lynn Liang, Giuseppe Deganutti, Tin T. Truong, Matthew J. Belousoff, Michael E. Christe, Sebastian G.B. Furness, Michael Gregory Bell, Christopher A. Reynolds, Denise Wootten, Kyle W. Sloop, Ming-Wei Wang, Francis S. Willard, Peishen Zhao, Laurence J. Miller, Madeleine M. Fletcher, and Radostin Danev
- Subjects
0301 basic medicine ,Agonist ,Models, Molecular ,medicine.drug_class ,Pyridines ,Phenylalanine ,CHO Cells ,Glucagon-Like Peptide-1 Receptor ,03 medical and health sciences ,0302 clinical medicine ,Protein structure ,Cricetulus ,Cricetinae ,medicine ,Functional selectivity ,Extracellular ,Animals ,Humans ,Receptor ,Protein Structure, Quaternary ,Glucagon-like peptide 1 receptor ,Multidisciplinary ,Chemistry ,Ligand (biochemistry) ,Isoquinolines ,Protein Structure, Tertiary ,Transmembrane domain ,Kinetics ,030104 developmental biology ,Structural Homology, Protein ,Biophysics ,030217 neurology & neurosurgery - Abstract
Class B G-protein-coupled receptors are major targets for the treatment of chronic diseases, including diabetes and obesity1. Structures of active receptors reveal peptide agonists engage deep within the receptor core, leading to an outward movement of extracellular loop 3 and the tops of transmembrane helices 6 and 7, an inward movement of transmembrane helix 1, reorganization of extracellular loop 2 and outward movement of the intracellular side of transmembrane helix 6, resulting in G-protein interaction and activation2,3,4,5,6. Here we solved the structure of a non-peptide agonist, TT-OAD2, bound to the glucagon-like peptide-1 (GLP-1) receptor. Our structure identified an unpredicted non-peptide agonist-binding pocket in which reorganization of extracellular loop 3 and transmembrane helices 6 and 7 manifests independently of direct ligand interaction within the deep transmembrane domain pocket. TT-OAD2 exhibits biased agonism, and kinetics of G-protein activation and signalling that are distinct from peptide agonists. Within the structure, TT-OAD2 protrudes beyond the receptor core to interact with the lipid or detergent, providing an explanation for the distinct activation kinetics that may contribute to the clinical efficacy of this compound series. This work alters our understanding of the events that drive the activation of class B receptors.
- Published
- 2019
20. Antagonism of the proinflammatory and pronociceptive actions of canonical and biased agonists of protease‐activated receptor‐2
- Author
-
Tina Marie Lieu, Laura E. Edgington-Mitchell, Nigel W. Bunnett, David P. Fairlie, Romke Bron, Daniel P. Poole, Peter McLean, Nicholas Barlow, E. Savage, Peishen Zhao, and Rink-Jan Lohman
- Subjects
Male ,0301 basic medicine ,Proteases ,Administration, Oral ,Proinflammatory cytokine ,Rats, Sprague-Dawley ,Mice ,Structure-Activity Relationship ,03 medical and health sciences ,medicine ,Animals ,Receptor, PAR-2 ,Protease-activated receptor 2 ,Cathepsin S ,Inflammation ,Mice, Knockout ,Pharmacology ,Cathepsin ,Dose-Response Relationship, Drug ,Chemistry ,Elastase ,Nociceptors ,Research Papers ,Rats ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Nociception ,Biochemistry ,Hyperalgesia ,medicine.symptom ,Oligopeptides ,Research Paper - Abstract
Background and Purpose Diverse proteases cleave protease-activated receptor-2 (PAR2) on primary sensory neurons and epithelial cells to evoke pain and inflammation. Trypsin and tryptase activate PAR2 by a canonical mechanism that entails cleavage within the extracellular N-terminus revealing a tethered ligand that activates the cleaved receptor. Cathepsin-S and elastase are biased agonists that cleave PAR2 at different sites to activate distinct signalling pathways. Although PAR2 is a therapeutic target for inflammatory and painful diseases, the divergent mechanisms of proteolytic activation complicate the development of therapeutically useful antagonists. Experimental Approach We investigated whether the PAR2 antagonist GB88 inhibits protease-evoked activation of nociceptors and protease-stimulated oedema and hyperalgesia in rodents. Key Results Intraplantar injection of trypsin, cathespsin-S or elastase stimulated mechanical and thermal hyperalgesia and oedema in mice. Oral GB88 or par2 deletion inhibited the algesic and proinflammatory actions of all three proteases, but did not affect basal responses. GB88 also prevented pronociceptive and proinflammatory effects of the PAR2-selective agonists 2-furoyl-LIGRLO-NH2 and AC264613. GB88 did not affect capsaicin-evoked hyperalgesia or inflammation. Trypsin, cathepsin-S and elastase increased [Ca2+]i in rat nociceptors, which expressed PAR2. GB88 inhibited this activation of nociceptors by all three proteases, but did not affect capsaicin-evoked activation of nociceptors or inhibit the catalytic activity of the three proteases. Conclusions and Implications GB88 inhibits the capacity of canonical and biased protease agonists of PAR2 to cause nociception and inflammation.
- Published
- 2016
- Full Text
- View/download PDF
21. Protein Kinase D and Gβγ Subunits Mediate Agonist-evoked Translocation of Protease-activated Receptor-2 from the Golgi Apparatus to the Plasma Membrane
- Author
-
Peishen Zhao, Stephen Vanner, Daniel P. Poole, Dane D. Jensen, Meritxell Canals, Marina Gerges, Holly R. Yeatman, Tina Marie Lieu, Nestor N. Jiménez-Vargas, and Nigel W. Bunnett
- Subjects
0301 basic medicine ,Proteases ,Biological Transport, Active ,Golgi Apparatus ,Endosomes ,Biology ,Endocytosis ,Biochemistry ,Cell Line ,Mice ,03 medical and health sciences ,symbols.namesake ,0302 clinical medicine ,GTP-Binding Protein gamma Subunits ,Extracellular ,Animals ,Humans ,Receptor, PAR-2 ,Calcium Signaling ,Receptor ,Protein Kinase Inhibitors ,Molecular Biology ,Protein Kinase C ,Protease-activated receptor 2 ,Activator (genetics) ,Cell Membrane ,GTP-Binding Protein beta Subunits ,Cell Biology ,Golgi apparatus ,Rats ,Cell biology ,Mice, Inbred C57BL ,HEK293 Cells ,Pyrimidines ,030104 developmental biology ,Xanthenes ,symbols ,030217 neurology & neurosurgery ,Intracellular ,Signal Transduction - Abstract
Agonist-evoked endocytosis of G protein-coupled receptors has been extensively studied. The mechanisms by which agonists stimulate mobilization and plasma membrane translocation of G protein-coupled receptors from intracellular stores are unexplored. Protease-activated receptor-2 (PAR2) traffics to lysosomes, and sustained protease signaling requires mobilization and plasma membrane trafficking of PAR2 from Golgi stores. We evaluated the contribution of protein kinase D (PKD) and Gβγ to this process. In HEK293 and KNRK cells, the PAR2 agonists trypsin and 2-furoyl-LIGRLO-NH2 activated PKD in the Golgi apparatus, where PKD regulates protein trafficking. PAR2 activation induced translocation of Gβγ, a PKD activator, to the Golgi apparatus, determined by bioluminescence resonance energy transfer between Gγ-Venus and giantin-Rluc8. Inhibitors of PKD (CRT0066101) and Gβγ (gallein) prevented PAR2-stimulated activation of PKD. CRT0066101, PKD1 siRNA, and gallein all inhibited recovery of PAR2-evoked Ca(2+) signaling. PAR2 with a photoconvertible Kaede tag was expressed in KNRK cells to examine receptor translocation from the Golgi apparatus to the plasma membrane. Irradiation of the Golgi region (405 nm) induced green-red photo-conversion of PAR2-Kaede. Trypsin depleted PAR2-Kaede from the Golgi apparatus and repleted PAR2-Kaede at the plasma membrane. CRT0066101 inhibited PAR2-Kaede translocation to the plasma membrane. CRT0066101 also inhibited sustained protease signaling to colonocytes and nociceptive neurons that naturally express PAR2 and mediate protease-evoked inflammation and nociception. Our results reveal a major role for PKD and Gβγ in agonist-evoked mobilization of intracellular PAR2 stores that is required for sustained signaling by extracellular proteases.
- Published
- 2016
- Full Text
- View/download PDF
22. Toward a Structural Understanding of Class B GPCR Peptide Binding and Activation
- Author
-
Radostin Danev, Patrick M. Sexton, Maryam Khoshouei, Peishen Zhao, Matthew J. Belousoff, Arthur Christopoulos, George Christopoulos, Yi Lynn Liang, Madeleine M. Fletcher, Denise Wootten, H. Eric Xu, Cassandra Koole, Yan Zhang, Tin T. Truong, and Villy Julita
- Subjects
Models, Molecular ,G protein ,Adenylate kinase ,Peptide binding ,Peptide ,Biology ,Ligands ,Receptors, Corticotropin-Releasing Hormone ,Receptors, G-Protein-Coupled ,Conserved sequence ,Corticotropin-releasing hormone receptor 1 ,03 medical and health sciences ,0302 clinical medicine ,Humans ,Amino Acid Sequence ,Protein Precursors ,Receptor ,Molecular Biology ,030304 developmental biology ,G protein-coupled receptor ,chemistry.chemical_classification ,0303 health sciences ,Cryoelectron Microscopy ,Enkephalins ,Cell Biology ,Cell biology ,chemistry ,Peptides ,030217 neurology & neurosurgery ,Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I ,Signal Transduction - Abstract
Class B G protein-coupled receptors (GPCRs) are important therapeutic targets for major diseases. Here, we present structures of peptide and Gs-bound pituitary adenylate cyclase-activating peptide, PAC1 receptor, and corticotropin-releasing factor (CRF), (CRF1) receptor. Together with recently solved structures, these provide coverage of the major class B GPCR subfamilies. Diverse orientations of the extracellular domain to the receptor core in different receptors are at least partially dependent on evolutionary conservation in the structure and nature of peptide interactions. Differences in peptide interactions to the receptor core also influence the interlinked TM2-TM1-TM6/ECL3/TM7 domain, and this is likely important in their diverse signaling. However, common conformational reorganization of ECL2, linked to reorganization of ICL2, modulates G protein contacts. Comparison between receptors reveals ICL2 as a key domain forming dynamic G protein interactions in a receptor- and ligand-specific manner. This work advances our understanding of class B GPCR activation and Gs coupling.
- Published
- 2020
- Full Text
- View/download PDF
23. Protein kinase D and Gβγ mediate sustained nociceptive signaling by biased agonists of protease-activated receptor-2
- Author
-
Luke A. Pattison, Cintya Lopez-Lopez, Stephen Vanner, Daniel P. Poole, Dane D. Jensen, Brian L. Schmidt, Nestor N. Jiménez-Vargas, Nigel W. Bunnett, Tina Marie Lieu, Josue Obed Jaramillo, Peishen Zhao, and Rocco Latorre
- Subjects
0301 basic medicine ,Proteases ,Gs alpha subunit ,medicine.medical_treatment ,Golgi Apparatus ,Biochemistry ,03 medical and health sciences ,symbols.namesake ,Mice ,GTP-Binding Protein gamma Subunits ,Ganglia, Spinal ,medicine ,Animals ,Humans ,Receptor, PAR-2 ,Molecular Biology ,Protease-activated receptor 2 ,Protein Kinase C ,Cathepsin ,Protease ,030102 biochemistry & molecular biology ,biology ,Chemistry ,Cell Membrane ,GTP-Binding Protein beta Subunits ,Cell Biology ,Golgi apparatus ,Cathepsins ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,HEK293 Cells ,Pyrimidines ,Gq alpha subunit ,Xanthenes ,Hyperalgesia ,symbols ,biology.protein ,Signal transduction ,Leukocyte Elastase ,Signal Transduction - Abstract
Proteases sustain hyperexcitability and pain by cleaving protease-activated receptor-2 (PAR(2)) on nociceptors through distinct mechanisms. Whereas trypsin induces PAR(2) coupling to Gα(q), Gα(s), and β-arrestins, cathepsin-S (CS) and neutrophil elastase (NE) cleave PAR(2) at distinct sites and activate it by biased mechanisms that induce coupling to Gα(s), but not to Gα(q) or β-arrestins. Because proteases activate PAR(2) by irreversible cleavage, and activated PAR(2) is degraded in lysosomes, sustained extracellular protease-mediated signaling requires mobilization of intact PAR(2) from the Golgi apparatus or de novo synthesis of new receptors by incompletely understood mechanisms. We found here that trypsin, CS, and NE stimulate PAR(2)-dependent activation of protein kinase D (PKD) in the Golgi of HEK293 cells, in which PKD regulates protein trafficking. The proteases stimulated translocation of the PKD activator Gβγ to the Golgi, coinciding with PAR(2) mobilization from the Golgi. Proteases also induced translocation of a photoconverted PAR(2)-Kaede fusion protein from the Golgi to the plasma membrane of KNRK cells. After incubation of HEK293 cells and dorsal root ganglia neurons with CS, NE, or trypsin, PAR(2) responsiveness initially declined, consistent with PAR(2) cleavage and desensitization, and then gradually recovered. Inhibitors of PKD, Gβγ, and protein translation inhibited recovery of PAR(2) responsiveness. PKD and Gβγ inhibitors also attenuated protease-evoked mechanical allodynia in mice. We conclude that proteases that activate PAR(2) by canonical and biased mechanisms stimulate PKD in the Golgi; PAR(2) mobilization and de novo synthesis repopulate the cell surface with intact receptors and sustain nociceptive signaling by extracellular proteases.
- Published
- 2018
24. Protease-activated receptor-2 in endosomes signals persistent pain of irritable bowel syndrome
- Author
-
Holly R. Yeatman, Stuart M. Brierley, Dane D. Jensen, Peishen Zhao, Aditi Bhattacharya, Bernard L. Flynn, Nigel W. Bunnett, Christopher J.H. Porter, Rocco Latorre, Elyssa Chen, Daniel P. Poole, Laura E. Edgington-Mitchell, Luke A. Pattison, Peter McLean, Tina Marie Lieu, Michelle L. Halls, Nicholas A. Veldhuis, Nicole N. Scheff, Brian L. Schmidt, Giang Thanh Le, Gareth A. Hicks, Meritxell Canals, Stephen Vanner, Nestor N. Jiménez-Vargas, Luigi Aurelio, Joel Castro, and Carmen Klein Herenbrink
- Subjects
0301 basic medicine ,Nociception ,Endosome ,Endosomes ,Pharmacology ,Endocytosis ,Irritable Bowel Syndrome ,03 medical and health sciences ,medicine ,Animals ,Humans ,Receptor, PAR-2 ,Trypsin ,Extracellular Signal-Regulated MAP Kinases ,Protease-activated receptor 2 ,Cathepsin S ,Cathepsin ,Multidisciplinary ,Chemistry ,Beta-Arrestins ,Nociceptors ,3. Good health ,030104 developmental biology ,Allodynia ,PNAS Plus ,medicine.symptom ,Chronic Pain ,Signal Transduction - Abstract
Once activated at the surface of cells, G protein-coupled receptors (GPCRs) redistribute to endosomes, where they can continue to signal. Whether GPCRs in endosomes generate signals that contribute to human disease is unknown. We evaluated endosomal signaling of protease-activated receptor-2 (PAR2), which has been proposed to mediate pain in patients with irritable bowel syndrome (IBS). Trypsin, elastase, and cathepsin S, which are activated in the colonic mucosa of patients with IBS and in experimental animals with colitis, caused persistent PAR2-dependent hyperexcitability of nociceptors, sensitization of colonic afferent neurons to mechanical stimuli, and somatic mechanical allodynia. Inhibitors of clathrin- and dynamin-dependent endocytosis and of mitogen-activated protein kinase kinase-1 prevented trypsin-induced hyperexcitability, sensitization, and allodynia. However, they did not affect elastase- or cathepsin S-induced hyperexcitability, sensitization, or allodynia. Trypsin stimulated endocytosis of PAR2, which signaled from endosomes to activate extracellular signal-regulated kinase. Elastase and cathepsin S did not stimulate endocytosis of PAR2, which signaled from the plasma membrane to activate adenylyl cyclase. Biopsies of colonic mucosa from IBS patients released proteases that induced persistent PAR2-dependent hyperexcitability of nociceptors, and PAR2 association with β-arrestins, which mediate endocytosis. Conjugation to cholestanol promoted delivery and retention of antagonists in endosomes containing PAR2 A cholestanol-conjugated PAR2 antagonist prevented persistent trypsin- and IBS protease-induced hyperexcitability of nociceptors. The results reveal that PAR2 signaling from endosomes underlies the persistent hyperexcitability of nociceptors that mediates chronic pain of IBS. Endosomally targeted PAR2 antagonists are potential therapies for IBS pain. GPCRs in endosomes transmit signals that contribute to human diseases.
- Published
- 2018
25. Cathepsin S Causes Inflammatory Pain via Biased Agonism of PAR2 and TRPV4
- Author
-
Erik Lindström, Stephen Vanner, Peishen Zhao, Dane D. Jensen, Matthew C. Metcalf, Peter McIntyre, Wolfgang Liedtke, Nicholas A. Veldhuis, Raquel Guerrero-Alba, Martina Kocan, Tina Marie Lieu, Silke Haerteis, Nigel W. Bunnett, Ian R. Henderson, Eduardo E. Valdez-Morales, Silvia Sostegni, Vera Baraznenok, Christoph Korbmacher, and Nicholas Barlow
- Subjects
medicine.medical_specialty ,Gs alpha subunit ,Pain ,TRPV Cation Channels ,Biology ,Biochemistry ,Adenylyl cyclase ,Mice ,Xenopus laevis ,chemistry.chemical_compound ,Internal medicine ,medicine ,Animals ,Humans ,Receptor, PAR-2 ,Protein kinase A ,Molecular Biology ,Cathepsin S ,Inflammation ,Mice, Knockout ,Cathepsin ,Neurogenic inflammation ,HEK 293 cells ,Cell Biology ,Cathepsins ,Cell biology ,HEK293 Cells ,Endocrinology ,chemistry ,Hyperalgesia ,Additions and Corrections ,medicine.symptom ,Adenylyl Cyclases ,Signal Transduction - Abstract
Serine proteases such as trypsin and mast cell tryptase cleave protease-activated receptor-2 (PAR2) at R(36)↓S(37) and reveal a tethered ligand that excites nociceptors, causing neurogenic inflammation and pain. Whether proteases that cleave PAR2 at distinct sites are biased agonists that also induce inflammation and pain is unexplored. Cathepsin S (Cat-S) is a lysosomal cysteine protease of antigen-presenting cells that is secreted during inflammation and which retains activity at extracellular pH. We observed that Cat-S cleaved PAR2 at E(56)↓T(57), which removed the canonical tethered ligand and prevented trypsin activation. In HEK and KNRK cell lines and in nociceptive neurons of mouse dorsal root ganglia, Cat-S and a decapeptide mimicking the Cat-S-revealed tethered ligand-stimulated PAR2 coupling to Gαs and formation of cAMP. In contrast to trypsin, Cat-S did not mobilize intracellular Ca(2+), activate ERK1/2, recruit β-arrestins, or induce PAR2 endocytosis. Cat-S caused PAR2-dependent activation of transient receptor potential vanilloid 4 (TRPV4) in Xenopus laevis oocytes, HEK cells and nociceptive neurons, and stimulated neuronal hyperexcitability by adenylyl cyclase and protein kinase A-dependent mechanisms. Intraplantar injection of Cat-S caused inflammation and hyperalgesia in mice that was attenuated by PAR2 or TRPV4 deletion and adenylyl cyclase inhibition. Cat-S and PAR2 antagonists suppressed formalin-induced inflammation and pain, which implicates endogenous Cat-S and PAR2 in inflammatory pain. Our results identify Cat-S as a biased agonist of PAR2 that causes PAR2- and TRPV4-dependent inflammation and pain. They expand the role of PAR2 as a mediator of protease-driven inflammatory pain.
- Published
- 2014
- Full Text
- View/download PDF
26. The nature of efficacy at G protein-coupled receptors
- Author
-
Sebastian G.B. Furness and Peishen Zhao
- Subjects
0301 basic medicine ,Pharmacology ,Cell signaling ,Drug Inverse Agonism ,Biology ,Ligands ,Biochemistry ,Receptors, G-Protein-Coupled ,GPCR Signaling ,03 medical and health sciences ,Treatment Outcome ,030104 developmental biology ,0302 clinical medicine ,Pharmaceutical Preparations ,030220 oncology & carcinogenesis ,Functional selectivity ,Animals ,Humans ,Receptor structure function ,Signal transduction ,Receptor ,Neuroscience ,Receptor theory ,Signal Transduction ,G protein-coupled receptor - Abstract
G protein-coupled receptors (GPCRs) participate in many pathophysiological processes as well as almost all aspects of normal physiology. They are present at the surface of all cell types making them amenable and attractive targets for pharmaceutical therapeutics. GPCRs possess complex pharmacology with the ability to be turned on to various extents, have their constitutive activity suppressed and even switch between signaling pathways to which they couple. Underlying this complex pharmacology is GPCR signaling efficacy, and differences in efficacy promoted by alternative ligands and in different tissues is of great interest to biology in general and also the pharmaceutical industry. In this review we hope to discuss what the molecular foundations of efficacy are and whether a new approach utilizing a rate-dependent model may provide new insights into this phenomenon.
- Published
- 2019
- Full Text
- View/download PDF
27. RGS2 is a component of the cellular stress response
- Author
-
Chau H. Nguyen, Peter Chidiac, Alina J. Sobiesiak, and Peishen Zhao
- Subjects
Proteome ,GTPase-activating protein ,Eukaryotic Initiation Factor-2 ,Biophysics ,Apoptosis ,Biology ,Biochemistry ,Cell Line ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Regulator of G protein signaling ,Downregulation and upregulation ,Stress, Physiological ,Cellular stress response ,Heterotrimeric G protein ,Animals ,RNA, Small Interfering ,Molecular Biology ,RGS2 ,030304 developmental biology ,0303 health sciences ,Cell Biology ,Fibroblasts ,RGS17 ,Molecular biology ,Cell biology ,Gene Knockdown Techniques ,Protein Biosynthesis ,Unfolded protein response ,Heat-Shock Response ,RGS Proteins ,030217 neurology & neurosurgery - Abstract
Regulator of G protein signaling (RGS) proteins are GTPase accelerating proteins for heterotrimeric G protein α-subunits. RGS2 has recently been shown to have additional G protein-independent functions including control of ion channel currents, microtubule polymerization, and protein synthesis. Cellular levels of RGS2 mRNA and protein are upregulated in response to various forms of stress suggesting that it may be a stress-adaptive protein; however, direct evidence to support this notion has remained elusive. In this report, we show that thermal stress upregulates RGS2 expression and this serves to arrest de novo protein synthesis. The latter is an established cellular response to stress. Inhibiting the stress-induced RGS2 upregulation by way of siRNA knockdown diminished the repression of global protein synthesis. The collective results of our study implicate RGS2 upregulation as a cellular mechanism of controlling de novo protein synthesis in response to stress. This work provides greater insight into the stress proteome and the role of RGS2.
- Published
- 2012
- Full Text
- View/download PDF
28. Demonstration of elevated levels of active cathepsin S in dextran sulfate sodium colitis using a new activatable probe
- Author
-
Raquel Guerrero-Alba, Joshua W. Conner, Daniel P. Poole, Yasmin Nasser, Tina Marie Lieu, Nicholas Barlow, Peishen Zhao, Stephanie Vanner, Bimbil Graham, Laura E. Edgington-Mitchell, Namit Sharma, Nicholas A. Veldhuis, Erik Lindström, Andrew W.B. Craig, and Nigel W. Bunnett
- Subjects
Proteases ,Physiology ,medicine.medical_treatment ,Inflammation ,Mice ,medicine ,Animals ,Colitis ,Receptor ,Cathepsin S ,Fluorescent Dyes ,Cathepsin ,Gastrointestinal tract ,Protease ,Endocrine and Autonomic Systems ,Chemistry ,Dextran Sulfate ,Gastroenterology ,medicine.disease ,Molecular biology ,Cathepsins ,Mice, Inbred C57BL ,Disease Models, Animal ,Biochemistry ,medicine.symptom - Abstract
Background Proteases play a major role in inflammatory diseases of the gastrointestinal tract. Activatable probes are a major technological advance, enabling sensitive detection of active proteases in tissue samples. Our aim was to synthesize an activatable probe for cathepsin S and validate its use in a mouse model of colitis. Methods We designed and synthesized a new fluorescent activatable probe, NB200, for the detection of active cathepsin S. Colitis was induced in C57BL/6 mice by the administration of 3% dextran sulfate sodium (DSS). Homogenized mouse colons, with or without the addition of the specific cathepsin S inhibitor MV026031, were incubated with NB200 in a fluorescent plate reader. Key Results NB200 selectively detected purified cathepsin S and not other common inflammatory proteases. Homogenates of colon from mice with DSS colitis induced a significant fluorescent increase when compared to control animals (control vs DSS: p
- Published
- 2015
29. Biased signaling of protease-activated receptors
- Author
-
Peishen, Zhao, Matthew, Metcalf, and Nigel W, Bunnett
- Subjects
Endocrinology ,General Commentary ,proteases ,PARs ,signal transduction ,biased signaling ,G proteins ,β-arrestins - Published
- 2014
30. Regulation of RGS5 GAP activity by GPSM3
- Author
-
Peter Chidiac and Peishen Zhao
- Subjects
Male ,GTP' ,GTPase-activating protein ,G protein ,Clinical Biochemistry ,Myocytes, Smooth Muscle ,GTPase ,CHO Cells ,Rats, Inbred WKY ,Cell Line ,RGS4 ,Cricetulus ,GTP-Binding Protein Regulators ,GTP-Binding Proteins ,Heterotrimeric G protein ,Animals ,Molecular Biology ,G alpha subunit ,Guanine Nucleotide Dissociation Inhibitors ,biology ,Cell Biology ,General Medicine ,Heterotrimeric GTP-Binding Proteins ,Cell biology ,Rats ,Biochemistry ,biology.protein ,Guanosine Triphosphate ,RGS Proteins ,Protein Binding ,Signal Transduction - Abstract
Heterotrimeric G protein signaling is limited by intracellular proteins that impede the binding of or accelerate the hydrolysis of the activating nucleotide GTP, exemplified respectively by the G protein-signaling modifier (GPSM) and regulator of G protein-signaling (RGS) families of proteins. Little is known about how members of these groups of proteins might influence the impact of the other on G protein activity. In the present study, we have identified novel binding and functional interactions between GPSM3 (also known as activator of G protein-signaling 4 (AGS4) or G18) and RGS5, both of which were found to be expressed in primary rat aortic smooth muscle cell cultures. The binding of GPSM3 to RGS5 appears to be selective as no interactions were detected with other RGS proteins tested. In solution-based experiments, the addition of GPSM3 was found to enhance the ability of RGS5 to accelerate GTP hydrolysis by Gαi1 but not that of RGS4. In membrane-based assays utilizing M2 muscarinic receptor-activated Gαi1, GPSM3 decreased the rate of GTP hydrolysis in the presence of RGS4 but not RGS5, suggesting that the enhancement of RGS5 activity by GPSM3 is maintained under these conditions and/or that the binding of RGS5 to GPSM3 impedes its inhibitory effect on GTP turnover. Overall these findings show that it is possible for GPSM and RGS proteins to bind to one another to produce distinct regulatory effects on heterotrimeric G protein activity.
- Published
- 2014
31. The bile acid receptor TGR5 activates the TRPA1 channel to induce itch in mice
- Author
-
Megan S. Grace, Nigel W. Bunnett, Tina Marie Lieu, Silke Haerteis, Serena Materazzi, Yvette M. Wilson, Daniel P. Poole, Christoph Korbmacher, Martin Steinhoff, Peishen Zhao, Gihan Jayaweera, Matteus Krappitz, Peter McIntyre, Pierangelo Geppetti, Dane D. Jensen, Romina Nassini, Romke Bron, and Carlos U. Corvera
- Subjects
medicine.medical_specialty ,Primary Cell Culture ,TRPV1 ,Neuropeptide ,In situ hybridization ,Article ,Receptors, G-Protein-Coupled ,Bile Acids and Salts ,Xenopus laevis ,Transient Receptor Potential Channels ,Internal medicine ,Ganglia, Spinal ,medicine ,Animals ,Humans ,Neurons, Afferent ,Receptor ,Natriuretic Peptides ,TRPA1 Cation Channel ,Cholestatic pruritus ,Mice, Knockout ,Cholestasis ,Hepatology ,Chemistry ,Pruritus ,Gastroenterology ,food and beverages ,Nociceptors ,medicine.disease ,G protein-coupled bile acid receptor ,Disease Models, Animal ,Endocrinology ,HEK293 Cells ,Gastrin-Releasing Peptide ,Nociceptor ,Oocytes ,Signal transduction ,psychological phenomena and processes - Abstract
Background & Aims Patients with cholestatic disease have increased systemic concentrations of bile acids (BAs) and profound pruritus. The G-protein–coupled BA receptor 1 TGR5 (encoded by GPBAR1 ) is expressed by primary sensory neurons; its activation induces neuronal hyperexcitability and scratching by unknown mechanisms. We investigated whether the transient receptor potential ankyrin 1 (TRPA1) is involved in BA-evoked, TGR5-dependent pruritus in mice. Methods Co-expression of TGR5 and TRPA1 in cutaneous afferent neurons isolated from mice was analyzed by immunofluorescence, in situ hybridization, and single-cell polymerase chain reaction. TGR5-induced activation of TRPA1 was studied in in HEK293 cells, Xenopus laevis oocytes, and primary sensory neurons by measuring Ca 2+ signals. The contribution of TRPA1 to TGR5-induced release of pruritogenic neuropeptides, activation of spinal neurons, and scratching behavior were studied using TRPA1 antagonists or Trpa1 −/− mice. Results TGR5 and TRPA1 protein and messenger RNA were expressed by cutaneous afferent neurons. In HEK cells, oocytes, and neurons co-expressing TGR5 and TRPA1, BAs caused TGR5-dependent activation and sensitization of TRPA1 by mechanisms that required Gβγ, protein kinase C, and Ca 2+ . Antagonists or deletion of TRPA1 prevented BA-stimulated release of the pruritogenic neuropeptides gastrin-releasing peptide and atrial natriuretic peptide B in the spinal cord. Disruption of Trpa1 in mice blocked BA-induced expression of Fos in spinal neurons and prevented BA-stimulated scratching. Spontaneous scratching was exacerbated in transgenic mice that overexpressed TRG5. Administration of a TRPA1 antagonist or the BA sequestrant colestipol, which lowered circulating levels of BAs, prevented exacerbated spontaneous scratching in TGR5 overexpressing mice. Conclusions BAs induce pruritus in mice by co-activation of TGR5 and TRPA1. Antagonists of TGR5 and TRPA1, or inhibitors of the signaling mechanism by which TGR5 activates TRPA1, might be developed for treatment of cholestatic pruritus.
- Published
- 2014
32. Fine-tuning of GPCR signals by intracellular G protein modulators
- Author
-
Peishen, Zhao, Wendy, Cladman, Hubert H M, Van Tol, and Peter, Chidiac
- Subjects
GTP-Binding Proteins ,Intracellular Space ,Animals ,Humans ,Models, Biological ,Protein Binding ,Receptors, G-Protein-Coupled ,Signal Transduction - Abstract
Heterotrimeric G proteins convey receptor signals to intracellular effectors. Superimposed over the basic GPCR-G protein-effector scheme are three types of auxiliary proteins that also modulate Gα. Regulator of G protein signaling proteins and G protein signaling modifier proteins respectively promote GTPase activity and hinder GTP/GDP exchange to limit Gα activation. There are also diverse proteins that, like GPCRs, can promote nucleotide exchange and thus activation. Here we review the impact of these auxiliary proteins on GPCR signaling. Although their precise physiological functions are not yet clear, all of them can produce significant effects in experimental systems. These signaling changes are generally consistent with established effects on isolated Gα; however, the activation state of Gα is seldom verified and many such changes appear also to reflect the physical disruption of or indirect effects on interactions between Gα and its associated GPCR, Gβγ, and/or effector.
- Published
- 2013
33. Fine-Tuning of GPCR Signals by Intracellular G Protein Modulators
- Author
-
Hubert H.M. Van Tol, Peishen Zhao, Wendy Cladman, and Peter Chidiac
- Subjects
G beta-gamma complex ,Regulator of G protein signaling ,GTPase-activating protein ,G protein ,Heterotrimeric G protein ,GTPase ,Biology ,G protein-coupled receptor ,Cell biology ,Protein–protein interaction - Abstract
Heterotrimeric G proteins convey receptor signals to intracellular effectors. Superimposed over the basic GPCR–G protein–effector scheme are three types of auxiliary proteins that also modulate Gα. Regulator of G protein signaling proteins and G protein signaling modifier proteins respectively promote GTPase activity and hinder GTP/GDP exchange to limit Gα activation. There are also diverse proteins that, like GPCRs, can promote nucleotide exchange and thus activation. Here we review the impact of these auxiliary proteins on GPCR signaling. Although their precise physiological functions are not yet clear, all of them can produce significant effects in experimental systems. These signaling changes are generally consistent with established effects on isolated Gα; however, the activation state of Gα is seldom verified and many such changes appear also to reflect the physical disruption of or indirect effects on interactions between Gα and its associated GPCR, Gβγ, and/or effector.
- Published
- 2013
- Full Text
- View/download PDF
34. 431 Protease-Activated Receptor 2 Signals from Endosomes to Control Sensitivity of Nociceptors
- Author
-
Peishen Zhao, Stephen Vanner, Nigel W. Bunnett, Luke A. Pattison, and Nestor N. Jiménez-Vargas
- Subjects
Hepatology ,Chemistry ,Endosome ,Gastroenterology ,Nociceptor ,Sensitivity (control systems) ,Protease-activated receptor 2 ,Cell biology - Published
- 2016
- Full Text
- View/download PDF
35. 562 Protein Kinase D and Gβγ Mediate Protease-Biased Translocation of Protease-activated Receptor-2 from the Golgi Apparatus to the Plasma Membrane
- Author
-
Peishen Zhao, Tina Marie Lieu, Dane D. Jensen, Daniel P. Poole, and Nigel W. Bunnett
- Subjects
Protease ,Hepatology ,Chemistry ,medicine.medical_treatment ,Gastroenterology ,Chromosomal translocation ,Golgi apparatus ,Cell biology ,symbols.namesake ,Membrane ,symbols ,medicine ,PROTEIN KINASE D ,Protease-activated receptor 2 - Published
- 2016
- Full Text
- View/download PDF
36. Resistance to age-related, normal body weight gain in RGS2 deficient mice
- Author
-
Caroline Nunn, Kelly M Summers, Peishen Zhao, Min Xu Zou, Peter Chidiac, and Christopher G. Guglielmo
- Subjects
Male ,medicine.medical_specialty ,Aging ,Adipocytes, White ,Adipose tissue ,White adipose tissue ,Biology ,Weight Gain ,Glucagon ,Mice ,Internal medicine ,Enhancer binding ,Brown adipose tissue ,Receptors, Adrenergic, beta ,medicine ,CCAAT-Enhancer-Binding Protein-alpha ,Lipolysis ,Animals ,Insulin ,Mice, Knockout ,Leptin ,Cell Biology ,Cold Temperature ,Mice, Inbred C57BL ,PPAR gamma ,Lipoprotein Lipase ,Endocrinology ,medicine.anatomical_structure ,Adipocytes, Brown ,Glucose ,Adipogenesis ,Hepatocytes ,Female ,Energy Metabolism ,RGS Proteins ,Signal Transduction - Abstract
RGS2 (regulator of G protein signaling 2) is known to limit signals mediated via Gq- and Gs-coupled GPCRs (G protein coupled receptors), and it has been implicated in the differentiation of several cells types. The physiology of RGS2 knockout mice ( rgs2 −/− ) has been studied in some detail, however, a metabolic phenotype has not previously been reported. We observed that old (21–24 month) rgs2 −/− mice weigh much less than wild-type C57BL/6 controls, and exhibit greatly reduced fat deposits, decreased serum lipids, and low leptin levels. Lower weight was evident as early as four weeks and continued throughout life. Younger adult male rgs2 −/− mice (4–8 months) were found to show similar strain-related differences as the aged animals, as well improved glucose clearance and insulin sensitivity, and enhanced beta-adrenergic and glucagon signaling in isolated hepatocytes. In addition, rgs2 −/− pre-adipocytes had reduced levels of differentiation markers (Peroxisome proliferator-activated receptor γ (PPARγ); lipoprotein lipase (Lpl); CCAAT/enhancer binding protein α (CEBPα)) and also rgs2 −/− white adipocytes were small relative to controls, suggesting altered adipogenesis. In wild-type animals, RGS2 mRNA was decreased in brown adipose tissue after cold exposure (7 h at 4 °C) but increased in white adipose tissue in response to a high fat diet, also suggesting a role in lipid storage. No differences between strains were detected with respect to food intake, energy expenditure, GPCR-stimulated lipolysis, or adaptive thermogenesis. In conclusion this study points to RGS2 as being an important regulatory factor in controlling body weight and adipose function.
- Published
- 2010
37. Translational control by RGS2
- Author
-
Chau Hoang Nguyen, Hong Ming, Robert Gros, Scot R. Kimball, Peter Chidiac, Peishen Zhao, and Lynne Hugendubler
- Subjects
Male ,GTPase-activating protein ,Molecular Sequence Data ,GTPase ,Biology ,Article ,Cell Line ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Regulator of G protein signaling ,GTP-binding protein regulators ,Protein biosynthesis ,Animals ,Humans ,Amino Acid Sequence ,RGS2 ,Research Articles ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Colforsin ,Cell Biology ,RGS17 ,Molecular biology ,Cell biology ,Protein Structure, Tertiary ,Eukaryotic Initiation Factor-2B ,Gene Expression Regulation ,Protein Biosynthesis ,RGS Proteins ,Sequence Alignment ,030217 neurology & neurosurgery - Abstract
A regulator of G protein signaling, RGS2, moonlights in protein synthesis control., The regulator of G protein signaling (RGS) proteins are a family of guanosine triphosphatase (GTPase)–accelerating proteins. We have discovered a novel function for RGS2 in the control of protein synthesis. RGS2 was found to bind to eIF2Bϵ (eukaryotic initiation factor 2B ϵ subunit) and inhibit the translation of messenger RNA (mRNA) into new protein. This effect was not observed for other RGS proteins tested. This novel function of RGS2 is distinct from its ability to regulate G protein–mediated signals and maps to a stretch of 37 amino acid residues within its conserved RGS domain. Moreover, RGS2 was capable of interfering with the eIF2–eIF2B GTPase cycle, which is a requisite step for the initiation of mRNA translation. Collectively, this study has identified a novel role for RGS2 in the control of protein synthesis that is independent of its established RGS domain function.
- Published
- 2009
38. Su1892 Cystatin C Attenuates Cathepsin S-Evoked and PAR2-Mediated Inflammation and Pain
- Author
-
Justine Mintern, Nigel W. Bunnett, Nicholas Barlow, Peishen Zhao, Tina Marie Lieu, and Erik Lindström
- Subjects
medicine.medical_specialty ,Hepatology ,biology ,business.industry ,Gastroenterology ,Inflammation ,Endocrinology ,Cystatin C ,Internal medicine ,medicine ,biology.protein ,medicine.symptom ,business ,Cathepsin S - Published
- 2015
- Full Text
- View/download PDF
39. 519 Neutrophil Elastase Activates a PAR2/TRPV4 Signaling Complex to Cause Inflammatory Pain
- Author
-
Peishen Zhao, Tina Marie Lieu, Stephen Vanner, and Nigel W. Bunnett
- Subjects
TRPV4 ,Hepatology ,biology ,business.industry ,Neutrophil elastase ,Immunology ,Gastroenterology ,biology.protein ,Medicine ,business ,Inflammatory pain - Published
- 2015
- Full Text
- View/download PDF
40. Tu1963 The Bile Acid Receptor TGR5 Sensitizes the TRPA1 Channel to Induce Cholestatic Itch
- Author
-
Megan S. Grace, Peter McIntyre, Romke Bron, Dane D. Jensen, Daniel P. Poole, Nigel W. Bunnett, Peishen Zhao, Gihan Jayaweera, and Tina Marie Lieu
- Subjects
medicine.medical_specialty ,Endocrinology ,Hepatology ,Chemistry ,Internal medicine ,Gastroenterology ,medicine ,TRPA1 Channel ,G protein-coupled bile acid receptor - Published
- 2014
- Full Text
- View/download PDF
41. 400 Cathepsin S Induces Inflammation and Pain via Biased Agonism of PAR2 and TRPV4
- Author
-
Erik Lindström, Silvia Sostegni, Matthew C. Metcalf, Dane D. Jensen, Nicholas A. Veldhuis, Nicholas Barlow, Martina Kocan, Tina Marie Lieu, Silke Haerteis, Peishen Zhao, Nigel W. Bunnett, and Christoph Korbmacher
- Subjects
TRPV4 ,medicine.medical_specialty ,Endocrinology ,Hepatology ,Chemistry ,Internal medicine ,Gastroenterology ,medicine ,Functional selectivity ,Inflammation ,medicine.symptom ,Cathepsin S - Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.