1
|
Rajala R, Griffin CT. Endothelial protease-activated receptor 4: impotent or important? Front Cardiovasc Med 2025; 12:1541879. [PMID: 39935714 PMCID: PMC11810968 DOI: 10.3389/fcvm.2025.1541879] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025] Open
Abstract
The protease thrombin, which increases its levels with various pathologies, can signal through the G protein-coupled receptors protease-activated receptors 1 and 4 (PAR1/PAR4). PAR1 is a high-affinity receptor for thrombin, whereas PAR4 is a low-affinity receptor. Finding functions for PAR4 in endothelial cells (ECs) has been an elusive goal over the last two decades. Several studies have demonstrated a lack of functionality for PAR4 in ECs, with many claiming that PAR4 function is confined mostly to platelets. A recent study from our lab identified low expressing but functional PAR4 in hepatic ECs in vivo. We also found that PAR4 likely has a higher signaling potency than PAR1. Given this potency, ECs seem to limit PAR4 signaling except for extreme cases. As a result, we claim PAR4 is not an impotent receptor because it is low expressing, but rather PAR4 is low expressing because it is a very potent receptor. Since we have finally shown PAR4 to be present and functional on ECs in vivo, it is important to outline why such controversy arose over the last two decades and, more importantly, why the receptor was undervalued on ECs. This timely review aims to inspire investigators in the field of vascular biology to study the regulatory aspect of endothelial PAR4 and its relationship with the more highly expressed PAR1.
Collapse
Affiliation(s)
- Rahul Rajala
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Courtney T. Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
2
|
Rajala R, Cleuren ACA, Griffin CT. Acetaminophen Overdose Reveals PAR4 as a Low-Expressing but Potent Receptor on the Hepatic Endothelium in Mice. Arterioscler Thromb Vasc Biol 2025; 45:53-71. [PMID: 39360412 DOI: 10.1161/atvbaha.124.321353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND The protease thrombin, which elicits multiple physiological and pathological effects on vascular endothelial cells (ECs), can signal through PARs (protease-activated receptors) 1 and 4. PAR1 is a high-affinity thrombin receptor known to signal on ECs, whereas PAR4 is a low-affinity thrombin receptor, and evidence for its expression and function on ECs is mixed. This study aims to exploit the high levels of thrombin generation and hepatic vascular dysfunction that occur during acetaminophen (APAP) overdose to determine (1) whether hepatic endothelial PAR4 is a functional receptor, and (2) the endothelial-specific functions for PAR1 and PAR4 in a high thrombin and pathological setting. METHODS We generated mice with conditional deletion of Par1/Par4 in ECs and overdosed them with APAP. Hepatic vascular permeability, erythrocyte accumulation in the liver, thrombin generation, and liver function were assessed following overdose. Additionally, we investigated the expression levels of endothelial PARs and how they influence transcription in APAP-overdosed liver ECs using endothelial translating ribosome affinity purification followed by next-generation sequencing. RESULTS We found that mice deficient in high-expressing endothelial Par1 or low-expressing Par4 had equivalent reductions in APAP-induced hepatic vascular instability, although mice deficient for both receptors had lower vascular permeability at an earlier timepoint after APAP overdose than either of the single mutants. Additionally, mice with loss of both endothelial Par1 and Par4 had reduced thrombin generation after APAP overdose, suggesting decreased hypercoagulability. Last, we found that endothelial PAR1-but not PAR4-can regulate transcription in hepatic ECs. CONCLUSIONS Low-expressing PAR4 can react similarly to high-expressing PAR1 in APAP-overdosed hepatic ECs, demonstrating that PAR4 is a potent thrombin receptor. Additionally, these receptors are functionally redundant but act divergently in their expression and ability to influence transcription in hepatic ECs.
Collapse
MESH Headings
- Animals
- Acetaminophen/toxicity
- Receptors, Thrombin/metabolism
- Receptors, Thrombin/genetics
- Liver/metabolism
- Liver/drug effects
- Liver/pathology
- Receptor, PAR-1/metabolism
- Receptor, PAR-1/genetics
- Receptor, PAR-1/deficiency
- Thrombin/metabolism
- Endothelial Cells/metabolism
- Endothelial Cells/drug effects
- Endothelial Cells/pathology
- Mice, Knockout
- Mice, Inbred C57BL
- Disease Models, Animal
- Capillary Permeability/drug effects
- Male
- Drug Overdose/metabolism
- Signal Transduction
- Mice
- Chemical and Drug Induced Liver Injury/metabolism
- Chemical and Drug Induced Liver Injury/pathology
- Chemical and Drug Induced Liver Injury/genetics
- Chemical and Drug Induced Liver Injury/etiology
- Cells, Cultured
- Receptors, Proteinase-Activated
Collapse
Affiliation(s)
- Rahul Rajala
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City (R.R., A.C.A.C., C.T.G.)
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City (R.R., A.C.A.C., C.T.G.)
- Harold Hamm Diabetes Center, Oklahoma City, OK (R.R.)
| | - Audrey C A Cleuren
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City (R.R., A.C.A.C., C.T.G.)
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City (R.R., A.C.A.C., C.T.G.)
| | - Courtney T Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City (R.R., A.C.A.C., C.T.G.)
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City (R.R., A.C.A.C., C.T.G.)
| |
Collapse
|
3
|
Kendrick NC, Nieman MT. Hidden Power: PAR4's Role in Liver Damage From Acetaminophen Overdose in Mice. Arterioscler Thromb Vasc Biol 2025; 45:72-73. [PMID: 39540282 DOI: 10.1161/atvbaha.124.321881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Affiliation(s)
- NaShea C Kendrick
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Marvin T Nieman
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
4
|
Rajala R, Cleuren AC, Griffin CT. Acetaminophen Overdose Reveals Protease-Activated Receptor 4 as a Low-Expressing but Potent Receptor on the Hepatic Endothelium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.07.598028. [PMID: 38895465 PMCID: PMC11185779 DOI: 10.1101/2024.06.07.598028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Background & Aims Hepatic endothelial cell (EC) dysfunction and centrilobular hepatocyte necrosis occur with acetaminophen (APAP) overdose. The protease thrombin, which is acutely generated during APAP overdose, can signal through protease-activated receptors 1 and 4 (PAR1/PAR4). PAR1 is a high-affinity thrombin receptor that is known to signal on ECs, whereas PAR4 is a low-affinity thrombin receptor, and evidence for its expression and function on ECs is mixed. This study aims to exploit the high levels of thrombin generated during APAP overdose to determine (1) if hepatic endothelial PAR4 is a functional receptor, and (2) endothelial-specific functions for PAR1 and PAR4 in a high thrombin setting. Methods We generated mice with conditional deletion(s) of Par1/Par4 in ECs and overdosed them with APAP. Hepatic vascular permeability, erythrocyte congestion/bleeding, and liver function were assessed following overdose. Additionally, we investigated the expression levels of endothelial PARs and how they influence transcription in APAP-overdosed liver ECs using endothelial Translating Ribosome Affinity Purification followed by next-generation sequencing (TRAPseq). Results We found that mice deficient in high-expressing endothelial Par1 or low-expressing Par4 had equivalent reductions in APAP-induced hepatic vascular instability but no effect on hepatocyte necrosis. Additionally, mice with loss of endothelial Par1 and Par4 had reduced permeability at an earlier time point after APAP overdose when compared to mice singly deficient in either receptor in ECs. We also found that endothelial PAR1-but not PAR4-can regulate transcription in hepatic ECs. Conclusions Low-expressing PAR4 can react similarly to high-expressing PAR1 in APAP-overdosed hepatic ECs, demonstrating that PAR4 is a potent thrombin receptor. Additionally, these receptors are functionally redundant but act divergently in their expression and ability to influence transcription in hepatic ECs.
Collapse
Affiliation(s)
- Rahul Rajala
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
- Harold Hamm Diabetes Center, Oklahoma City, OK
| | - Audrey C.A. Cleuren
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Courtney T. Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
5
|
Blockade of L-type Ca 2+ channel attenuates doxorubicin-induced cardiomyopathy via suppression of CaMKII-NF-κB pathway. Sci Rep 2019; 9:9850. [PMID: 31285514 PMCID: PMC6614470 DOI: 10.1038/s41598-019-46367-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 06/27/2019] [Indexed: 12/30/2022] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) and nuclear factor-kappa B (NF-κB) play crucial roles in pathogenesis of doxorubicin (DOX)-induced cardiomyopathy. Their activities are regulated by intracellular Ca2+. We hypothesized that blockade of L-type Ca2+ channel (LTCC) could attenuate DOX-induced cardiomyopathy by regulating CaMKII and NF-κB. DOX activated CaMKII and NF-κB through their phosphorylation and increased cleaved caspase 3 in cardiomyocytes. Pharmacological blockade or gene knockdown of LTCC by nifedipine or small interfering RNA, respectively, suppressed DOX-induced phosphorylation of CaMKII and NF-κB and apoptosis in cardiomyocytes, accompanied by decreasing intracellular Ca2+ concentration. Autocamtide 2-related inhibitory peptide (AIP), a selective CaMKII inhibitor, inhibited DOX-induced phosphorylation of NF-κB and cardiomyocyte apoptosis. Inhibition of NF-κB activity by ammonium pyrrolidinedithiocarbamate (PDTC) suppressed DOX-induced cardiomyocyte apoptosis. DOX-treatment (18 mg/kg via intravenous 3 injections over 1 week) increased phosphorylation of CaMKII and NF-κB in mouse hearts. Nifedipine (10 mg/kg/day) significantly suppressed DOX-induced phosphorylation of CaMKII and NF-κB and cardiomyocyte injury and apoptosis in mouse hearts. Moreover, it attenuated DOX-induced left ventricular dysfunction and dilatation. Our findings suggest that blockade of LTCC attenuates DOX-induced cardiomyocyte apoptosis via suppressing intracellular Ca2+ elevation and activation of CaMKII-NF-κB pathway. LTCC blockers might be potential therapeutic agents against DOX-induced cardiomyopathy.
Collapse
|
6
|
Liu W, Hashimoto T, Yamashita T, Hirano K. Coagulation factor XI induces Ca 2+ response and accelerates cell migration in vascular smooth muscle cells via proteinase-activated receptor 1. Am J Physiol Cell Physiol 2019; 316:C377-C392. [PMID: 30566391 DOI: 10.1152/ajpcell.00426.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Activated coagulation factor XI (FXIa) is a serine proteinase that plays a key role in the intrinsic coagulation pathway. The analysis of FXI-knockout mice has indicated the contribution of FXI to the pathogenesis of atherosclerosis. However, the underlying mechanism remains unknown. We hypothesized that FXIa exerts vascular smooth muscle effects via proteinase-activated receptor 1 (PAR1). Fura-2 fluorometry revealed that FXIa elicited intracellular Ca2+ signal in rat embryo aorta smooth muscle A7r5 cells. The influx of extracellular Ca2+ played a greater role in generating Ca2+ signal than the Ca2+ release from intracellular stores. The FXIa-induced Ca2+ signal was abolished by the pretreatment with atopaxar, an antagonist of PAR1, or 4-amidinophenylmethanesulfonyl fluoride (p-APMSF), an inhibitor of proteinase, while it was also lost in embryonic fibroblasts derived from PAR1-/- mice. FXIa cleaved the recombinant protein containing the extracellular region of PAR1 at the same site (R45/S46) as that of thrombin, a canonical PAR1 agonist. The FXIa-induced Ca2+ influx was inhibited by diltiazem, an L-type Ca2+ channel blocker, and by siRNA targeted to CaV1.2. The FXIa-induced Ca2+ influx was also inhibited by GF109203X and rottlerin, inhibitors of protein kinase C. In a wound healing assay, FXIa increased the rate of cell migration by 2.46-fold of control, which was partly inhibited by atopaxar or diltiazem. In conclusion, FXIa mainly elicits the Ca2+ signal via the PAR1/CaV1.2-mediated Ca2+ influx and accelerates the migration in vascular smooth muscle cells. The present study provides the first evidence that FXIa exerts a direct cellular effect on vascular smooth muscle.
Collapse
Affiliation(s)
- Wenhua Liu
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University , Kagawa , Japan
| | - Takeshi Hashimoto
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University , Kagawa , Japan
| | - Tetsuo Yamashita
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University , Kagawa , Japan
| | - Katsuya Hirano
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University , Kagawa , Japan
| |
Collapse
|
7
|
Slezak J, Kura B, Babal P, Barancik M, Ferko M, Frimmel K, Kalocayova B, Kukreja RC, Lazou A, Mezesova L, Okruhlicova L, Ravingerova T, Singal PK, Szeiffova Bacova B, Viczenczova C, Vrbjar N, Tribulova N. Potential markers and metabolic processes involved in the mechanism of radiation-induced heart injury. Can J Physiol Pharmacol 2017; 95:1190-1203. [PMID: 28750189 DOI: 10.1139/cjpp-2017-0121] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Irradiation of normal tissues leads to acute increase in reactive oxygen/nitrogen species that serve as intra- and inter-cellular signaling to alter cell and tissue function. In the case of chest irradiation, it can affect the heart, blood vessels, and lungs, with consequent tissue remodelation and adverse side effects and symptoms. This complex process is orchestrated by a large number of interacting molecular signals, including cytokines, chemokines, and growth factors. Inflammation, endothelial cell dysfunction, thrombogenesis, organ dysfunction, and ultimate failing of the heart occur as a pathological entity - "radiation-induced heart disease" (RIHD) that is major source of morbidity and mortality. The purpose of this review is to bring insights into the basic mechanisms of RIHD that may lead to the identification of targets for intervention in the radiotherapy side effect. Studies of authors also provide knowledge about how to select targeted drugs or biological molecules to modify the progression of radiation damage in the heart. New prospective studies are needed to validate that assessed factors and changes are useful as early markers of cardiac damage.
Collapse
Affiliation(s)
- Jan Slezak
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Branislav Kura
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Pavel Babal
- b Institute of Pathology, Medical Faculty of Comenius University, Bratislava, Slovakia
| | - Miroslav Barancik
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Miroslav Ferko
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Karel Frimmel
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Barbora Kalocayova
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Rakesh C Kukreja
- c Division of Cardiology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, USA
| | - Antigone Lazou
- d School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Lucia Mezesova
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Ludmila Okruhlicova
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Tanya Ravingerova
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Pawan K Singal
- e University of Manitoba, St. Boniface Research Centre, Winnipeg, MB R2H 2A6, Canada
| | | | - Csilla Viczenczova
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Norbert Vrbjar
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| | - Narcis Tribulova
- a Institute for Heart Research, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| |
Collapse
|
8
|
Ramachandran R, Mihara K, Thibeault P, Vanderboor CM, Petri B, Saifeddine M, Bouvier M, Hollenberg MD. Targeting a Proteinase-Activated Receptor 4 (PAR4) Carboxyl Terminal Motif to Regulate Platelet Function. Mol Pharmacol 2017; 91:287-295. [PMID: 28126849 DOI: 10.1124/mol.116.106526] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 01/18/2017] [Indexed: 12/22/2022] Open
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 β-arrestin interactions. By disrupting this PAR4 calcium/β-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.
Collapse
Affiliation(s)
- Rithwik Ramachandran
- Snyder Institute for Chronic Diseases and Department of Physiology and Pharmacology (R.R., K.M., M.S., M.D.H.), Mouse Phenomics Resource Laboratory, Snyder Institute for Chronic Diseases and Department of Microbiology, Immunology, and Infectious Diseases (B.P.), and Department of Medicine (M.D.H.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada (R.R., P.T., C.M.V.); and IRIC-Université de Montréal, Montréal, Québec, Canada (M.B.)
| | - Koichiro Mihara
- Snyder Institute for Chronic Diseases and Department of Physiology and Pharmacology (R.R., K.M., M.S., M.D.H.), Mouse Phenomics Resource Laboratory, Snyder Institute for Chronic Diseases and Department of Microbiology, Immunology, and Infectious Diseases (B.P.), and Department of Medicine (M.D.H.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada (R.R., P.T., C.M.V.); and IRIC-Université de Montréal, Montréal, Québec, Canada (M.B.)
| | - Pierre Thibeault
- Snyder Institute for Chronic Diseases and Department of Physiology and Pharmacology (R.R., K.M., M.S., M.D.H.), Mouse Phenomics Resource Laboratory, Snyder Institute for Chronic Diseases and Department of Microbiology, Immunology, and Infectious Diseases (B.P.), and Department of Medicine (M.D.H.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada (R.R., P.T., C.M.V.); and IRIC-Université de Montréal, Montréal, Québec, Canada (M.B.)
| | - Christina M Vanderboor
- Snyder Institute for Chronic Diseases and Department of Physiology and Pharmacology (R.R., K.M., M.S., M.D.H.), Mouse Phenomics Resource Laboratory, Snyder Institute for Chronic Diseases and Department of Microbiology, Immunology, and Infectious Diseases (B.P.), and Department of Medicine (M.D.H.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada (R.R., P.T., C.M.V.); and IRIC-Université de Montréal, Montréal, Québec, Canada (M.B.)
| | - Björn Petri
- Snyder Institute for Chronic Diseases and Department of Physiology and Pharmacology (R.R., K.M., M.S., M.D.H.), Mouse Phenomics Resource Laboratory, Snyder Institute for Chronic Diseases and Department of Microbiology, Immunology, and Infectious Diseases (B.P.), and Department of Medicine (M.D.H.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada (R.R., P.T., C.M.V.); and IRIC-Université de Montréal, Montréal, Québec, Canada (M.B.)
| | - Mahmoud Saifeddine
- Snyder Institute for Chronic Diseases and Department of Physiology and Pharmacology (R.R., K.M., M.S., M.D.H.), Mouse Phenomics Resource Laboratory, Snyder Institute for Chronic Diseases and Department of Microbiology, Immunology, and Infectious Diseases (B.P.), and Department of Medicine (M.D.H.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada (R.R., P.T., C.M.V.); and IRIC-Université de Montréal, Montréal, Québec, Canada (M.B.)
| | - Michel Bouvier
- Snyder Institute for Chronic Diseases and Department of Physiology and Pharmacology (R.R., K.M., M.S., M.D.H.), Mouse Phenomics Resource Laboratory, Snyder Institute for Chronic Diseases and Department of Microbiology, Immunology, and Infectious Diseases (B.P.), and Department of Medicine (M.D.H.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada (R.R., P.T., C.M.V.); and IRIC-Université de Montréal, Montréal, Québec, Canada (M.B.)
| | - Morley D Hollenberg
- Snyder Institute for Chronic Diseases and Department of Physiology and Pharmacology (R.R., K.M., M.S., M.D.H.), Mouse Phenomics Resource Laboratory, Snyder Institute for Chronic Diseases and Department of Microbiology, Immunology, and Infectious Diseases (B.P.), and Department of Medicine (M.D.H.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada (R.R., P.T., C.M.V.); and IRIC-Université de Montréal, Montréal, Québec, Canada (M.B.)
| |
Collapse
|
9
|
Arai S, Ikeda M, Ide T, Matsuo Y, Fujino T, Hirano K, Sunagawa K, Tsutsui H. Functional loss of DHRS7C induces intracellular Ca2+ overload and myotube enlargement in C2C12 cells via calpain activation. Am J Physiol Cell Physiol 2017; 312:C29-C39. [DOI: 10.1152/ajpcell.00090.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 10/17/2016] [Indexed: 02/03/2023]
Abstract
Dehydrogenase/reductase member 7C (DHRS7C) is a newly identified NAD/NADH-dependent dehydrogenase that is expressed in cardiac and skeletal muscle and localized in the endoplasmic/sarcoplasmic reticulum (ER/SR). However, its functional role in muscle cells remains to be fully elucidated. Here, we investigated the role of DHRS7C by analyzing mouse C2C12 myoblasts deficient in DHRS7C (DHRS7C-KO cells), overexpressing wild-type DHRS7C (DHRS7C-WT cells), or expressing mutant DHRS7C [DHRS7C-Y191F or DHRS7C-K195Q cells, harboring point mutations in the NAD/NADH-dependent dehydrogenase catalytic core domain (YXXXK)]. DHRS7C expression was induced as C2C12 myoblasts differentiated into mature myotubes, whereas DHRS7C-KO myotubes exhibited enlarged cellular morphology after differentiation. Notably, both DHRS7C-Y191F and DHRS7C-K195Q cells also showed similar enlarged cellular morphology, suggesting that the NAD/NADH-dependent dehydrogenase catalytic core domain is pivotal for DHRS7C function. In DHRS7C-KO, DHRS7C-Y191F, and DHRS7C-K195Q cells, the resting level of cytosolic Ca2+ and total amount of Ca2+ storage in the ER/SR were significantly higher than those in control C2C12 and DHRS7C-WT cells after differentiation. Additionally, Ca2+ release from the ER/SR induced by thapsigargin and 4-chloro-m-cresol was augmented in these cells and calpain, a calcium-dependent protease, was significantly activated in DHRS7C-KO, DHRS7C-Y191F, and DHRS7C-K195Q myotubes, consistent with the higher resting level of cytosolic Ca2+ concentration and enlarged morphology after differentiation. Furthermore, treatment with a calpain inhibitor abolished the enlarged cellular morphology. Taken together, our findings suggested that DHRS7C maintains intracellular Ca2+ homeostasis involving the ER/SR and that functional loss of DHRS7C leads to Ca2+ overload in the cytosol and ER/SR, resulting in enlarged cellular morphology via calpain activation.
Collapse
Affiliation(s)
- Shinobu Arai
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masataka Ikeda
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomomi Ide
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuka Matsuo
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeo Fujino
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Katsuya Hirano
- Department of Cardiovascular Physiology Faculty of Medicine, Kagawa University, Kagawa, Japan; and
| | - Kenji Sunagawa
- Department of Therapeutic Regulation of Cardiovascular Homeostasis, Center for Disruptive Cardiovascular Medicine, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
10
|
Tillery LC, Epperson TA, Eguchi S, Motley ED. Featured Article: Differential regulation of endothelial nitric oxide synthase phosphorylation by protease-activated receptors in adult human endothelial cells. Exp Biol Med (Maywood) 2016; 241:569-80. [PMID: 26729042 DOI: 10.1177/1535370215622584] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 11/06/2015] [Indexed: 11/17/2022] Open
Abstract
Protease-activated receptors have been shown to regulate endothelial nitric oxide synthase through the phosphorylation of specific sites on the enzyme. It has been established that PAR-2 activation phosphorylates eNOS-Ser-1177 and leads to the production of the potent vasodilator nitric oxide, while PAR-1 activation phosphorylates eNOS-Thr-495 and decreases nitric oxide production in human umbilical vein endothelial cells. In this study, we hypothesize a differential coupling of protease-activated receptors to the signaling pathways that regulates endothelial nitric oxide synthase and nitric oxide production in primary adult human coronary artery endothelial cells. Using Western Blot analysis, we showed that thrombin and the PAR-1 activating peptide, TFLLR, lead to the phosphorylation of eNOS-Ser-1177 in human coronary artery endothelial cells, which was blocked by SCH-79797 (SCH), a PAR-1 inhibitor. Using the nitrate/nitrite assay, we also demonstrated that the thrombin- and TFLLR-induced production of nitric oxide was inhibited by SCH and L-NAME, a NOS inhibitor. In addition, we observed that TFLLR, unlike thrombin, significantly phosphorylated eNOS-Thr-495, which may explain the observed delay in nitric oxide production in comparison to that of thrombin. Activation of PAR-2 by SLIGRL, a PAR-2 specific ligand, leads to dual phosphorylation of both catalytic sites but primarily regulated eNOS-Thr-495 phosphorylation with no change in nitric oxide production in human coronary artery endothelial cells. PAR-3, known as the non-signaling receptor, was activated by TFRGAP, a PAR-3 mimicking peptide, and significantly induced the phosphorylation of eNOS-Thr-495 with minimal phosphorylation of eNOS-Ser-1177 with no change in nitric oxide production. In addition, we confirmed that PAR-mediated eNOS-Ser-1177 phosphorylation was Ca(2+)-dependent using the Ca(2+) chelator, BAPTA, while eNOS-Thr-495 phosphorylation was mediated via Rho kinase using the ROCK inhibitor, Y-27632, suggesting protease-activated receptor coupling to Gq and G12/13, respectively. These data suggest a vascular bed specific differential coupling of protease-activated receptors to the signaling pathways that regulate endothelial nitric oxide synthase and nitric oxide production that may be responsible for endothelial dysfunction associated with cardiovascular disease.
Collapse
Affiliation(s)
- Lakeisha C Tillery
- Department of Microbiology and Immunology, Meharry Medical College, Nashville, TN 37208, USA
| | - Tenille A Epperson
- Department of Physiology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Satoru Eguchi
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Evangeline D Motley
- Department of Physiology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| |
Collapse
|
11
|
Bao Y, Gao Y, Yang L, Kong X, Zheng H, Hou W, Hua B. New insights into protease-activated receptor 4 signaling pathways in the pathogenesis of inflammation and neuropathic pain: a literature review. Channels (Austin) 2015; 9:5-13. [PMID: 25664811 DOI: 10.4161/19336950.2014.995001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Pain is an unpleasant sensory and emotional experience that is commonly associated with actual or potential tissue damage. Despite decades of pain research, many patients continue to suffer from chronic pain that is refractory to current treatments. Accumulating evidence has indicated an important role of protease-activated receptor 4 (PAR4) in the pathogenesis of inflammation and neuropathic pain. Here we reviewed PAR4 expression and activation via intracellular signaling pathways and the role of PAR4 signaling pathways in the development and maintenance of pain. Understanding PAR4 and its corresponding signaling pathways will provide insight to further explore the molecular basis of pain, which will also help to identify new targets for pharmacological intervention for pain relief.
Collapse
Affiliation(s)
- Yanju Bao
- a Department of Oncology ; Guang'anmen Hospital ; China Academy of Chinese Medical Sciences; Beixiange 5 ; Xicheng District , Beijing , P. R. China
| | | | | | | | | | | | | |
Collapse
|
12
|
Fu Q, Cheng J, Gao Y, Zhang Y, Chen X, Xie J. Protease-activated receptor 4: a critical participator in inflammatory response. Inflammation 2015; 38:886-95. [PMID: 25120239 DOI: 10.1007/s10753-014-9999-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Protease-activated receptors (PARs) are G protein-coupled receptors of which four members PAR1, PAR2, PAR3, and PAR4 have been identified, characterized by a typical mechanism of activation involving various related proteases. The amino-terminal sequence of PARs is cleaved by a broad array of proteases, leading to specific proteolytic cleavage which forms endogenous tethered ligands to induce agonist-biased PAR activation. The biological effect of PARs activated by coagulation proteases to regulate hemostasis and thrombosis plays an enormous role in the cardiovascular system, while PAR4 can also be activated by trypsin, cathepsin G, the activated factor X of the coagulation cascade, and trypsin IV. Irrespective of its role in thrombin-induced platelet aggregation, PAR4 activation is believed to be involved in inflammatory lesions, as show by investigations that have unmasked the effects of PAR4 on neutrophil recruitment, the regulation of edema, and plasma extravasation. This review summarizes the roles of PAR4 in coagulation and other extracellular protease pathways, which activate PAR4 to participate in normal regulation and disease.
Collapse
Affiliation(s)
- Qiang Fu
- Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008, China
| | | | | | | | | | | |
Collapse
|
13
|
Platelets and protease-activated receptor-4 contribute to acetaminophen-induced liver injury in mice. Blood 2015; 126:1835-43. [PMID: 26179083 DOI: 10.1182/blood-2014-09-598656] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 07/13/2015] [Indexed: 12/15/2022] Open
Abstract
Acetaminophen (APAP)-induced liver injury in humans is associated with robust coagulation cascade activation and thrombocytopenia. However, it is not known whether coagulation-driven platelet activation participates in APAP hepatotoxicity. Here, we found that APAP overdose in mice caused liver damage accompanied by significant thrombocytopenia and accumulation of platelets in the liver. These changes were attenuated by administration of the direct thrombin inhibitor lepirudin. Platelet depletion with an anti-CD41 antibody also significantly reduced APAP-mediated liver injury and thrombin generation, indicated by the concentration of thrombin-antithrombin (TAT) complexes in plasma. Compared with APAP-treated wild-type mice, biomarkers of hepatocellular and endothelial damage, plasma TAT concentration, and hepatic platelet accumulation were reduced in mice lacking protease-activated receptor (PAR)-4, which mediates thrombin signaling in mouse platelets. However, selective hematopoietic cell PAR-4 deficiency did not affect APAP-induced liver injury or plasma TAT levels. These results suggest that interconnections between coagulation and hepatic platelet accumulation promote APAP-induced liver injury, independent of platelet PAR-4 signaling. Moreover, the results highlight a potential contribution of nonhematopoietic cell PAR-4 signaling to APAP hepatotoxicity.
Collapse
|
14
|
Carrasco L, Cea P, Rocco P, Peña-Oyarzún D, Rivera-Mejias P, Sotomayor-Flores C, Quiroga C, Criollo A, Ibarra C, Chiong M, Lavandero S. Role of Heterotrimeric G Protein and Calcium in Cardiomyocyte Hypertrophy Induced by IGF-1. J Cell Biochem 2014; 115:712-20. [DOI: 10.1002/jcb.24712] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 11/05/2013] [Indexed: 01/09/2023]
Affiliation(s)
- Loreto Carrasco
- Advanced Center for Chronic Diseases; Universidad de Chile; Santiago Chile
- Centro Estudios Moleculares de la Celula; Facultad de Ciencias y Farmacéuticas; Universidad de Chile; Santiago Chile
| | - Paola Cea
- Advanced Center for Chronic Diseases; Universidad de Chile; Santiago Chile
- Centro Estudios Moleculares de la Celula; Facultad de Ciencias y Farmacéuticas; Universidad de Chile; Santiago Chile
| | - Paola Rocco
- Centro Estudios Moleculares de la Celula; Facultad de Ciencias y Farmacéuticas; Universidad de Chile; Santiago Chile
| | - Daniel Peña-Oyarzún
- Advanced Center for Chronic Diseases; Universidad de Chile; Santiago Chile
- Centro Estudios Moleculares de la Celula; Facultad de Ciencias y Farmacéuticas; Universidad de Chile; Santiago Chile
| | - Pablo Rivera-Mejias
- Advanced Center for Chronic Diseases; Universidad de Chile; Santiago Chile
- Centro Estudios Moleculares de la Celula; Facultad de Ciencias y Farmacéuticas; Universidad de Chile; Santiago Chile
| | - Cristian Sotomayor-Flores
- Advanced Center for Chronic Diseases; Universidad de Chile; Santiago Chile
- Centro Estudios Moleculares de la Celula; Facultad de Ciencias y Farmacéuticas; Universidad de Chile; Santiago Chile
| | - Clara Quiroga
- Advanced Center for Chronic Diseases; Universidad de Chile; Santiago Chile
- Centro Estudios Moleculares de la Celula; Facultad de Ciencias y Farmacéuticas; Universidad de Chile; Santiago Chile
| | - Alfredo Criollo
- Advanced Center for Chronic Diseases; Universidad de Chile; Santiago Chile
- Departamento Ciencias Básicas y Comunitarias; Facultad Odontología; Universidad de Chile; Santiago Chile
| | - Cristian Ibarra
- Advanced Center for Chronic Diseases; Universidad de Chile; Santiago Chile
- Department of Medical Biochemistry and Biophysics; Karolinska Institutet; Stockholm Sweden
| | - Mario Chiong
- Advanced Center for Chronic Diseases; Universidad de Chile; Santiago Chile
- Centro Estudios Moleculares de la Celula; Facultad de Ciencias y Farmacéuticas; Universidad de Chile; Santiago Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases; Universidad de Chile; Santiago Chile
- Centro Estudios Moleculares de la Celula; Facultad de Ciencias y Farmacéuticas; Universidad de Chile; Santiago Chile
- Instituto de Ciencias Biomédicas; Facultad Medicina; Universidad de Chile; Santiago Chile
- Cardiology Division; Department of Internal Medicine; University of Texas Southwestern Medical Center; Dallas Texas
| |
Collapse
|
15
|
Asteriti S, Daniele S, Porchia F, Dell'Anno MT, Fazzini A, Pugliesi I, Trincavelli ML, Taliani S, Martini C, Mazzoni MR, Gilchrist A. Modulation of PAR(1) signalling by benzimidazole compounds. Br J Pharmacol 2013; 167:80-94. [PMID: 22519452 DOI: 10.1111/j.1476-5381.2012.01974.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Recently, a small molecule (Q94) was reported to selectively block PAR(1) /Gα(q) interaction and signalling. Here, we describe the pharmacological properties of Q94 and two analogues that share its benzimidazole scaffold (Q109, Q89). Q109 presents a modest variation from Q94 in the substituent group at the 2-position, while Q89 has quite different groups at the 1- and 2-positions. EXPERIMENTAL APPROACH Using human microvascular endothelial cells, we examined intracellular Ca(2+) mobilization and inositol 1,4,5-trisphosphate accumulation as well as isoprenaline- or forskolin-stimulated cAMP production in response to thrombin. KEY RESULTS Q89 (10 µM) produced a leftward shift in the thrombin-mediated intracellular Ca(2+) mobilization concentration-response curve while having no effect on the E(max) . Both Q94 (10 µM) and Q109 (10 µM) reduced intracellular Ca(2+) mobilization, leading to a decrease in E(max) and an increase in EC(50) values. Experiments utilizing receptor-specific activating peptides confirmed that Q94 and Q109 were selective for PAR(1) as they did not alter the Ca(2+) response mediated by a PAR(2) activating peptide. Consistent with our Ca(2+) results, micromolar concentrations of either Q94 or Q109 significantly reduced thrombin-induced inositol 1,4,5-trisphosphate production. Neither Q94 nor Q109 diminished the inhibitory effects of thrombin on cAMP production, indicating they inhibit signalling selectively through the G(q) pathway. Our results also suggest the 1,2-disubstituted benzimidazole derivatives act as 'allosteric agonists' of PAR(1) . CONCLUSIONS AND IMPLICATIONS The Q94 and Q109 benzimidazole derivatives represent a novel scaffold for the development of new PAR(1) inhibitors and provide a starting point to develop dual signalling pathway-selective positive/negative modulators of PAR(1) .
Collapse
Affiliation(s)
- S Asteriti
- Department of Psychiatry, Neurobiology, Pharmacology and Biotechnology, University of Pisa, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Canto I, Soh UJK, Trejo J. Allosteric modulation of protease-activated receptor signaling. Mini Rev Med Chem 2012; 12:804-11. [PMID: 22681248 DOI: 10.2174/138955712800959116] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 08/17/2011] [Accepted: 09/02/2011] [Indexed: 12/23/2022]
Abstract
The protease-activated receptors (PARs) are G protein-coupled receptors (GPCRs) that are uniquely activated by proteolysis. PARs mediate hemostasis, thrombosis, inflammation, embryonic development and progression of certain malignant cancers. The family of PARs include four members: PAR1, PAR2, PAR3 and PAR4. PARs harbor a cryptic ligand sequence within their N-terminus that is exposed following proteolytic cleavage. The newly formed PAR Nterminus functions as a tethered ligand that binds intramolecularly to the receptor to trigger transmembrane signaling. This unique mechanism of activation would indicate that regardless of the activating protease, cleavage of PARs would unmask a tethered ligand sequence that would induce a similar active receptor conformation and signaling response. However, this is not the case. Recent studies demonstrate that PARs can be differentially activated by synthetic peptide agonists, proteases or through dimerization, that ultimately result in distinct cellular responses. In some cases, allosteric modulation of PARs involves compartmentalization in caveolae, plasma membrane microdomains enriched in cholesterol. Here, we discuss some mechanisms that lead to allosteric modulation of PAR signaling.
Collapse
Affiliation(s)
- I Canto
- Department of Pharmacology, School of Medicine, University of California, San Diego, Biomedical Sciences Building, Room 3044A, 9500 Gilman Drive, La Jolla, CA 92093-0636, USA.
| | | | | |
Collapse
|
17
|
Xu T, Zhao W, Zhu JM, Albanna MZ, Yoo JJ, Atala A. Complex heterogeneous tissue constructs containing multiple cell types prepared by inkjet printing technology. Biomaterials 2012; 34:130-9. [PMID: 23063369 DOI: 10.1016/j.biomaterials.2012.09.035] [Citation(s) in RCA: 336] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 09/17/2012] [Indexed: 02/07/2023]
Abstract
This study was designed to develop a versatile method for fabricating complex and heterogeneous three-dimensional (3D) tissue constructs using simultaneous ink-jetting of multiple cell types. Human amniotic fluid-derived stem cells (hAFSCs), canine smooth muscle cells (dSMCs), and bovine aortic endothelial cells (bECs), were separately mixed with ionic cross-linker calcium chloride (CaCl(2)), loaded into separate ink cartridges and printed using a modified thermal inkjet printer. The three cell types were delivered layer-by-layer to pre-determined locations in a sodium alginate-collagen composite located in a chamber under the printer. The reaction between CaCl(2) and sodium alginate resulted in a rapid formation of a solid composite gel and the printed cells were anchored in designated areas within the gel. The printing process was repeated for several cycles leading to a complex 3D multi-cell hybrid construct. The biological functions of the 3D printed constructs were evaluated in vitro and in vivo. Each of the printed cell types maintained their viability and normal proliferation rates, phenotypic expression, and physiological functions within the heterogeneous constructs. The bioprinted constructs were able to survive and mature into functional tissues with adequate vascularization in vivo. These findings demonstrate the feasibility of fabricating complex heterogeneous tissue constructs containing multiple cell types using inkjet printing technology.
Collapse
Affiliation(s)
- Tao Xu
- Wake Forest Institute for Regenerative Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | | | | | | | | | | |
Collapse
|
18
|
Moccia F, Berra-Romani R, Tanzi F. Update on vascular endothelial Ca 2+ signalling: A tale of ion channels, pumps and transporters. World J Biol Chem 2012; 3:127-58. [PMID: 22905291 PMCID: PMC3421132 DOI: 10.4331/wjbc.v3.i7.127] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 07/04/2012] [Accepted: 07/11/2012] [Indexed: 02/05/2023] Open
Abstract
A monolayer of endothelial cells (ECs) lines the lumen of blood vessels and forms a multifunctional transducing organ that mediates a plethora of cardiovascular processes. The activation of ECs from as state of quiescence is, therefore, regarded among the early events leading to the onset and progression of potentially lethal diseases, such as hypertension, myocardial infarction, brain stroke, and tumor. Intracellular Ca2+ signals have long been know to play a central role in the complex network of signaling pathways regulating the endothelial functions. Notably, recent work has outlined how any change in the pattern of expression of endothelial channels, transporters and pumps involved in the modulation of intracellular Ca2+ levels may dramatically affect whole body homeostasis. Vascular ECs may react to both mechanical and chemical stimuli by generating a variety of intracellular Ca2+ signals, ranging from brief, localized Ca2+ pulses to prolonged Ca2+ oscillations engulfing the whole cytoplasm. The well-defined spatiotemporal profile of the subcellular Ca2+ signals elicited in ECs by specific extracellular inputs depends on the interaction between Ca2+ releasing channels, which are located both on the plasma membrane and in a number of intracellular organelles, and Ca2+ removing systems. The present article aims to summarize both the past and recent literature in the field to provide a clear-cut picture of our current knowledge on the molecular nature and the role played by the components of the Ca2+ machinery in vascular ECs under both physiological and pathological conditions.
Collapse
Affiliation(s)
- Francesco Moccia
- Francesco Moccia, Franco Tanzi, Department of Biology and Biotechnologies "Lazzaro Spallanzani", Laboratory of Physiology, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy
| | | | | |
Collapse
|
19
|
Komatsu H, Shimose A, Shimizu T, Mukai Y, Kobayashi J, Ohama T, Sato K. Trypsin inhibits lipopolysaccharide signaling in macrophages via toll-like receptor 4 accessory molecules. Life Sci 2012; 91:143-50. [PMID: 22771700 DOI: 10.1016/j.lfs.2012.06.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 06/23/2012] [Indexed: 12/21/2022]
Abstract
AIMS To examine the role of trypsin in the immune response of macrophages and to determine whether protease-activated receptors (PARs) are involved in the effects of trypsin. MAIN METHODS We used RAW264.7 cells and peritoneal macrophages isolated from C57BL/6 wild-type mice, PAR2 knockout mice, and ddY mice. Macrophages were stimulated with lipopolysaccharide (LPS) in the presence or absence of trypsin, thrombin, and PAR subtype-specific agonists (PARs-AP). Activation of macrophages was quantified by nitric oxide production and expression of inflammatory mediators, such as inducible nitric oxide synthase, interleukin-1β, and interleukin-6. To clarify the effect of trypsin on LPS receptors, we also investigated the expression of toll-like receptor 4 (TLR4), soluble MD-2 (sMD-2), membrane-bound MD-2 (mMD-2), soluble CD14 (sCD14), and membrane-bound CD14 (mCD14). To directly investigate the effect of trypsin on CD14 protein, we expressed recombinant CD14 protein. KEY FINDINGS Trypsin inhibited LPS-induced nitric oxide production and expression of inducible nitric oxide synthase, interleukin-1β, and interleukin-6. The same inhibitory effects of trypsin were observed in wild-type macrophages and in PAR2 knockout macrophages. Furthermore, the other PAR agonists, thrombin, PAR1-AP, PAR2-AP, and PAR4-AP, did not mimic the effect of trypsin. Although trypsin did not affect TLR4 or mMD-2 expression, sCD14, mCD14, and sMD-2 expressions were decreased by trypsin. Furthermore, trypsin also degraded recombinant CD14 protein. SIGNIFICANCE Trypsin inhibited LPS signaling PAR-independently via degradation of TLR4 accessory molecules. This observation provides a better understanding of the complicated immune response in acute pancreatitis.
Collapse
Affiliation(s)
- Hiroyuki Komatsu
- Laboratory of Veterinary Pharmacology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Japan
| | | | | | | | | | | | | |
Collapse
|
20
|
Cunningham MR, McIntosh KA, Pediani JD, Robben J, Cooke AE, Nilsson M, Gould GW, Mundell S, Milligan G, Plevin R. Novel role for proteinase-activated receptor 2 (PAR2) in membrane trafficking of proteinase-activated receptor 4 (PAR4). J Biol Chem 2012; 287:16656-69. [PMID: 22411985 PMCID: PMC3351358 DOI: 10.1074/jbc.m111.315911] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Proteinase-activated receptors 4 (PAR4) is a class A G protein-coupled receptor (GPCR) recognized through the ability of serine proteases such as thrombin and trypsin to mediate receptor activation. Due to the irreversible nature of activation, a fresh supply of receptor is required to be mobilized to the cell surface for responsiveness to agonist to be sustained. Unlike other PAR subtypes, the mechanisms regulating receptor trafficking of PAR4 remain unknown. Here, we report novel features of the intracellular trafficking of PAR4 to the plasma membrane. PAR4 was poorly expressed at the plasma membrane and largely retained in the endoplasmic reticulum (ER) in a complex with the COPI protein subunit β-COP1. Analysis of the PAR4 protein sequence identified an arginine-based (RXR) ER retention sequence located within intracellular loop-2 (R183AR → A183AA), mutation of which allowed efficient membrane delivery of PAR4. Interestingly, co-expression with PAR2 facilitated plasma membrane delivery of PAR4, an effect produced through disruption of β-COP1 binding and facilitation of interaction with the chaperone protein 14-3-3ζ. Intermolecular FRET studies confirmed heterodimerization between PAR2 and PAR4. PAR2 also enhanced glycosylation of PAR4 and activation of PAR4 signaling. Our results identify a novel regulatory role for PAR2 in the anterograde traffic of PAR4. PAR2 was shown to both facilitate and abrogate protein interactions with PAR4, impacting upon receptor localization and cell signal transduction. This work is likely to impact markedly upon the understanding of the receptor pharmacology of PAR4 in normal physiology and disease.
Collapse
Affiliation(s)
- Margaret R Cunningham
- Department of Physiology and Pharmacology, Strathclyde Institute for Biomedical Sciences, Univesity of Strathclyde, 27 Taylor Street, Glasgow G4 0NR, Scotland, United Kingdom.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
Abstract population are ubiquitous background radiation and medical exposure of patients. From the early 1980s to 2006, the average dose per individual in the United States for all sources of radiation increased by a factor of 1.7-6.2 mSv, with this increase due to the growth of medical imaging procedures. Radiation can place individuals at an increased risk of developing cardiovascular disease. Excess risk of cardiovascular disease occurs a long time after exposure to lower doses of radiation as demonstrated in Japanese atomic bomb survivors. This review examines sources of radiation (atomic bombs, radiation accidents, radiological terrorism, cancer treatment, space exploration, radiosurgery for cardiac arrhythmia, and computed tomography) and the risk for developing cardiovascular disease. The evidence presented suggests an association between cardiovascular disease and exposure to low-to-moderate levels of radiation, as well as the well-known association at high doses. Studies are needed to define the extent that diagnostic and therapeutic radiation results in increased risk factors for cardiovascular disease, to understand the mechanisms involved, and to develop strategies to mitigate or treat radiation-induced cardiovascular disease.
Collapse
Affiliation(s)
- John E Baker
- Division of Cardiothoracic Surgery, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| | | | | |
Collapse
|
22
|
Adams MN, Ramachandran R, Yau MK, Suen JY, Fairlie DP, Hollenberg MD, Hooper JD. Structure, function and pathophysiology of protease activated receptors. Pharmacol Ther 2011; 130:248-82. [PMID: 21277892 DOI: 10.1016/j.pharmthera.2011.01.003] [Citation(s) in RCA: 280] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 01/03/2011] [Indexed: 12/18/2022]
Abstract
Discovered in the 1990s, protease activated receptors(1) (PARs) are membrane-spanning cell surface proteins that belong to the G protein coupled receptor (GPCR) family. A defining feature of these receptors is their irreversible activation by proteases; mainly serine. Proteolytic agonists remove the PAR extracellular amino terminal pro-domain to expose a new amino terminus, or tethered ligand, that binds intramolecularly to induce intracellular signal transduction via a number of molecular pathways that regulate a variety of cellular responses. By these mechanisms PARs function as cell surface sensors of extracellular and cell surface associated proteases, contributing extensively to regulation of homeostasis, as well as to dysfunctional responses required for progression of a number of diseases. This review examines common and distinguishing structural features of PARs, mechanisms of receptor activation, trafficking and signal termination, and discusses the physiological and pathological roles of these receptors and emerging approaches for modulating PAR-mediated signaling in disease.
Collapse
Affiliation(s)
- Mark N Adams
- Mater Medical Research Institute, Aubigny Place, Raymond Terrace, South Brisbane Qld 4101, Australia
| | | | | | | | | | | | | |
Collapse
|
23
|
The Indazole Derivative YD-3 Specifically Inhibits Thrombin-Induced Angiogenesis In Vitro and In Vivo. Shock 2010; 34:580-5. [DOI: 10.1097/shk.0b013e3181df00a3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
24
|
Hirano K, Hirano M, Hanada A. Involvement of STIM1 in the proteinase-activated receptor 1-mediated Ca2+ influx in vascular endothelial cells. J Cell Biochem 2009; 108:499-507. [PMID: 19626660 DOI: 10.1002/jcb.22279] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Thrombin increases the cytosolic Ca(2+) concentrations and induces NO production by activating proteinase-activated receptor 1 (PAR(1)) in vascular endothelial cells. The store-operated Ca(2+) influx is a major Ca(2+) influx pathway in non-excitable cells including endothelial cells and it has been reported to play a role in the thrombin-induced Ca(2+) signaling in endothelial cells. Recent studies have identified stromal interaction molecule 1 (STIM1) to function as a sensor of the store site Ca(2+) content, thereby regulating the store-operated Ca(2+) influx. However, the functional role of STIM1 in the thrombin-induced Ca(2+) influx and NO production in endothelial cells still remains to be elucidated. Fura-2 and diaminorhodamine-4M fluorometry was utilized to evaluate the thrombin-induced changes in cytosolic Ca(2+) concentrations and NO production, respectively, in porcine aortic endothelial cells transfected with small interfering RNA (siRNA) targeted to STIM1. STIM1-targeted siRNA suppressed the STIM1 expression and the thapsigargin-induced Ca(2+) influx. The degree of suppression of the STIM1 expression correlated well to the degree of suppression of the Ca(2+) influx. The knockdown of STIM1 was associated with a substantial inhibition of the Ca(2+) influx and a partial reduction of the NO production induced by thrombin. The thrombin-induced Ca(2+) influx exhibited the similar sensitivity toward the Ca(2+) influx inhibitors to that seen with the thapsigargin-induced Ca(2+) influx. The present study provides the first evidence that STIM1 plays a critical role in the PAR(1)-mediated Ca(2+) influx and Ca(2+)-dependent component of the NO production in endothelial cells.
Collapse
Affiliation(s)
- Katsuya Hirano
- Division of Molecular Cardiology, Research Institute of Angiocardiology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, Japan.
| | | | | |
Collapse
|
25
|
Augé C, Balz-Hara D, Steinhoff M, Vergnolle N, Cenac N. Protease-activated receptor-4 (PAR 4): a role as inhibitor of visceral pain and hypersensitivity. Neurogastroenterol Motil 2009; 21:1189-e107. [PMID: 19413681 DOI: 10.1111/j.1365-2982.2009.01310.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Protease-activated receptor-4 (PAR(4)) belongs to the family of receptors activated by the proteolytic cleavage of their extracellular N-terminal domain and the subsequent binding of the newly released N-terminus. While largely expressed in the colon, the role of PAR(4) in gut functions has not been defined. We have investigated the effects of PAR(4) agonist on colonic sensations and sensory neuron signalling, and its role in visceral pain. We observed that a single administration of the PAR(4) agonist peptide (AYPGKF-NH(2)), but not the control peptide (YAPGKF-NH(2)) into the colon lumen of mice significantly reduced the visceromotor response to colorectal distension at different pressures of distension. Further, intracolonic administration of the PAR(4) agonist, but not the control peptide, was able to significantly inhibit PAR(2) agonist- and transcient receptor potential vanilloid-4 (TRPV4) agonist-induced allodynia and hyperalgesia in response to colorectal distension. Protease-activated receptor-4 was detected in sensory neurons projecting from the colon, and isolated from the dorsal root ganglia, where it co-expressed with PAR(2) and TRPV4. In total sensory neurons, PAR(4) agonist exposure inhibited free intracellular calcium mobilization induced by the pro-nociceptive agonists of PAR(2) and TRPV4. Finally, PAR(4)-deficient mice experienced increased pain behaviour in response to intracolonic administration of mustard oil, compared with wild-type littermates. These results show that PAR(4) agonists modulate colonic nociceptive response, inhibit colonic hypersensitivity and primary afferent responses to pro-nociceptive mediators. Endogenous activation of PAR(4) also plays a major role in controlling visceral pain. These results identify PAR(4) as a previously unknown modulator of visceral nociception.
Collapse
Affiliation(s)
- C Augé
- INSERM U563, Centre de Physiopathologie de Toulouse Purpan, Toulouse, France
| | | | | | | | | |
Collapse
|
26
|
Waismam K, Chudzinski-Tavassi AM, Carrijo-Carvalho LC, Fernandes Pacheco MT, Farsky SH. Lopap: A non-inflammatory and cytoprotective molecule in neutrophils and endothelial cells. Toxicon 2009; 53:652-9. [DOI: 10.1016/j.toxicon.2009.01.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
27
|
Suzuki H, Motley ED, Eguchi K, Hinoki A, Shirai H, Watts V, Stemmle LN, Fields TA, Eguchi S. Distinct roles of protease-activated receptors in signal transduction regulation of endothelial nitric oxide synthase. Hypertension 2009; 53:182-8. [PMID: 19064814 PMCID: PMC2679177 DOI: 10.1161/hypertensionaha.108.125229] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2008] [Accepted: 11/13/2008] [Indexed: 01/29/2023]
Abstract
Protease-activated receptors (PARs), such as PAR1 and PAR2, have been implicated in the regulation of endothelial NO production. We hypothesized that PAR1 and PAR2 distinctly regulate the activity of endothelial NO synthase through the selective phosphorylation of a positive regulatory site, Ser(1179), and a negative regulatory site, Thr(497), in bovine aortic endothelial cells. A selective PAR1 ligand, TFLLR, stimulated the phosphorylation of endothelial NO synthase at Thr(497). It had a minimal effect on Ser(1179) phosphorylation. In contrast, a selective PAR2 ligand, SLIGRL, stimulated the phosphorylation of Ser(1179) with no noticeable effect on Thr(497). Thrombin has been shown to transactivate PAR2 through PAR1. Thus, thrombin, as well as a peptide mimicking the PAR1 tethered ligand, TRAP, stimulated phosphorylation of both sites. Also, thrombin and SLIGRL, but not TFLLR, stimulated cGMP production. A G(q) inhibitor blocked thrombin- and SLIGRL-induced Ser(1179) phosphorylation, whereas it enhanced thrombin-induced Thr(497) phosphorylation. In contrast, a G(12/13) inhibitor blocked thrombin- and TFLLR-induced Thr(497) phosphorylation, whereas it enhanced the Ser(1179) phosphorylation. Although a Rho-kinase inhibitor, Y27632, blocked the Thr(497) phosphorylation, other inhibitors that targeted Rho-kinase failed to block TFLLR-induced Thr(497) phosphorylation. These data suggest that PAR1 and PAR2 distinctly regulate endothelial NO synthase phosphorylation and activity through G(12/13) and G(q), respectively, delineating the novel signaling pathways by which the proteases act on protease-activated receptors to potentially modulate endothelial functions.
Collapse
Affiliation(s)
- Hiroyuki Suzuki
- Cardiovascular Research Center and Department of Physiology, Temple University School of Medicine, 3420 N Broad St, Philadelphia, PA 19140, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Sangawa T, Nogi T, Takagi J. A Murine Monoclonal Antibody That Binds N-Terminal Extracellular Segment of Human Protease-Activated Receptor-4. Hybridoma (Larchmt) 2008; 27:331-5. [DOI: 10.1089/hyb.2008.0027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Takeshi Sangawa
- Laboratory of Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Terukazu Nogi
- Laboratory of Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Junichi Takagi
- Laboratory of Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Osaka, Japan
| |
Collapse
|
29
|
Kai Y, Maeda Y, Sasaki T, Kanaide H, Hirano K. Basic and Translational Research on Proteinase-Activated Receptors: The Role of Thrombin Receptor in Cerebral Vasospasm in Subarachnoid Hemorrhage. J Pharmacol Sci 2008; 108:426-32. [DOI: 10.1254/jphs.08r11fm] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|