1
|
Oe Y, Tanaka T, Takahashi N. The Many Faces of Protease-Activated Receptor 2 in Kidney Injury. Biomedicines 2025; 13:414. [PMID: 40002827 PMCID: PMC11852827 DOI: 10.3390/biomedicines13020414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/15/2025] [Accepted: 02/02/2025] [Indexed: 02/27/2025] Open
Abstract
Protease-activated receptor 2 (PAR2) is a seven-transmembrane, G-protein-coupled receptor that is activated by coagulation proteases such as factor VIIa and factor Xa and other serine proteases. It is a potential therapeutic target for kidney injury, as it enhances inflammatory and fibrotic responses via the nuclear factor-kappa B and mitogen-activated protein kinase cascades. The body of knowledge regarding the role of PAR2 in kidney disease is currently growing, and its role in various kidney disease models, such as acute kidney injury, renal fibrosis, diabetic kidney disease, aging, and thrombotic microangiopathy, has been reported. Here, we review the literature to better understand the various aspects of PAR2 in kidney disease.
Collapse
Affiliation(s)
- Yuji Oe
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Tetsuhiro Tanaka
- Department of Nephrology, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Nobuyuki Takahashi
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences & Faculty of Pharmaceutical Sciences, Sendai 980-0845, Japan
| |
Collapse
|
2
|
Vesey DA, Iyer A, Owen E, Kamato D, Johnson DW, Gobe GC, Fairlie DP, Nikolic-Paterson DJ. PAR2 activation on human tubular epithelial cells engages converging signaling pathways to induce an inflammatory and fibrotic milieu. Front Pharmacol 2024; 15:1382094. [PMID: 39005931 PMCID: PMC11239397 DOI: 10.3389/fphar.2024.1382094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/31/2024] [Indexed: 07/16/2024] Open
Abstract
Key features of chronic kidney disease (CKD) include tubulointerstitial inflammation and fibrosis. Protease activated receptor-2 (PAR2), a G-protein coupled receptor (GPCR) expressed by the kidney proximal tubular cells, induces potent proinflammatory responses in these cells. The hypothesis tested here was that PAR2 signalling can contribute to both inflammation and fibrosis in the kidney by transactivating known disease associated pathways. Using a primary cell culture model of human kidney tubular epithelial cells (HTEC), PAR2 activation induced a concentration dependent, PAR2 antagonist sensitive, secretion of TNF, CSF2, MMP-9, PAI-1 and CTGF. Transcription factors activated by the PAR2 agonist 2F, including NFκB, AP1 and Smad2, were critical for production of these cytokines. A TGF-β receptor-1 (TGF-βRI) kinase inhibitor, SB431542, and an EGFR kinase inhibitor, AG1478, ameliorated 2F induced secretion of TNF, CSF2, MMP-9, and PAI-1. Whilst an EGFR blocking antibody, cetuximab, blocked PAR2 induced EGFR and ERK phosphorylation, a TGF-βRII blocking antibody failed to influence PAR2 induced secretion of PAI-1. Notably simultaneous activation of TGF-βRII (TGF-β1) and PAR2 (2F) synergistically enhanced secretion of TNF (2.2-fold), CSF2 (4.4-fold), MMP-9 (15-fold), and PAI-1 (2.5-fold). In summary PAR2 activates critical inflammatory and fibrotic signalling pathways in human kidney tubular epithelial cells. Biased antagonists of PAR2 should be explored as a potential therapy for CKD.
Collapse
Affiliation(s)
- David A Vesey
- Centre for Kidney Disease Research, Translational Research Institute, The University of Queensland at the Princess Alexandra Hospital, Brisbane, QLD, Australia
- Department of Kidney and Transplant Services, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Abishek Iyer
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- Centre for Inflammation and Disease Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Evan Owen
- Centre for Kidney Disease Research, Translational Research Institute, The University of Queensland at the Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Danielle Kamato
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - David W Johnson
- Centre for Kidney Disease Research, Translational Research Institute, The University of Queensland at the Princess Alexandra Hospital, Brisbane, QLD, Australia
- Department of Kidney and Transplant Services, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Glenda C Gobe
- Centre for Kidney Disease Research, Translational Research Institute, The University of Queensland at the Princess Alexandra Hospital, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - David P Fairlie
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- Centre for Inflammation and Disease Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - David J Nikolic-Paterson
- Department of Nephrology, Monash Health and Monash University Centre for Inflammatory Diseases, Monash Medical Centre, Clayton, VIC, Australia
| |
Collapse
|
3
|
Bagang N, Gupta K, Singh G, Kanuri SH, Mehan S. Protease-activated receptors in kidney diseases: A comprehensive review of pathological roles, therapeutic outcomes and challenges. Chem Biol Interact 2023; 377:110470. [PMID: 37011708 DOI: 10.1016/j.cbi.2023.110470] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/21/2023] [Accepted: 03/31/2023] [Indexed: 04/03/2023]
Abstract
Studies have demonstrated that protease-activated receptors (PARs) with four subtypes (PAR1-4) are mainly expressed in the renal epithelial, endothelial, and podocyte cells. Some endogenous and urinary proteases, namely thrombin, trypsin, urokinase, and kallikrein released during diseased conditions, are responsible for activating different subtypes of PARs. Each PAR receptor subtype is involved in kidney disease of distinct aetiology. PAR1 and PAR2 have shown differential therapeutic outcomes in rodent models of type-1 and type-2 diabetic kidney diseases due to the distinct etiological basis of each disease type, however such findings need to be confirmed in other diabetic renal injury models. PAR1 and PAR2 blockers have been observed to abolish drug-induced nephrotoxicity in rodents by suppressing tubular inflammation and fibrosis and preventing mitochondrial dysfunction. Notably, PAR2 inhibition improved autophagy and prevented fibrosis, inflammation, and remodeling in the urethral obstruction model. Only the PAR1/4 subtypes have emerged as a therapeutic target for treating experimentally induced nephrotic syndrome, where their respective antibodies attenuated the podocyte apoptosis induced upon thrombin activation. Strikingly PAR2 and PAR4 subtypes involvement has been tested in sepsis-induced acute kidney injury (AKI) and renal ischemia-reperfusion injury models. Thus, more studies are required to delineate the role of other subtypes in the sepsis-AKI model. Evidence suggests that PARs regulate oxidative, inflammatory stress, immune cell activation, fibrosis, autophagic flux, and apoptosis during kidney diseases.
Collapse
|
4
|
Oe Y, Miyazaki M, Takahashi N. Coagulation, Protease-Activated Receptors, and Diabetic Kidney Disease: Lessons from eNOS-Deficient Mice. TOHOKU J EXP MED 2021; 255:1-8. [PMID: 34511578 DOI: 10.1620/tjem.255.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Endothelial nitric oxide synthase (eNOS) dysfunction is known to exacerbate the progression and prognosis of diabetic kidney disease (DKD). One of the mechanisms through which this is achieved is that low eNOS levels are associated with hypercoagulability, which promotes kidney injury. In the extrinsic coagulation cascade, the tissue factor (factor III) and downstream coagulation factors, such as active factor X (FXa), exacerbate inflammation through activation of the protease-activated receptors (PARs). Recently, it has been shown that the lack of or reduced eNOS expression in diabetic mice, as a model of advanced DKD, increases renal tissue factor levels and PAR1 and 2 expression in their kidneys. Furthermore, pharmaceutical inhibition or genetic deletion of coagulation factors or PARs ameliorated inflammation in DKD in mice lacking eNOS. In this review, we summarize the relationship between eNOS, coagulation, and PARs and propose a novel therapeutic option for the management of patients with DKD.
Collapse
Affiliation(s)
- Yuji Oe
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine
| | - Mariko Miyazaki
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine
| | - Nobuyuki Takahashi
- Division of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences & Faculty of Pharmaceutical Sciences
| |
Collapse
|
5
|
Humphries TLR, Shen K, Iyer A, Johnson DW, Gobe GC, Nikolic-Paterson D, Fairlie DP, Vesey DA. PAR2-Induced Tissue Factor Synthesis by Primary Cultures of Human Kidney Tubular Epithelial Cells Is Modified by Glucose Availability. Int J Mol Sci 2021; 22:ijms22147532. [PMID: 34299151 PMCID: PMC8304776 DOI: 10.3390/ijms22147532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/07/2021] [Accepted: 07/07/2021] [Indexed: 01/05/2023] Open
Abstract
Coagulopathies common to patients with diabetes and chronic kidney disease (CKD) are not fully understood. Fibrin deposits in the kidney suggest the local presence of clotting factors including tissue factor (TF). In this study, we investigated the effect of glucose availability on the synthesis of TF by cultured human kidney tubular epithelial cells (HTECs) in response to activation of protease-activated receptor 2 (PAR2). PAR2 activation by peptide 2f-LIGRLO-NH2 (2F, 2 µM) enhanced the synthesis and secretion of active TF (~45 kDa) which was blocked by a PAR2 antagonist (I-191). Treatment with 2F also significantly increased the consumption of glucose from the cell medium and lactate secretion. Culturing HTECs in 25 mM glucose enhanced TF synthesis and secretion over 5 mM glucose, while addition of 5 mM 2-deoxyglucose (2DOG) significantly decreased TF synthesis and reduced its molecular weight (~40 kDa). Blocking glycosylation with tunicamycin also reduced 2F-induced TF synthesis while reducing its molecular weight (~36 kDa). In conclusion, PAR2-induced TF synthesis in HTECs is enhanced by culture in high concentrations of glucose and suppressed by inhibiting either PAR2 activation (I-191), glycolysis (2DOG) or glycosylation (tunicamycin). These results may help explain how elevated concentrations of glucose promote clotting abnormities in diabetic kidney disease. The application of PAR2 antagonists to treat CKD should be investigated further.
Collapse
Affiliation(s)
- Tyrone L. R. Humphries
- Centre for Kidney Disease Research, Translational Research Institute, Faulty of Medicine, The University of Queensland at the Princess Alexandra, Brisbane, QLD 4072, Australia; (T.L.R.H.); (K.S.); (D.W.J.); (G.C.G.)
| | - Kunyu Shen
- Centre for Kidney Disease Research, Translational Research Institute, Faulty of Medicine, The University of Queensland at the Princess Alexandra, Brisbane, QLD 4072, Australia; (T.L.R.H.); (K.S.); (D.W.J.); (G.C.G.)
| | - Abishek Iyer
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia; (A.I.); (D.P.F.)
- Centre for Inflammation and Disease Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - David W. Johnson
- Centre for Kidney Disease Research, Translational Research Institute, Faulty of Medicine, The University of Queensland at the Princess Alexandra, Brisbane, QLD 4072, Australia; (T.L.R.H.); (K.S.); (D.W.J.); (G.C.G.)
- Department of Nephrology, The University of Queensland at Princess Alexandra Hospital, Brisbane, QLD 4102, Australia
| | - Glenda C. Gobe
- Centre for Kidney Disease Research, Translational Research Institute, Faulty of Medicine, The University of Queensland at the Princess Alexandra, Brisbane, QLD 4072, Australia; (T.L.R.H.); (K.S.); (D.W.J.); (G.C.G.)
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland at the Translational Research Institute, Brisbane, QLD 4072, Australia
| | - David Nikolic-Paterson
- Department of Nephrology, Monash Medical Centre and Monash University Centre for Inflammatory Diseases, Melbourne, VIC 3168, Australia;
| | - David P. Fairlie
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia; (A.I.); (D.P.F.)
- Centre for Inflammation and Disease Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - David A. Vesey
- Centre for Kidney Disease Research, Translational Research Institute, Faulty of Medicine, The University of Queensland at the Princess Alexandra, Brisbane, QLD 4072, Australia; (T.L.R.H.); (K.S.); (D.W.J.); (G.C.G.)
- Department of Nephrology, The University of Queensland at Princess Alexandra Hospital, Brisbane, QLD 4102, Australia
- Correspondence: ; Tel.: +61-7-3443-8013
| |
Collapse
|
6
|
Myeloid cell-derived coagulation tissue factor is associated with renal tubular damage in mice fed an adenine diet. Sci Rep 2021; 11:12159. [PMID: 34108522 PMCID: PMC8190319 DOI: 10.1038/s41598-021-91586-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 05/13/2021] [Indexed: 02/04/2023] Open
Abstract
Patients with chronic kidney disease (CKD) commonly exhibit hypercoagulability. Increased levels of uremic toxins cause thrombogenicity by increasing tissue factor (TF) expression and activating the extrinsic coagulation cascade. TF is induced in monocytes and macrophages under pathological conditions, such as inflammatory diseases. However, the role of monocyte myeloid cell TF in CKD progression remains unclear. We aimed to clarify this issue, and the present study found that patients with CKD had elevated levels of D-dimer, a marker of fibrin degradation, which was associated with decreased estimated glomerular filtration rate and increased serum levels of uremic toxins, such as indoxyl sulfate. In vitro studies showed that several uremic toxins increased cellular TF levels in monocytic THP-1 cells. Mice with TF specifically deleted in myeloid cells were fed an adenine diet to cause uremic kidney injury. Myeloid TF deletion reduced tubular injury and pro-inflammatory gene expression in the kidneys of adenine-induced CKD but did not improve renal function as measured by plasma creatinine or blood urea nitrogen. Collectively, our findings suggest a novel concept of pathogenesis of coagulation-mediated kidney injury, in which elevated TF levels in monocytes under uremic conditions is partly involved in the development of CKD.
Collapse
|
7
|
Morais C, Rajandram R, Blakeney JS, Iyer A, Suen JY, Johnson DW, Gobe GC, Fairlie DP, Vesey DA. Expression of protease activated receptor-2 is reduced in renal cell carcinoma biopsies and cell lines. PLoS One 2021; 16:e0248983. [PMID: 33765016 PMCID: PMC7993771 DOI: 10.1371/journal.pone.0248983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/09/2021] [Indexed: 01/09/2023] Open
Abstract
Expression of the protease sensing receptor, protease activated receptor-2 (PAR2), is elevated in a variety of cancers and has been promoted as a potential therapeutic target. With the development of potent antagonists for this receptor, we hypothesised that they could be used to treat renal cell carcinoma (RCC). The expression of PAR2 was, therefore, examined in human RCC tissues and selected RCC cell lines. Histologically confirmed cases of RCC, together with paired non-involved kidney tissue, were used to produce a tissue microarray (TMA) and to extract total tissue RNA. Immunohistochemistry and qPCR were then used to assess PAR2 expression. In culture, RCC cell lines versus primary human kidney tubular epithelial cells (HTEC) were used to assess PAR2 expression by qPCR, immunocytochemistry and an intracellular calcium mobilization assay. The TMA revealed an 85% decrease in PAR2 expression in tumour tissue compared with normal kidney tissue. Likewise, qPCR showed a striking reduction in PAR2 mRNA in RCC compared with normal kidney. All RCC cell lines showed lower levels of PAR2 expression than HTEC. In conclusion, we found that PAR2 was reduced in RCC compared with normal kidney and is unlikely to be a target of interest in the treatment of this type of cancer.
Collapse
Affiliation(s)
- Christudas Morais
- Centre for Kidney Disease Research, The University of Queensland, Translational Research Institute, Brisbane, Australia
- Department of Urology, Princess Alexandra Hospital, Brisbane, Australia
| | - Retnagowri Rajandram
- Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Wilayah Persekutuan, Malaysia
| | - Jade S. Blakeney
- Centre for Inflammation and Disease Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Abishek Iyer
- Centre for Inflammation and Disease Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Jacky Y. Suen
- Centre for Inflammation and Disease Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - David W. Johnson
- Centre for Kidney Disease Research, The University of Queensland, Translational Research Institute, Brisbane, Australia
- Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
| | - Glenda C. Gobe
- Centre for Kidney Disease Research, The University of Queensland, Translational Research Institute, Brisbane, Australia
| | - David P. Fairlie
- Centre for Inflammation and Disease Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - David A. Vesey
- Centre for Kidney Disease Research, The University of Queensland, Translational Research Institute, Brisbane, Australia
- Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
- * E-mail:
| |
Collapse
|
8
|
Iyer A, Humphries TLR, Owens EP, Zhao KN, Masci PP, Johnson DW, Nikolic-Paterson D, Gobe GC, Fairlie DP, Vesey DA. PAR2 Activation on Human Kidney Tubular Epithelial Cells Induces Tissue Factor Synthesis, That Enhances Blood Clotting. Front Physiol 2021; 12:615428. [PMID: 33776786 PMCID: PMC7987918 DOI: 10.3389/fphys.2021.615428] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/12/2021] [Indexed: 12/14/2022] Open
Abstract
Coagulation abnormalities and increased risk of atherothrombosis are common in patients with chronic kidney diseases (CKD). Mechanisms that alter renal hemostasis and lead to thrombotic events are not fully understood. Here we show that activation of protease activated receptor-2 (PAR2) on human kidney tubular epithelial cells (HTECs), induces tissue factor (TF) synthesis and secretion that enhances blood clotting. PAR-activating coagulation-associated protease (thrombin), as well as specific PAR2 activators (matriptase, trypsin, or synthetic agonist 2f-LIGRLO-NH2 (2F), induced TF synthesis and secretion that were potently inhibited by PAR2 antagonist, I-191. Thrombin-induced TF was also inhibited by a PAR1 antagonist, Vorapaxar. Peptide activators of PAR1, PAR3, and PAR4 failed to induce TF synthesis. Differential centrifugation of the 2F-conditoned medium sedimented the secreted TF, together with the exosome marker ALG-2 interacting protein X (ALIX), indicating that secreted TF was associated with extracellular vesicles. 2F-treated HTEC conditioned medium significantly enhanced blood clotting, which was prevented by pre-incubating this medium with an antibody for TF. In summary, activation of PAR2 on HTEC stimulates synthesis and secretion of TF that induces blood clotting, and this is attenuated by PAR2 antagonism. Thrombin-induced TF synthesis is at least partly mediated by PAR1 transactivation of PAR2. These findings reveal how underlying hemostatic imbalances might increase thrombosis risk and subsequent chronic fibrin deposition in the kidneys of patients with CKD and suggest PAR2 antagonism as a potential therapeutic strategy for intervening in CKD progression.
Collapse
Affiliation(s)
- Abishek Iyer
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- Centre for Inflammation and Disease Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Tyrone L. R. Humphries
- Centre for Kidney Disease Research, Translational Research Institute, Faculty of Medicine at the Princess Alexandra Hospital, The University of Queensland, Woolloongabba, QLD, Australia
| | - Evan P. Owens
- Centre for Kidney Disease Research, Translational Research Institute, Faculty of Medicine at the Princess Alexandra Hospital, The University of Queensland, Woolloongabba, QLD, Australia
| | - Kong-Nan Zhao
- Centre for Venomics Research, Faculty of Medicine, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Paul P. Masci
- Centre for Venomics Research, Faculty of Medicine, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - David W. Johnson
- Centre for Kidney Disease Research, Translational Research Institute, Faculty of Medicine at the Princess Alexandra Hospital, The University of Queensland, Woolloongabba, QLD, Australia
- Department of Nephrology, The University of Queensland at Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| | - David Nikolic-Paterson
- Department of Nephrology, Monash Medical Centre and Monash University Centre for Inflammatory Diseases, Melbourne, VIC, Australia
| | - Glenda C. Gobe
- Centre for Kidney Disease Research, Translational Research Institute, Faculty of Medicine at the Princess Alexandra Hospital, The University of Queensland, Woolloongabba, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - David P. Fairlie
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- Centre for Inflammation and Disease Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - David A. Vesey
- Centre for Kidney Disease Research, Translational Research Institute, Faculty of Medicine at the Princess Alexandra Hospital, The University of Queensland, Woolloongabba, QLD, Australia
- Department of Nephrology, The University of Queensland at Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| |
Collapse
|
9
|
A direct thrombin inhibitor, dabigatran etexilate protects from renal fibrosis by inhibiting protease activated receptor-1. Eur J Pharmacol 2020; 893:173838. [PMID: 33359646 DOI: 10.1016/j.ejphar.2020.173838] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/16/2020] [Accepted: 12/22/2020] [Indexed: 11/23/2022]
Abstract
Chronic kidney disease (CKD) involves interstitial fibrosis as an influential underlying pathological process associated with compromised renal function regardless of etiological cause of the injury. The tubulointerstitial fibrosis is found to be well correlated with declining renal function and its subsequent culmination into renal failure. Given the prominent role of thrombin in multiple diseases, it was tempting for us to investigate the outcome of a direct thrombin inhibitor in renal injury. We investigated the involvement of thrombin in renal injury and fibrosis by using an FDA approved orally active, direct thrombin inhibitor, dabigatran etexilate (DB). We used a robust experimental model of unilateral ureteral obstruction (UUO)-induced renal injury which shows progressive tubulointerstitial fibrosis (TIF) along with tubular injury and inflammation. The obstructed kidney showed severe TIF as compared to control kidneys. The administration of DB significantly inhibited UUO-induced collagen-1 and TIF by inhibition of thrombin activated protease activated receptor (PAR)-1 expression in fibrotic kidney. In addition, DB administration improved histoarchitecture of obstructed kidney, inhibited TGF-β and SNAI2-induced epithelial-mesenchymal transition (EMT) program. Our study highlights the importance of thrombin signalling in TIF and provides strong evidences to support the notion that a direct thrombin inhibitor ameliorates TIF by PAR-1 mediated mechanism.
Collapse
|
10
|
Iio A, Kaji K, Kaji N, Hori M, Yonezawa T, Momoi Y, Maeda S. Expression analysis of protease-activated receptor-2 in cats. Vet Immunol Immunopathol 2020; 229:110115. [PMID: 32932190 DOI: 10.1016/j.vetimm.2020.110115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 08/24/2020] [Accepted: 08/27/2020] [Indexed: 10/23/2022]
Abstract
Chronic kidney disease (CKD) is a common disease in geriatric cats. Despite its high prevalence, the pathogenesis of feline CKD is poorly understood. Recently, there has been increasing evidence for the role of protease-activated receptor-2 (PAR-2) in the progression of CKD in humans and rodents. However, the role of PAR-2 in feline CKD has not been evaluated. In this study, we determined nucleotide sequence of feline PAR-2 from the kidney, evaluated PAR-2 mRNA and protein expression in normal feline tissues, and analyzed functional expression in the feline kidney epithelial cell line Crandell-Rees Feline Kidney (CRFK). The open reading frame of feline PAR-2 comprised 1,194 bp and encoded 397 amino acids, showing 90%, 90%, and 85% identities to human, dog, and mouse PAR-2, respectively. In healthy cats, expression levels of the PAR-2 mRNA and protein were relatively higher in the gastrointestinal tract and kidney, and was lowest in the heart. The feline PAR-2 protein expression was confirmed, and stimulation of trypsin and PAR-2 agonists induced a prompt increase in the intracellular calcium ion concentration in CRFK cells. The present study will provide fundamental information for investigation of the involvement of PAR-2 in the pathogenesis of CKD in cats.
Collapse
Affiliation(s)
- Aki Iio
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Kenjiro Kaji
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Noriyuki Kaji
- Department of Veterinary Pharmacology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Masatoshi Hori
- Department of Veterinary Pharmacology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Tomohiro Yonezawa
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Yasuyuki Momoi
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Shingo Maeda
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan.
| |
Collapse
|
11
|
Kanazawa N, Iyoda M, Tachibana S, Matsumoto K, Wada Y, Suzuki T, Iseri K, Shibata T. Therapeutic Potential of Thrombomodulin in Renal Fibrosis of Nephrotoxic Serum Nephritis in Wistar-Kyoto Rats. Kidney Blood Press Res 2020; 45:391-406. [PMID: 32146474 DOI: 10.1159/000506286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/30/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Recombinant human soluble thrombomodulin (rhTM) was approved in 2008 and has been used for treatment of disseminated intravascular coagulation in Japan. The antifibrotic effects of rhTM in acute exacerbation of idiopathic pulmonary fibrosis are well established, but the therapeutic potential of rhTM in renal fibrosis remains poorly understood. METHODS Nephrotoxic serum nephritis (NTS-N) was induced in 22 female Wistar-Kyoto (WKY) rats on day 0. Rats were administered either rhTM or vehicle intraperitoneally, every day from day 4 to day 55. Rats were sacrificed on day 56 when renal fibrosis was established and renal morphological investigations were performed. In vitro, rat renal fibroblasts (NRK-49F) were pretreated with rhTM or saline, and expression levels of profibrogenic gene induced by thrombin were analyzed by real-time reverse transcription polymerase chain reaction. RESULTS Compared to WKY-GN-vehicle rats, the body weights of WKY-GN-rhTM rats were significantly greater on day 55. By day 56, rhTM had significantly reduced serum creatinine levels in NTS-N. On the other hand, urinary protein excretion was comparable between the two treatment groups throughout the study. The percentage of Masson trichrome-positive areas in WKY-GN-rhTM rats was significantly lower compared to that in WKY-GN-vehicle rats. Glomerular fibrin deposition was significantly reduced in WKY-GN-rhTM rats. In addition, rhTM significantly reduced the renal cortical mRNA expression levels of TNF-α, Toll-like receptor 4, MYD88, TGF-β, αSMA, collagen I, collagen III, fibronectin, and protease-activated receptor 1 (PAR1), a thrombin receptor. In vitro, thrombin stimulation of NRK-49F cells significantly enhanced the mRNA expression levels of αSMA and PAR1, and these upregulations were significantly reduced by pretreatment with rhTM. CONCLUSIONS Administration of rhTM after establishment of crescentic glomerulonephritis (GN) attenuated the subsequent development of renal fibrosis in NTS-N, possibly in part by inhibiting thrombin-mediated fibrogenesis. Our results suggest that rhTM may offer a therapeutic option for limiting the progression of chronic kidney disease in crescentic GN.
Collapse
Affiliation(s)
- Nobuhiro Kanazawa
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Masayuki Iyoda
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan,
| | - Shohei Tachibana
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Kei Matsumoto
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yukihiro Wada
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Taihei Suzuki
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Ken Iseri
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Takanori Shibata
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
12
|
Ma FY, Han Y, Ozols E, Chew P, Vesey DA, Gobe GC, Morais C, Lohman RJ, Suen JY, Johnson DW, Fairlie DP, Nikolic-Paterson DJ. Protease-activated receptor 2 does not contribute to renal inflammation or fibrosis in the obstructed kidney. Nephrology (Carlton) 2019; 24:983-991. [PMID: 31314137 DOI: 10.1111/nep.13635] [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] [Accepted: 07/08/2019] [Indexed: 12/19/2022]
Abstract
AIM Protease-activated receptor 2 (PAR2) has been implicated in the development of renal inflammation and fibrosis. In particular, activation of PAR2 in cultured tubular epithelial cells induces extracellular signal-regulated kinase signalling and secretion of fibronectin, C-C Motif Chemokine Ligand 2 (CCL2) and transforming growth factor-β1 (TGF-β1), suggesting a role in tubulointerstitial inflammation and fibrosis. We tested this hypothesis in unilateral ureteric obstruction (UUO) in which ongoing tubular epithelial cell damage drives tubulointerstitial inflammation and fibrosis. METHODS Unilateral ureteric obstruction surgery was performed in groups (n = 9/10) of Par2-/- and wild type (WT) littermate mice which were killed 7 days later. Non-experimental mice were controls. RESULTS Wild type mice exhibited a 5-fold increase in Par2 messenger RNA (mRNA) levels in the UUO kidney. In situ hybridization localized Par2 mRNA expression to tubular epithelial cells in normal kidney, with a marked increase in Par2 mRNA expression by tubular cells, including damaged tubular cells, in WT UUO kidney. Tubular damage (tubular dilation, increased KIM-1 and decreased α-Klotho expression) and tubular signalling (extracellular signal-regulated kinase phosphorylation) seen in WT UUO were not altered in Par2-/- UUO. In addition, macrophage infiltration, up-regulation of M1 (NOS2) and M2 (CD206) macrophage markers, and up-regulation of pro-inflammatory molecules (tumour necrosis factor, CCL2, interleukin-36α) in WT UUO kidney were unchanged in Par2-/- UUO. Finally, the accumulation of α-SMA+ myofibroblasts, deposition of collagen IV and expression of pro-fibrotic factors (CTGF, TGF-β1) were not different between WT and Par2-/- UUO mice. CONCLUSION Protease-activated receptor 2 expression is substantially up-regulated in tubular epithelial cells in the obstructed kidney, but this does not contribute to the development of tubular damage, renal inflammation or fibrosis.
Collapse
Affiliation(s)
- Frank Y Ma
- Department of Nephrology, Monash Medical Centre, Monash University, Melbourne, Victoria, Australia.,Centre for Inflammatory Diseases, Monash University, Melbourne, Victoria, Australia
| | - Yingjie Han
- Department of Nephrology, Monash Medical Centre, Monash University, Melbourne, Victoria, Australia.,Centre for Inflammatory Diseases, Monash University, Melbourne, Victoria, Australia
| | - Elyce Ozols
- Department of Nephrology, Monash Medical Centre, Monash University, Melbourne, Victoria, Australia.,Centre for Inflammatory Diseases, Monash University, Melbourne, Victoria, Australia
| | - Phyllis Chew
- Department of Nephrology, Monash Medical Centre, Monash University, Melbourne, Victoria, Australia.,Centre for Inflammatory Diseases, Monash University, Melbourne, Victoria, Australia
| | - David A Vesey
- Centre for Kidney Disease Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Glenda C Gobe
- Centre for Kidney Disease Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Christudas Morais
- Centre for Kidney Disease Research, The University of Queensland, Brisbane, Queensland, Australia.,Department of Urology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Rink-Jan Lohman
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Jacky Y Suen
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - David W Johnson
- Centre for Kidney Disease Research, The University of Queensland, Brisbane, Queensland, Australia.,Translational Research Institute, Brisbane, Queensland, Australia.,Department of Nephrology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - David P Fairlie
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - David J Nikolic-Paterson
- Department of Nephrology, Monash Medical Centre, Monash University, Melbourne, Victoria, Australia.,Centre for Inflammatory Diseases, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
13
|
Han Y, Tian L, Ma F, Tesch G, Vesey DA, Gobe GC, Lohman RJ, Morais C, Suen JY, Fairlie DP, Nikolic-Paterson DJ. Pharmacological inhibition of protease-activated receptor-2 reduces crescent formation in rat nephrotoxic serum nephritis. Clin Exp Pharmacol Physiol 2019; 46:456-464. [PMID: 30811624 DOI: 10.1111/1440-1681.13077] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/20/2019] [Accepted: 02/22/2019] [Indexed: 01/06/2023]
Abstract
Glomerular crescent formation is a hallmark of rapidly progressive forms of glomerulonephritis. Thrombosis and macrophage infiltration are features of crescent formation in human and experimental kidney disease. Protease-activated receptor-2 (PAR-2) is a G-protein coupled receptor that links coagulation and inflammation. This study investigated whether pharmacological inhibition of PAR-2 can suppress glomerular crescent formation in rat nephrotoxic serum nephritis (NTN). Disease was induced in Wistar Kyoto rats by immunisation with sheep IgG followed by administration of sheep nephrotoxic serum. Rats (n = 8/group) received the PAR-2 antagonist (GB88, 10 mg/kg/p.o.), vehicle or no treatment starting 3 days before nephrotoxic serum injection and continuing until day 14. Vehicle and untreated rats developed thrombosis and macrophage infiltration in the glomerular tuft and Bowman's space in conjunction with prominent crescent formation. Activation of JNK signalling and proliferation in parietal epithelial cells was associated with crescent formation. GB88 treatment significantly reduced crescent formation with a substantial reduction in glomerular thrombosis, reduced macrophage infiltration in Bowman's space, and reduced activation of parietal epithelial cells. However, GB88 did not protect against the development of proteinuria, renal function impairment, inflammation or tubular cell damage in the NTN model. In conclusion, PAR-2 plays a specific role in glomerular crescent formation by promoting glomerular thrombosis, macrophage accumulation in Bowman's space and activation of parietal epithelial cells.
Collapse
Affiliation(s)
- Yingjie Han
- Department of Nephrology, Monash Medical Centre, Clayton, Victoria, Australia.,Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria, Australia
| | - Lifang Tian
- Department of Nephrology, Monash Medical Centre, Clayton, Victoria, Australia.,Department of Nephrology, Second Affiliated Hospital of Xi'an, Shannxi Province, China
| | - Frank Ma
- Department of Nephrology, Monash Medical Centre, Clayton, Victoria, Australia.,Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria, Australia
| | - Greg Tesch
- Department of Nephrology, Monash Medical Centre, Clayton, Victoria, Australia.,Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria, Australia
| | - David A Vesey
- Faculty of Medicine, Centre for Kidney Disease Research, Translational Research Institute, The University of Queensland, Brisbane, Queensland, Australia.,Department of Nephrology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Glenda C Gobe
- Faculty of Medicine, Centre for Kidney Disease Research, Translational Research Institute, The University of Queensland, Brisbane, Queensland, Australia.,Faculty of Medicine, School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Rink-Jan Lohman
- Centre for Inflammation and Disease Research and ARC Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Christudas Morais
- Department of Nephrology, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,Centre for Inflammation and Disease Research and ARC Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Jacky Y Suen
- Centre for Inflammation and Disease Research and ARC Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - David P Fairlie
- Centre for Inflammation and Disease Research and ARC Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - David J Nikolic-Paterson
- Department of Nephrology, Monash Medical Centre, Clayton, Victoria, Australia.,Department of Medicine, Monash Medical Centre, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
14
|
Guan Y, Nakano D, Zhang Y, Li L, Liu W, Nishida M, Kuwabara T, Morishita A, Hitomi H, Mori K, Mukoyama M, Masaki T, Hirano K, Nishiyama A. A protease-activated receptor-1 antagonist protects against podocyte injury in a mouse model of nephropathy. J Pharmacol Sci 2017; 135:S1347-8613(17)30128-7. [PMID: 29110957 DOI: 10.1016/j.jphs.2017.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/13/2017] [Accepted: 08/15/2017] [Indexed: 12/19/2022] Open
Abstract
The kidney expresses protease-activated receptor-1 (PAR-1). PAR-1 is known as a thrombin receptor, but its role in kidney injury is not well understood. In this study, we examined the contribution of PAR-1 to kidney glomerular injury and the effects of its inhibition on development of nephropathy. Mice were divided into 3 groups: control, doxorubicin + vehicle (15 mg/kg doxorubicin and saline) and doxorubicin + Q94 (doxorubicin at 15 mg/kg and the PAR-1 antagonist Q94 at 5 mg/kg/d) groups. Where indicated, doxorubicin was administered intravenously and PAR-1 antagonist or saline vehicle by subcutaneous osmotic mini-pump. PAR-1 expression was increased in glomeruli of mice treated with doxorubicin. Q94 treatment significantly suppressed the increased albuminuria in these nephropathic mice. Pathological analysis showed that Q94 treatment significantly attenuated periodic acid-Schiff and desmin staining, indicators of podocyte injury, and also decreased glomerular levels of podocin and nephrin. Furthermore, thrombin increased intracellular calcium levels in podocytes. This increase was suppressed by Q94 and Rox4560, a transient receptor potential cation channel (TRPC)3/6 antagonist. In addition, both Q94 and Rox4560 suppressed the doxorubicin-induced increase in activities of caspase-9 and caspase-3 in podocytes. These data suggested that PAR-1 contributes to development of podocyte and glomerular injury and that PAR-1 antagonists have therapeutic potential.
Collapse
Affiliation(s)
- Yu Guan
- Department of Pharmacology, Kagawa University, Kagawa, Japan
| | - Daisuke Nakano
- Department of Pharmacology, Kagawa University, Kagawa, Japan.
| | - Yifan Zhang
- Department of Pharmacology, Kagawa University, Kagawa, Japan
| | - Lei Li
- Department of Pharmacology, Kagawa University, Kagawa, Japan
| | - Wenhua Liu
- Department of Cardiovascular Physiology, Kagawa University, Kagawa, Japan
| | - Motohiro Nishida
- Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
| | - Takashige Kuwabara
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Asahiro Morishita
- Department of Gastroenterology & Neurology, Kagawa University, Kagawa, Japan
| | - Hirofumi Hitomi
- Department of Pharmacology, Kagawa University, Kagawa, Japan
| | - Kiyoshi Mori
- Department of Nephrology and Kidney Research, Shizuoka General Hospital, Shizuoka, Japan
| | - Masashi Mukoyama
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology & Neurology, Kagawa University, Kagawa, Japan
| | - Katsuya Hirano
- Department of Cardiovascular Physiology, Kagawa University, Kagawa, Japan
| | - Akira Nishiyama
- Department of Pharmacology, Kagawa University, Kagawa, Japan
| |
Collapse
|
15
|
Latosinska A, Hulko M, Speidel R, Mischak H, Storr M, Krause B. Removal of Cell-Activating Substances Using Dialyzers With Various Permeability Profiles. Artif Organs 2017; 42:78-87. [DOI: 10.1111/aor.12952] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 03/13/2017] [Accepted: 03/14/2017] [Indexed: 12/24/2022]
Affiliation(s)
| | - Michael Hulko
- Gambro Dialysatoren GmbH (part of Baxter International Inc.), Research & Development; Hechingen Germany
| | - Rose Speidel
- Gambro Dialysatoren GmbH (part of Baxter International Inc.), Research & Development; Hechingen Germany
| | | | - Markus Storr
- Gambro Dialysatoren GmbH (part of Baxter International Inc.), Research & Development; Hechingen Germany
| | - Bernd Krause
- Gambro Dialysatoren GmbH (part of Baxter International Inc.), Research & Development; Hechingen Germany
| |
Collapse
|
16
|
Dong D, Cai GY, Ning YC, Wang JC, Lv Y, Hong Q, Cui SY, Fu B, Guo YN, Chen XM. Alleviation of senescence and epithelial-mesenchymal transition in aging kidney by short-term caloric restriction and caloric restriction mimetics via modulation of AMPK/mTOR signaling. Oncotarget 2017; 8:16109-16121. [PMID: 28147330 PMCID: PMC5369951 DOI: 10.18632/oncotarget.14884] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/16/2017] [Indexed: 01/01/2023] Open
Abstract
Renal fibrosis contributes to declining renal function in the elderly. What is unclear however, is whether epithelial-mesenchymal transition (EMT) contributes to this age-related renal fibrosis. Here, we analyzed indicators of EMT during kidney aging and investigated the protective effects and mechanisms of short-term regimens of caloric restriction (CR) or caloric restriction mimetics (CRMs), including resveratrol and metformin. High glucose was used to induce premature senescence and EMT in human primary proximal tubular cells (PTCs) in vitro. To test the role of AMPK-mTOR signaling, siRNA was used to deplete AMPK. Cellular senescence and AMPK-mTOR signaling markers associated with EMT were detected. CR or CRMs treatment alleviated age-related EMT in aging kidneys, which was accompanied by activation of AMPK-mTOR signaling. High glucose induced premature senescence and EMT in PTCs in vitro, which was accompanied by down-regulation of AMPK/mTOR signaling. CRMs alleviated high glucose-induced senescence and EMT via stimulation of AMPK/mTOR signaling. Activation of AMPK/mTOR signaling protected PTCs from high glucose-induced EMT and cellular senescence. Short-term regimens of CR and CRMs alleviated age-related EMT via AMPK-mTOR signaling, suggesting a potential approach to reducing renal fibrosis during aging.
Collapse
Affiliation(s)
- Dan Dong
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Beijing, P. R. China.,Department of Nephrology, First Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Guang-Yan Cai
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Beijing, P. R. China
| | - Yi-Chun Ning
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Beijing, P. R. China
| | - Jing-Chao Wang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Beijing, P. R. China
| | - Yang Lv
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Beijing, P. R. China
| | - Quan Hong
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Beijing, P. R. China
| | - Shao-Yuan Cui
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Beijing, P. R. China
| | - Bo Fu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Beijing, P. R. China
| | - Ya-Nan Guo
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Beijing, P. R. China
| | - Xiang-Mei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center of Kidney Diseases, Beijing, P. R. China
| |
Collapse
|
17
|
Palygin O, Ilatovskaya DV, Staruschenko A. Protease-activated receptors in kidney disease progression. Am J Physiol Renal Physiol 2016; 311:F1140-F1144. [PMID: 27733370 DOI: 10.1152/ajprenal.00460.2016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 10/07/2016] [Indexed: 01/05/2023] Open
Abstract
Protease-activated receptors (PARs) are members of a well-known family of transmembrane G protein-coupled receptors (GPCRs). Four PARs have been identified to date, of which PAR1 and PAR2 are the most abundant receptors, and have been shown to be expressed in the kidney vascular and tubular cells. PAR signaling is mediated by an N-terminus tethered ligand that can be unmasked by serine protease cleavage. The receptors are activated by endogenous serine proteases, such as thrombin (acts on PARs 1, 3, and 4) and trypsin (PAR2). PARs can be involved in glomerular, microvascular, and inflammatory regulation of renal function in both normal and pathological conditions. As an example, it was shown that human glomerular epithelial and mesangial cells express PARs, and these receptors are involved in the pathogenesis of crescentic glomerulonephritis, glomerular fibrin deposition, and macrophage infiltration. Activation of these receptors in the kidney also modulates renal hemodynamics and glomerular filtration rate. Clinical studies further demonstrated that the concentration of urinary thrombin is associated with glomerulonephritis and type 2 diabetic nephropathy; thus, molecular and functional mechanisms of PARs activation can be directly involved in renal disease progression. We briefly discuss here the recent literature related to activation of PAR signaling in glomeruli and the kidney in general and provide some examples of PAR1 signaling in glomeruli podocytes.
Collapse
Affiliation(s)
- Oleg Palygin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Daria V Ilatovskaya
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | |
Collapse
|
18
|
Protease-activated receptor-1 deficiency protects against streptozotocin-induced diabetic nephropathy in mice. Sci Rep 2016; 6:33030. [PMID: 27618774 PMCID: PMC5020504 DOI: 10.1038/srep33030] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 08/18/2016] [Indexed: 12/19/2022] Open
Abstract
Endogenously administered activated protein C ameliorates diabetic nephropathy (DN) in a protease-activated receptor-1 (PAR-1)-dependent manner, suggesting that PAR-1 activation limits the progression of DN. Activation of PAR-1 in fibroblast-like cells, however, induces proliferation and extracellular matrix production, thereby driving fibrotic disease. Considering the key role of mesangial proliferation and extracellular matrix production during DN, PAR-1 may in fact potentiate diabetes-induced kidney injury. To determine the net effect of PAR-1 in DN, streptozotocin-induced DN was studied in wild type and PAR-1 deficient mice. Subsequent mechanistic insight was obtained by assessing profibrotic responses of mesangial and tubular epithelial cells in vitro, following PAR-1 stimulation and inhibition. Despite having similar glucose levels, PAR-1 deficient mice developed less kidney damage after induction of diabetes, as evidenced by diminished proteinuria, plasma cystatin C levels, expansion of the mesangial area, and tubular atrophy. In vitro, PAR-1 signaling in mesangial cells led to increased proliferation and expression of matrix proteins fibronectin and collagen IV. Conversely, a reduction in both proliferation and fibronectin deposition was observed in diabetic PAR-1 deficient mice. Overall, we show that PAR-1 plays an important role in the development of DN and PAR-1 might therefore be an attractive therapeutic target to pursue in DN.
Collapse
|
19
|
Madhusudhan T, Kerlin BA, Isermann B. The emerging role of coagulation proteases in kidney disease. Nat Rev Nephrol 2015; 12:94-109. [PMID: 26592189 DOI: 10.1038/nrneph.2015.177] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A role of coagulation proteases in kidney disease beyond their function in normal haemostasis and thrombosis has long been suspected, and studies performed in the past 15 years have provided novel insights into the mechanisms involved. The expression of protease-activated receptors (PARs) in renal cells provides a molecular link between coagulation proteases and renal cell function and revitalizes research evaluating the role of haemostasis regulators in renal disease. Renal cell-specific expression and activity of coagulation proteases, their regulators and their receptors are dynamically altered during disease processes. Furthermore, renal inflammation and tissue remodelling are not only associated, but are causally linked with altered coagulation activation and protease-dependent signalling. Intriguingly, coagulation proteases signal through more than one receptor or induce formation of receptor complexes in a cell-specific manner, emphasizing context specificity. Understanding these cell-specific signalosomes and their regulation in kidney disease is crucial to unravelling the pathophysiological relevance of coagulation regulators in renal disease. In addition, the clinical availability of small molecule targeted anticoagulants as well as the development of PAR antagonists increases the need for in-depth knowledge of the mechanisms through which coagulation proteases might regulate renal physiology.
Collapse
Affiliation(s)
- Thati Madhusudhan
- Institute of Clinical Chemistry and Pathobiochemistry, Medical Faculty, Otto-von-Guericke-University, Magdeburg, Leipziger Strasse 44, Magdeburg D-39120, Germany
| | - Bryce A Kerlin
- Center for Clinical and Translational Research, Nationwide Children's Hospital, 700 Children's Drive, W325 Columbus, Ohio 43205, USA
| | - Berend Isermann
- Institute of Clinical Chemistry and Pathobiochemistry, Medical Faculty, Otto-von-Guericke-University, Magdeburg, Leipziger Strasse 44, Magdeburg D-39120, Germany
| |
Collapse
|
20
|
Ishibashi Y, Matsui T, Fukami K, Ueda S, Okuda S, Yamagishi SI. Rivaroxaban inhibits oxidative and inflammatory reactions in advanced glycation end product-exposed tubular cells by blocking thrombin/protease-activated receptor-2 system. Thromb Res 2015; 135:770-3. [PMID: 25636461 DOI: 10.1016/j.thromres.2015.01.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 12/03/2014] [Accepted: 01/19/2015] [Indexed: 11/27/2022]
Affiliation(s)
- Yuji Ishibashi
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Japan
| | - Takanori Matsui
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Japan
| | - Kei Fukami
- Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Seiji Ueda
- Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Seiya Okuda
- Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Sho-ichi Yamagishi
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine, Kurume, Japan.
| |
Collapse
|
21
|
Suen JY, Cotterell A, Lohman RJ, Lim J, Han A, Yau MK, Liu L, Cooper MA, Vesey DA, Fairlie DP. Pathway-selective antagonism of proteinase activated receptor 2. Br J Pharmacol 2014; 171:4112-24. [PMID: 24821440 DOI: 10.1111/bph.12757] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 04/04/2014] [Accepted: 04/30/2014] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Proteinase activated receptor 2 (PAR2) is a GPCR associated with inflammation, metabolism and disease. Clues to understanding how to block PAR2 signalling associated with disease without inhibiting PAR2 activation in normal physiology could be provided by studies of biased signalling. EXPERIMENTAL APPROACH PAR2 ligand GB88 was profiled for PAR2 agonist and antagonist properties by several functional assays associated with intracellular G-protein-coupled signalling in vitro in three cell types and with PAR2-induced rat paw oedema in vivo. KEY RESULTS In HT29 cells, GB88 was a PAR2 antagonist in terms of Ca(2+) mobilization and PKC phosphorylation, but a PAR2 agonist in attenuating forskolin-induced cAMP accumulation, increasing ERK1/2 phosphorylation, RhoA activation, myosin phosphatase phosphorylation and actin filament rearrangement. In CHO-hPAR2 cells, GB88 inhibited Ca(2+) release, but activated G(i/o) and increased ERK1/2 phosphorylation. In human kidney tubule cells, GB88 inhibited cytokine secretion (IL6, IL8, GM-CSF, TNF-α) mediated by PAR2. A rat paw oedema induced by PAR2 agonists was also inhibited by orally administered GB88 and compared with effects of locally administered inhibitors of G-protein coupled pathways. CONCLUSIONS AND IMPLICATIONS GB88 is a biased antagonist of PAR2 that selectively inhibits PAR2/G(q/11)/Ca(2+)/PKC signalling, leading to anti-inflammatory activity in vivo, while being an agonist in activating three other PAR2-activated pathways (cAMP, ERK, Rho) in human cells. These findings highlight opportunities to design drugs to block specific PAR2-linked signalling pathways in disease, without blocking beneficial PAR2 signalling in normal physiology, and to dissect PAR2-associated mechanisms of disease in vivo.
Collapse
Affiliation(s)
- J Y Suen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Yiu WH, Wong DWL, Chan LYY, Leung JCK, Chan KW, Lan HY, Lai KN, Tang SCW. Tissue kallikrein mediates pro-inflammatory pathways and activation of protease-activated receptor-4 in proximal tubular epithelial cells. PLoS One 2014; 9:e88894. [PMID: 24586431 PMCID: PMC3931644 DOI: 10.1371/journal.pone.0088894] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 01/13/2014] [Indexed: 12/11/2022] Open
Abstract
Tissue kallikrein (KLK1) expression is up-regulated in human diabetic kidney tissue and induced by high glucose (HG) in human proximal tubular epithelial cells (PTEC). Since the kallikrein-kinin system (KKS) has been linked to cellular inflammatory process in many diseases, it is likely that KLK1 expression may mediate the inflammatory process during the development of diabetic nephropathy. In this study, we explored the role of KLK1 in tubular pro-inflammatory responses under the diabetic milieu. Recombinant KLK1 stimulated the production of inflammatory cytokines in PTEC via the activation of p42/44 and p38 MAPK signaling pathways. Molecular knockdown of endogenous KLK1 expression by siRNA transfection in PTEC attenuated advanced glycation end-products (AGE)-induced IL-8 and ICAM-1 productions in vitro. Interestingly, exposure of PTEC to KLK1 induced the expression of protease-activated receptors (PARs). There was a 2.9-fold increase in PAR-4, 1.4-fold increase in PAR-1 and 1.2-fold increase in PAR-2 mRNA levels. Activation of PAR-4 by a selective agonist was found to elicit the pro-inflammatory and pro-fibrotic phenotypes in PTEC while blockade of the receptor by specific antagonist attenuated high glucose-induced IL-6, CCL-2, CTGF and collagen IV expression. Calcium mobilization by the PAR-4 agonist in PTEC was desensitized by pretreatment with KLK1. Consistent with these in vitro findings, there was a markedly up-regulation of tubular PAR-4 expression in human diabetic renal cortical tissues. Together, these results suggest that up-regulation of KLK1 in tubular epithelial cells may mediate pro-inflammatory pathway and PAR activation during diabetic nephropathy and provide a new therapeutic target for further investigation.
Collapse
Affiliation(s)
- Wai Han Yiu
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | - Dickson W. L. Wong
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | - Loretta Y. Y. Chan
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | - Joseph C. K. Leung
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | - Kwok Wah Chan
- Department of Pathology, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | - Hui Yao Lan
- Department of Medicine and Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Kar Neng Lai
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | - Sydney C. W. Tang
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
- * E-mail:
| |
Collapse
|
23
|
Gieseler F, Ungefroren H, Settmacher U, Hollenberg MD, Kaufmann R. Proteinase-activated receptors (PARs) - focus on receptor-receptor-interactions and their physiological and pathophysiological impact. Cell Commun Signal 2013; 11:86. [PMID: 24215724 PMCID: PMC3842752 DOI: 10.1186/1478-811x-11-86] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 10/25/2013] [Indexed: 02/07/2023] Open
Abstract
Proteinase-activated receptors (PARs) are a subfamily of G protein-coupled receptors (GPCRs) with four members, PAR1, PAR2, PAR3 and PAR4, playing critical functions in hemostasis, thrombosis, embryonic development, wound healing, inflammation and cancer progression. PARs are characterized by a unique activation mechanism involving receptor cleavage by different proteinases at specific sites within the extracellular amino-terminus and the exposure of amino-terminal “tethered ligand“ domains that bind to and activate the cleaved receptors. After activation, the PAR family members are able to stimulate complex intracellular signalling networks via classical G protein-mediated pathways and beta-arrestin signalling. In addition, different receptor crosstalk mechanisms critically contribute to a high diversity of PAR signal transduction and receptor-trafficking processes that result in multiple physiological effects. In this review, we summarize current information about PAR-initiated physical and functional receptor interactions and their physiological and pathological roles. We focus especially on PAR homo- and heterodimerization, transactivation of receptor tyrosine kinases (RTKs) and receptor serine/threonine kinases (RSTKs), communication with other GPCRs, toll-like receptors and NOD-like receptors, ion channel receptors, and on PAR association with cargo receptors. In addition, we discuss the suitability of these receptor interaction mechanisms as targets for modulating PAR signalling in disease.
Collapse
Affiliation(s)
| | | | | | | | - Roland Kaufmann
- Department of General, Visceral and Vascular Surgery, Experimental Transplantation Surgery, Jena University Hospital, Drackendorfer Str, 1, D-07747, Jena, Germany.
| |
Collapse
|
24
|
Li T, Zhang P, Yuan B, Zhao D, Chen Y, Zhang X. Thrombin-induced TGF-β1 pathway: a cause of communicating hydrocephalus post subarachnoid hemorrhage. Int J Mol Med 2013; 31:660-6. [PMID: 23338707 DOI: 10.3892/ijmm.2013.1253] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 12/11/2012] [Indexed: 11/06/2022] Open
Abstract
The mechanism of communicating hydrocephalus after subarachnoid hemorrhage (SAH) remains unclear. Revealing a signaling cascade may provide significant insights into the molecular etiology of the accumulation of cerebrospinal fluid (CSF) in cerebral compartments during SAH. To investigate the mechanism of the communicating hydrocephalus following SAH, we infused CSF with thrombin (TH), resulting in proinflammatory and proliferative responses in rat meninges of SAH. The effect of TH could be completely blocked by a transforming growth factor β1 (TGF-β1) inhibitor, SB-431542, suggesting that TH-stimulated proliferation of meninges is through the TGF-β1 signaling pathway. The cascade of TGF β1-Smad3 was significantly upregulated by TH, which, in turn, stimulated the proliferation of subarachnoid meninges. TH-induced overexpression of TGF-β1 and activation of its downstream factors might be a mechanism of communicating hydrocephalus after SAH.
Collapse
Affiliation(s)
- Tong Li
- Department of Neurology, First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, PR China
| | | | | | | | | | | |
Collapse
|
25
|
Hamamoto S, Yasui T, Okada A, Hirose M, Matsui Y, Kon S, Sakai F, Kojima Y, Hayashi Y, Tozawa K, Uede T, Kohri K. Crucial role of the cryptic epitope SLAYGLR within osteopontin in renal crystal formation of mice. J Bone Miner Res 2011; 26:2967-77. [PMID: 21898593 DOI: 10.1002/jbmr.495] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Osteopontin plays a crucial role in the formation of renal calcium crystals, which are primarily induced by renal tubular cell injury, especially mitochondrial damage. We have previously shown that the impaired Arg-Gly-Asp (RGD) sequence of osteopontin inhibits renal crystal formation by using OPN-transgenic mice and OPN-knockout (OPN-KO) mice. Here, we investigated the effects of an antimurine osteopontin antibody (35B6-Ab) that specifically reacts with the (162) SLAYGLR(168) sequence, which is exposed by thrombin cleavage and is located adjacent to the RGD sequence, on renal crystal formation. Renal crystals induced by daily administration of glyoxylate over 9 days (from days 1 to 9) in a murine model were sporadically detected in the renal tubular cells at the corticomedullary junction, where thrombin-cleaved osteopontin expression was also coincidentally detected. On days 0, 3, 6, and 9, 35B6-Ab administration inhibited renal crystal formation and induced significant morphological changes in a dose-dependent manner (250, 500, and 1000 µg per mouse). Scanning electron microscopy showed that the crystals in 35B6-Ab-treated mice were aberrantly formed and their density was low; in contrast, the crystals in untreated mice that were not administered 35B6-Ab had a radial pattern of growth (rosette petal-like crystals), and their density was high. Microstructure analysis of renal tubular cells by transmission electron microscopy revealed that untreated mice showed collapsed mitochondria in the flattened cytoplasm of renal tubular cells, unlike the corresponding structures in 35B6-Ab-treated mice, in which renal tubular cell injury was inhibited. In vitro, 35B6-Ab was found to inhibit the attachment of (14) C-labeled crystals to renal tubular culture cells and reduce morphological damage to these cells. We conclude that thrombin-cleaved osteopontin plays an important role in formation of renal calcium crystals and that 35B6-Ab contributes to the remarkable inhibition of early-stage renal crystal formation by preventing renal tubular cell injury and crystal-cell attachment.
Collapse
Affiliation(s)
- Shuzo Hamamoto
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Bae JS, Kim IS, Rezaie AR. Thrombin down-regulates the TGF-beta-mediated synthesis of collagen and fibronectin by human proximal tubule epithelial cells through the EPCR-dependent activation of PAR-1. J Cell Physiol 2010; 225:233-9. [PMID: 20506163 DOI: 10.1002/jcp.22249] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Human proximal tubule (HK-2) cells are commonly used as cellular models to understand the mechanism by which inflammatory mediators cause renal injury. It has been observed that thrombin stimulates the expression of TGF-beta, extracellular matrix (ECM) proteins and proinflammatory cytokines by HK-2 cells. These in vitro responses correlate well with the pathology of glomerular and tubular diseases observed in acute renal injury. HK-2 cells express PAR-1 and the thrombin activation of this receptor has been reported to up-regulate the TGF-beta-mediated expression of ECM proteins, suggesting a possible pathogenic role for PAR-1 signaling by thrombin in acute renal injury. On the other hand, several recent studies have indicated that activated protein C plays a renoprotective role, thus inhibiting the inflammatory responses and attenuating renal injury, presumably by activating the same cell surface receptor. In this study, we show that HK-2 cells express endothelial protein C receptor (EPCR) and that the occupancy of this receptor by protein C switches the signaling specificity of thrombin so that the activation of PAR-1 by thrombin inhibits the TNF-alpha-mediated synthesis of IL-6 and IL-8 and down-regulates the TGF-beta-mediated expression of ECM proteins. These results suggest a possible protective role for EPCR in acute kidney injury.
Collapse
Affiliation(s)
- Jong-Sup Bae
- Department of Herbal Pharmaceutical Engineering, College of Herbal Bio-Industry, Daegu Haany University, Gyeongsangbuk-do, Republic of Korea.
| | | | | |
Collapse
|
27
|
Protease-activated receptor 2 blocking peptide counteracts endotoxin-induced inflammation and coagulation and ameliorates renal fibrin deposition in a rat model of acute renal failure. Shock 2010; 32:626-32. [PMID: 19333145 DOI: 10.1097/shk.0b013e3181a5359c] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Glomerular and microvascular thrombosis due to the activation of inflammation and coagulation pathway contribute to the occurrence of acute renal failure in sepsis. The protease-activated receptors (PARs) have been shown to play an important role in the interplay between inflammation and coagulation. We hypothesized that PAR-2 blocking would improve glomerular and vascular thrombosis by attenuating inflammation and coagulation, leading to the prevention of acute renal failure, and assessed the effects of the PAR-2 blocking peptide (PAR-2 BP) in a rat model of LPS-induced acute renal failure. Levels of TNF-alpha were significantly expressed 1 h after LPS administration, followed by 1) an increase in levels of tissue factor, factor VIIa, factor Xa, thrombin and plasminogen activator inhibitor 1; 2) unchanged levels of tissue factor pathway inhibitor; and 3) subsequent deposition of fibrin in kidney tissues, which led to the elevation of creatinine and blood urea nitrogen. Time-dependent PAR-2 expression was observed at both the gene and protein levels. Immunoreactivities of PAR-2 and fibrin were observed in the glomerulus and small arteries. Protease-activated receptor blocking peptide suppressed TNF-alpha elevation and attenuated activation of the coagulation, thus leading to a decrease in fibrin formation and its deposition in the glomerulus. However, the levels of creatinine and blood urea nitrogen remained unchanged. These results show that PAR-2 plays a key role in the inflammatory and coagulation process of LPS-induced renal failure; however, PAR-2 inhibition alone does not affect improvement in the renal function.
Collapse
|
28
|
Protease-activated receptors as drug targets in inflammation and pain. Pharmacol Ther 2009; 123:292-309. [PMID: 19481569 DOI: 10.1016/j.pharmthera.2009.05.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Accepted: 05/05/2009] [Indexed: 12/12/2022]
Abstract
Proteases have been shown to signal to cells through the activation of a novel class of receptors coupled to G proteins: the protease-activated receptors (PARs). Those receptors are expressed in a wide range of cells, which ultimately are all involved in mechanisms of inflammation and pain. Numerous studies have considered the role of PARs in cells, organ systems or in vivo, highlighting the fact that PAR activation results in signs of inflammation. A growing body of evidences discussed here suggests that these receptors, and the proteases that activate them, interfere with inflammation and pain processes. Whether a role for PARs has been clearly defined in inflammatory and pain pathologies is discussed in this review. Further, the pros and cons for considering PARs as targets for the development of therapeutic options for the treatment of inflammation and pain are discussed.
Collapse
|
29
|
Lassen MR, Laux V. Emergence of new oral antithrombotics: a critical appraisal of their clinical potential. Vasc Health Risk Manag 2008; 4:1373-86. [PMID: 19337550 PMCID: PMC2663445 DOI: 10.2147/vhrm.s3266] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In Western countries, venous thromboembolism (VTE) is a widespread and serious disorder, with hospital admission rates that appear to be increasing. Current anticoagulant therapies available for the prevention and treatment of VTE have several drawbacks that make them either difficult to manage effectively, due to a need for careful monitoring to control coagulation, or, in the case of parenterally administered agents, inconvenient for long-term use. To address some of these issues, new anticoagulants are in clinical development that can be orally administered and directly target specific factors in the coagulation cascade. This article reviews the rationale behind development of these novel agents and provides a critical appraisal of their clinical potential. In addition, the impact that the introduction of such agents into clinical practice would have is discussed from the patient perspective.
Collapse
Affiliation(s)
- Michael Rud Lassen
- Department of Orthopaedics and Clinical Trial Unit, Nordsjaellands Hospital Hørsholm, University of Copenhagen, Usserød Kongevej 102, Hørsholm, Denmark.
| | | |
Collapse
|
30
|
Materazzi S, Pellerito S, Di Serio C, Paglierani M, Naldini A, Ardinghi C, Carraro F, Geppetti P, Cirino G, Santucci M, Tarantini F, Massi D. Analysis of protease-activated receptor-1 and -2 in human scar formation. J Pathol 2007; 212:440-9. [PMID: 17597495 DOI: 10.1002/path.2197] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Protease-activated receptor (PAR)-1 and PAR-2 are reported to contribute to the fibrotic process in a number of organs, including lung, liver, pancreas, and kidney. The aim of this study was to localize expression and biological activity of PAR-1 and PAR-2 in normal and pathological cutaneous scars. First, we investigated the immunohistochemical expression of PAR-1 and PAR-2 proteins in a series of human normal scars (NS, n = 10), hypertrophic scars (HS, n = 10), and keloids (K, n = 10). Expression of PAR-1 and PAR-2 was observed in all types of scar. Specifically, in HS and K, diffuse PAR-1 and PAR-2 positivity was found in dermal cellular areas composed of myofibroblasts, while no or minor staining was observed in the scattered fibroblasts embedded in abundant extracellular matrix in the context of the more collagenous nodules, irrespective of the type of scar. The hyperplastic epidermis overlying K was also found to be strongly PAR-1 and PAR-2 positive, whilst in most NS and HS the epidermis was faintly to moderately stained. Second, ribonuclease protection assay on paraffin-embedded specimens showed overexpression of PAR-1 and PAR-2 mRNA in K compared to NS and HS. Third, cultured human fibroblasts exposed to TGF-beta1 expressed a myofibroblast phenotype associated with overexpression of PAR-2, while PAR-1 expression was unaffected. Intracellular Ca(2+) mobilization by PAR-2 agonists in myofibroblasts was increased as compared to fibroblasts, whereas the effect of PAR-1 agonists was unchanged. Our in vivo study indicates that PAR-1 and PAR-2 are expressed in cells involved in physiological and pathological scar formation and suggests that in vitro overexpression and exaggerated functional response of PAR-2 may play a role in the function of myofibroblasts in scar evolution from a physiological repair process to a pathological tissue response.
Collapse
Affiliation(s)
- S Materazzi
- Department of Critical Care Medicine and Surgery, University of Florence, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Vesey DA, Kruger WA, Poronnik P, Gobé GC, Johnson DW. Proinflammatory and proliferative responses of human proximal tubule cells to PAR-2 activation. Am J Physiol Renal Physiol 2007; 293:F1441-9. [PMID: 17699557 DOI: 10.1152/ajprenal.00088.2007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Despite the abundant expression of protease-activated receptor (PAR)-2 in the kidney, its relevance to renal physiology is not well understood. A role for this receptor in inflammation and cell proliferation has recently been suggested in nonrenal tissues. The aims of this study were to demonstrate that human proximal tubule cells (PTC) express functional PAR-2 and to investigate whether its activation can mediate proinflammatory and proliferative responses in these cells. Primary human PTC were cultured under serum-free conditions with or without the PAR-2-activating peptide SLIGKV-NH2 (up to 800 microM), a control peptide, VKGILS-NH2 (200 microM), or trypsin (0.01-100 nM). PAR-2 expression (RT-PCR), intracellular Ca2+ mobilization (fura-2 fluorimetry), DNA synthesis (thymidine incorporation), fibronectin production (ELISA, Western blotting), and monocyte chemotactic protein (MCP)-1 secretion (ELISA) were measured. Trypsinogen expression in kidney and PTC cultures was determined by immunohistochemistry and Western blotting. In the kidney PTC were the predominant cell type expressing PAR-2. SLIGKV-NH2, but not VKGILS-NH2, stimulated a rapid concentration-dependent mobilization of intracellular Ca2+ and ERK1/2 phosphorylation and, by 24 h, increases in DNA synthesis, fibronectin secretion, and MCP-1 secretion. These delayed responses appeared to be independent of ERK1/2. Trypsin produced similar rapid but not delayed responses. Trypsinogen was weakly expressed by PTC in the kidney and in culture. In summary, PTC are the main site of PAR-2 expression in the human kidney. In PTC cultures SLIGKV-NH2 initiates proinflammatory and proliferative responses. Trypsinogen expressed within the kidney has the potential to contribute to PAR-2 activation in certain circumstances.
Collapse
Affiliation(s)
- David A Vesey
- 1Centre for Kidney Disease Research, University of Queensland Department of Medicine, Princess Alexandra Hospital, Brisbane, Qld 4102, Australia.
| | | | | | | | | |
Collapse
|
32
|
Veronese F, Rotman S, Smith RN, Pelle TD, Farrell ML, Kawai T, Benedict Cosimi A, Colvin RB. Pathological and clinical correlates of FOXP3+ cells in renal allografts during acute rejection. Am J Transplant 2007; 7:914-22. [PMID: 17286616 DOI: 10.1111/j.1600-6143.2006.01704.x] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The localization and significance of regulatory T cells (Treg) in allograft rejection is of considerable clinical and immunological interest. We analyzed 80 human renal transplant biopsies (including seven donor biopsies) with a double immunohistochemical marker for the Treg transcription factor FOXP3, combined with a second marker for CD4 or CD8. Quantitative FOXP3 cell counts were performed and analyzed for clinical and pathologic correlates. FOXP3(+) cells were present in the interstitium in acute cellular rejection (ACR) type I and II, at a greater density than in acute humoral rejection or CNI toxicity (p < 0.01). Most FOXP3(+) cells were CD4(+) (96%); a minority expressed CD8. FOXP3(+)CD4(+) cells were concentrated in the tubules (p < 0.001), suggesting a selective attraction or generation at that site. Considering only patients with ACR, a higher density of FOXP3(+) correlated with HLA class II match (p = 0.03), but paradoxically with worse graft survival. We conclude that infiltration of FOXP3(+) cells occurs in ACR to a greater degree than in humoral rejection, however, within the ACR group, no beneficial effect on outcome was evident. Tregs concentrate in tubules, probably contributing to FOXP3 mRNA in urine; the significance and pathogenesis of 'Treg tubulitis' remains to be determined.
Collapse
Affiliation(s)
- F Veronese
- Pathology and Surgical Services, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Wang H, Yi T, Zheng Y, He S. Induction of monocyte chemoattractant protein-1 release from A549 cells by agonists of protease-activated receptor-1 and -2. Eur J Cell Biol 2007; 86:233-42. [PMID: 17280738 DOI: 10.1016/j.ejcb.2006.12.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2006] [Revised: 10/27/2006] [Accepted: 12/04/2006] [Indexed: 02/05/2023] Open
Abstract
Hypersecretion of cytokines and serine proteases has been observed in asthma. However, the influence of proteases and protease-activated receptors (PARs) on monocyte chemoattractant protein-1 (MCP-1) release from airway epithelial cells remains largely unknown. In the present study, A549 cells were challenged with agonists of PARs, and levels of MCP-1 released in the supernatant and mRNA expression were examined by ELISA and real time polymerase chain reaction (PCR), respectively. The results show that thrombin, tryptase, elastase and trypsin induced an up to 6.5-, 1.8-, 1.6-, and 3.1-fold increase in MCP-1 release from A549 cells, respectively, following a 16-h incubation period. The protease-induced secretion of MCP-1 can be abolished by specific protease inhibitors. Agonist peptides of PAR-1 and PAR-2 stimulate MCP-1 secretion up to 15- and 12.7-fold, respectively. Real-time PCR showed that MCP-1 mRNA is up-regulated by the serine proteases tested and by agonist peptides of PAR-1 and PAR-2. In conclusion, serine proteases can stimulate MCP-1 release from A549 cells possibly through a PARs-related mechanism, suggesting that they are likely to contribute to MCP-1-related airway inflammatory disorders in man.
Collapse
Affiliation(s)
- Haiyan Wang
- Allergy and Inflammation Research Institute, The Key Immunopharmacology Laboratory of Guangdong Province, Shantou University Medical College, Shantou 515041, China
| | | | | | | |
Collapse
|
34
|
Turpie AGG. Oral, direct factor Xa inhibitors in development for the prevention and treatment of thromboembolic diseases. Arterioscler Thromb Vasc Biol 2007; 27:1238-47. [PMID: 17379841 DOI: 10.1161/atvbaha.107.139402] [Citation(s) in RCA: 172] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Anticoagulants are recommended for the prevention and treatment of a wide variety of thromboembolic events. Although existing anticoagulants are effective, their use is limited by parenteral administration or the requirement for frequent monitoring and subsequent dose adjustment. Therefore, there is an urgent need for novel, oral agents with a predictable anticoagulant action. Because of its key position in the coagulation cascade and its limited roles outside of coagulation, Factor Xa has emerged as an attractive target for novel anticoagulants. As a result, the past decade has witnessed an explosion of research into small-molecule, oral, direct Factor Xa inhibitors, and several are now in clinical development. Rivaroxaban, LY517717, YM150, apixaban, PRT054021, and DU-176b, among others, have shown considerable promise; rivaroxaban is currently furthest ahead in its developmental program, having entered phase III in 3 indications. It is hoped that, before long, these anticoagulants will allow us to enter an era of convenient, oral anticoagulation, without the need for regular monitoring or dose adjustment.
Collapse
|
35
|
McLaughlin JN, Patterson MM, Malik AB. Protease-activated receptor-3 (PAR3) regulates PAR1 signaling by receptor dimerization. Proc Natl Acad Sci U S A 2007; 104:5662-7. [PMID: 17376866 PMCID: PMC1838494 DOI: 10.1073/pnas.0700763104] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Thrombin activates endothelial cell signaling by cleaving the protease-activated receptor-1 (PAR1). However, the function of the apparently nonsignaling receptor PAR3 also expressed in endothelial cells is unknown. We demonstrate here the crucial role of PAR3 in potentiating the responsiveness of PAR1 to thrombin. We tested the hypothesis that PAR1/PAR3 heterodimerization and its effect in modifying G protein selectivity was responsible for PAR3 regulation of PAR1 sensitivity. Using bioluminescent resonance energy transfer-2, we showed that PAR1 had comparable dimerization affinity for PAR3 as for itself. We observed increased Galpha(13) coupling between the PAR1/3 heterodimer compared with the PAR1/1 homodimer. Moreover, knockdown of PAR3 moderated the PAR1-activated increase in endothelial permeability. These results demonstrate a role of PAR3 in allosterically regulating PAR1 signaling governing increased endothelial permeability. Because PAR3 is a critical determinant of PAR1 function, targeting of PAR3 may mitigate the effects of PAR1 in activating endothelial responses such as vascular inflammation.
Collapse
Affiliation(s)
- Joseph N McLaughlin
- Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA.
| | | | | |
Collapse
|
36
|
Hatsukari I, Singh P, Hitosugi N, Messmer D, Valderrama E, Teichberg S, Chaung W, Gross E, Schmidtmayerova H, Singhal PC. DEC-205-mediated internalization of HIV-1 results in the establishment of silent infection in renal tubular cells. J Am Soc Nephrol 2007; 18:780-7. [PMID: 17287423 DOI: 10.1681/asn.2006121307] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
HIV-1 infection of renal cells has been proposed to play a role in HIV-1-associated nephropathy. Renal biopsy data further suggest that renal tubular cells may serve as reservoir for HIV-1. The mechanism by which HIV-1 enters these cells has not been identified. Renal tubular cells do not express any of the known HIV-1 receptors, and our results confirmed lack of the expression of CD4, CCR5, CXCR4, DC-SIGN, or mannose receptors in tubular cells. The aim of this study, therefore, was to determine the mechanism that enables viral entry into renal tubular cells. An in vitro model was used to study the HIV-1 infection of human kidney tubular (HK2) cells and to identify the receptor that enables the virus to enter these cells. Results of these studies demonstrate that the C-type lectin DEC-205 acts as an HIV-1 receptor in HK2 cells. Interaction of HIV-1 with DEC-205 results in the internalization of the virus and establishment of a nonproductive infection. HIV-1-specific strong-stop DNA is detected in the infected HK2 cells for at least 7 d, and the virus can be transmitted in trans to sensitive target cells. HIV-1 entry is blocked by pretreatment with specific anti-DEC-205 antibody. Moreover, expression of DEC-205 in cells that lack the DEC-205 receptors renders them susceptible to HIV-1 infection. These findings suggest that DEC-205 acts as an HIV-1 receptor that mediates internalization of the virus into renal tubular cells, from which the virus can be rescued and disseminated by encountering immune cells.
Collapse
Affiliation(s)
- Ikusuke Hatsukari
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Vesey DA, Hooper JD, Gobe GC, Johnson DW. Potential physiological and pathophysiological roles for protease-activated receptor-2 in the kidney (Review Article). Nephrology (Carlton) 2007; 12:36-43. [PMID: 17295659 DOI: 10.1111/j.1440-1797.2006.00746.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The protease-activated receptor-2 (PAR-2), the second of four members of a unique subfamily of G-protein coupled receptors, is abundantly expressed in the kidney. In a similar manner to other PAR cleavage of its extracellular N-terminus exposes a tethered ligand, SLIGKV in humans, which acts as an intramolecular ligand to activate itself. In the kidney, PAR-2 expression has been variably reported in collecting duct cells, mesangial cells, interstitial fibroblasts, vascular endothelial cells, vascular smooth muscle cells and proximal tubular cells. Despite this renal expression data, the function of PAR-2 in the kidney remains unknown. More than 15 different mammalian serine proteases have been shown to activate PAR-2 in an in vitro setting, but it is still unclear which of these are physiologically relevant activators of PAR-2 in specific tissues. Their identification could provide novel therapeutic targets. PAR-2 activates a number of down-stream signalling molecules that include protein kinase C, extracellular signal regulated kinase and nuclear factor kappa-B. Proteases that can activate PAR-2 are generated and released from cells during injury, inflammation and malignancy and can thus signal to cells under these conditions. Potential physiological and pathophysiological roles for PAR-2 in the kidney include the regulation of inflammation, blood flow, and ion transport and tissue protection, repair and fibrosis. In this review the potential roles of PAR-2 in the kidney are highlighted and discussed.
Collapse
Affiliation(s)
- David A Vesey
- Centre for Kidney Disease Research, University of Queensland Department of Medicine, Princess Alexandra Hospital, Woolloongabba, Brisbane, Queensland, Australia.
| | | | | | | |
Collapse
|