1
|
Tisch LJ, Bartone RD, Antoniak S, Bonner JC. Protease-activated receptor-2 (PAR2) mutation attenuates airway fibrosis in mice during the exacerbation of house dust mite‑induced allergic lung disease by multi‑walled carbon nanotubes. Respir Res 2025; 26:90. [PMID: 40057754 PMCID: PMC11889904 DOI: 10.1186/s12931-025-03168-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/24/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Pulmonary exposure to multi-walled carbon nanotubes (MWCNTs) induces potent pro-inflammatory and pro-fibrotic responses in mouse models of allergic lung disease. We recently reported that MWCNTs exacerbated components of house dust mite (HDM)-induced allergic lung disease, including eosinophilic inflammation, mucous cell metaplasia and airway fibrosis. Protease-activated receptor 2 (PAR2) plays a significant role in the development of various respiratory diseases, including asthma and pulmonary fibrosis. However, studies investigating the function of PAR2 in allergic lung disease have produced variable results. To further define the role of PAR2 in pulmonary pathology, we investigated the effects of MWCNTs on HDM-induced allergic lung disease in PAR2-mutant mice. METHODS The PAR2-mutant mice used were previously generated by replacing a 1.8-kb region of the PAR2 coding sequence with a neomycin resistance gene, which did not entirely delete the gene. Wild-type (WT) male C57BL/6J mice and PAR2-mutant male mice were exposed to a vehicle solution, MWCNTs, HDM extract, or both via oropharyngeal aspiration six times over 3 weeks. Bronchoalveolar lavage fluid (BALF) was collected to measure changes in inflammatory cells, total protein, and lactate dehydrogenase (LDH). Lung protein and mRNA were assayed for pro-inflammatory and profibrotic mediators, and formalin-fixed lung sections were evaluated for histopathology. RESULTS In WT and PAR2-mutant mice, co-exposure to MWCNTs and HDM extract significantly increased eosinophilic lung inflammation, mucous cell metaplasia, increased BALF cellularity, BALF total protein, and LDH levels. These results were not significantly different between genotypes. Additionally, MWCNTs and HDM extract co-exposure significantly increased airway fibrosis in WT and PAR2-mutant mice, characterized by increased airway collagen deposition and Col1a1 mRNA expression. Quantitative morphometry revealed a significant decrease in airway fibrosis in PAR2-mutant mice compared to WT mice, accompanied by reduced Col1a1 mRNA as detected by PCR. Despite this reduction, the pro-fibrotic mediator arginase 1 (Arg-1) protein and mRNA levels were significantly upregulated in PAR2-mutant mice. CONCLUSION Our study demonstrates that PAR2 mediates airway fibrosis but does not influence eosinophilic lung inflammation or mucous cell metaplasia caused by co-exposure to MWCNTs and HDM allergen.
Collapse
Affiliation(s)
- Logan J Tisch
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Ryan D Bartone
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Silvio Antoniak
- UNC Blood Research Center, Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - James C Bonner
- Toxicology Program, Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
2
|
Lundgren JG, Flynn MG, List K. GPI-anchored serine proteases: essential roles in development, homeostasis, and disease. Biol Chem 2025; 406:1-28. [PMID: 40094301 DOI: 10.1515/hsz-2024-0135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 02/23/2025] [Indexed: 03/19/2025]
Abstract
The glycosylphosphatidylinositol (GPI)-anchored serine proteases, prostasin and testisin, have essential roles in diverse physiological functions including development, reproduction, homeostasis and barrier function of epithelia, angiogenesis, coagulation, and fibrinolysis. Important functions in pathological conditions such as cancer, kidney disease and cardiovascular disease have also been reported. In this review, we summarize current knowledge of the cellular and in vivo roles of prostasin and testisin in physiology and pathophysiology and explore the underlying molecular mechanisms. We discuss how new insights of their role in cancer and cardiovascular disease may facilitate translation into clinical settings in the future.
Collapse
Affiliation(s)
- Joseph G Lundgren
- Department of Pharmacology, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, Wayne State University, Detroit, MI 48201, USA
| | - Michael G Flynn
- Department of Pharmacology, Wayne State University, Detroit, MI 48201, USA
| | - Karin List
- Department of Pharmacology, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
3
|
Maity J, Pal P, Pal R, Mukhopadhyay PK. Co-administration of L-Ascorbic Acid and α-Tocopherol Alleviates Arsenic-Induced Immunotoxicities in the Thymus and Spleen by Dwindling Oxidative Stress-Induced Inflammation. Biol Trace Elem Res 2024; 202:2199-2227. [PMID: 37704839 DOI: 10.1007/s12011-023-03841-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/01/2023] [Indexed: 09/15/2023]
Abstract
Herein, we investigated whether L-ascorbic acid (L-AA) and α-tocopherol (α-T) co-administration has the potential to alleviate arsenic-induced immunotoxicities in the thymus, spleen, and circulating leukocytes. Forty-eight adult male Wistar rats were randomly divided into four groups before the treatment: group I (control); group II (sodium arsenite, 3 mg/kg/day/rat); group III (sodium arsenite + L-AA (200 mg/kg/day/rat) and α-T (400 mg/kg/day/rat)); group IV (L-AA and α-T). The result showed that sodium arsenite exposure (consecutive 30 days) caused weight reduction, structural alterations in the thymus and spleen, accompanied by a decrease in thymocyte and splenocyte count. Decreased superoxide dismutase and catalase activity, increased malondialdehyde and protein-carbonyl content, reduced Nrf2 and Bcl2 expression, and increased p-ERK, NF-kβ, Bax, and cleaved-caspase-3 expression were also observed in the thymus and spleen of arsenic-exposed rats. Enhanced plasma ACTH and corticosterone, ROS-induced apoptosis of lymphocytes were also observed. L-AA and α-T co-administration has the potential to abrogate the deleterious impact of arsenic on the thymus, spleen, and circulating lymphocytes. Whole transcriptome analysis of leukocytes revealed that arsenic treatment augmented the expression of Itga4, Itgam, and MMP9 genes, which might help in transient migration of the leukocytes through the endothelial cell layer. Co-administration with L-AA and α-T maintained Itga4, Itgam, and MMP9 gene expression within leukocytes at a lower level.
Collapse
Affiliation(s)
- Jeet Maity
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | - Priyankar Pal
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | - Ranjana Pal
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata, 700073, India
| | | |
Collapse
|
4
|
Berger M, Guiraud L, Dumas A, Sagnat D, Payros G, Rolland C, Vergnolle N, Deraison C, Cenac N, Racaud-Sultan C. Prenatal stress induces changes in PAR2- and M3-dependent regulation of colon primitive cells. Am J Physiol Gastrointest Liver Physiol 2022; 323:G609-G626. [PMID: 36283083 PMCID: PMC9722261 DOI: 10.1152/ajpgi.00061.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Prenatal stress is associated with a high risk of developing adult intestinal pathologies, such as irritable bowel syndrome, chronic inflammation, and cancer. Although epithelial stem cells and progenitors have been implicated in intestinal pathophysiology, how prenatal stress could impact their functions is still unknown. We have investigated the proliferative and differentiation capacities of primitive cells using epithelial crypts isolated from colons of adult male and female mice whose mothers have been stressed during late gestation. Our results show that stem cell/progenitor proliferation and differentiation in vitro are negatively impacted by prenatal stress in male progeny. This is promoted by a reinforcement of the negative proliferative/differentiation control by the protease-activated receptor 2 (PAR2) and the muscarinic receptor 3 (M3), two G protein-coupled receptors present in the crypt. Conversely, prenatal stress does not change in vitro proliferation of colon primitive cells in female progeny. Importantly, this maintenance is associated with a functional switch in the M3 negative control of colonoid growth, becoming proliferative after prenatal stress. In addition, the proliferative role of PAR2 specific to females is maintained under prenatal stress, even though PAR2-targeted stress signals Dusp6 and activated GSK3β are increased, reaching the levels of males. An epithelial serine protease could play a critical role in the activation of the survival kinase GSK3β in colonoids from prenatally stressed female progeny. Altogether, our results show that following prenatal stress, colon primitive cells cope with stress through sexually dimorphic mechanisms that could pave the way to dysregulated crypt regeneration and intestinal pathologies.NEW & NOTEWORTHY Primitive cells isolated from mouse colon following prenatal stress and exposed to additional stress conditions such as in vitro culture, present sexually dimorphic mechanisms based on PAR2- and M3-dependent regulation of proliferation and differentiation. Whereas prenatal stress reinforces the physiological negative control exerted by PAR2 and M3 in crypts from males, in females, it induces a switch in M3- and PAR2-dependent regulation leading to a resistant and proliferative phenotype of progenitor.
Collapse
Affiliation(s)
- Mathieu Berger
- 1Institut de Recherche en Santé Digestive, INSERM U1220, Institut
National de Recherche pour l’Agriculture, l’Alimentation et
l’Environnement, Ecole Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Laura Guiraud
- 1Institut de Recherche en Santé Digestive, INSERM U1220, Institut
National de Recherche pour l’Agriculture, l’Alimentation et
l’Environnement, Ecole Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Alexia Dumas
- 1Institut de Recherche en Santé Digestive, INSERM U1220, Institut
National de Recherche pour l’Agriculture, l’Alimentation et
l’Environnement, Ecole Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - David Sagnat
- 1Institut de Recherche en Santé Digestive, INSERM U1220, Institut
National de Recherche pour l’Agriculture, l’Alimentation et
l’Environnement, Ecole Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Gaëlle Payros
- 1Institut de Recherche en Santé Digestive, INSERM U1220, Institut
National de Recherche pour l’Agriculture, l’Alimentation et
l’Environnement, Ecole Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Corinne Rolland
- 1Institut de Recherche en Santé Digestive, INSERM U1220, Institut
National de Recherche pour l’Agriculture, l’Alimentation et
l’Environnement, Ecole Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Nathalie Vergnolle
- 1Institut de Recherche en Santé Digestive, INSERM U1220, Institut
National de Recherche pour l’Agriculture, l’Alimentation et
l’Environnement, Ecole Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France,2Department of Physiology and Pharmacology, Cumming School of
Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Céline Deraison
- 1Institut de Recherche en Santé Digestive, INSERM U1220, Institut
National de Recherche pour l’Agriculture, l’Alimentation et
l’Environnement, Ecole Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Nicolas Cenac
- 1Institut de Recherche en Santé Digestive, INSERM U1220, Institut
National de Recherche pour l’Agriculture, l’Alimentation et
l’Environnement, Ecole Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Claire Racaud-Sultan
- 1Institut de Recherche en Santé Digestive, INSERM U1220, Institut
National de Recherche pour l’Agriculture, l’Alimentation et
l’Environnement, Ecole Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| |
Collapse
|
5
|
Jia J, Zeng M, Zhu D, Jiao X, Zhang B, Yang R, Feng W, Zheng X. An Amide Alkaloid Isolated from Ephedra sinica Ameliorates OVA-Induced Allergic Asthma by Inhibiting Mast Cell Activation and Dendritic Cell Maturation. Int J Mol Sci 2022; 23:13541. [PMID: 36362328 PMCID: PMC9655655 DOI: 10.3390/ijms232113541] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/30/2022] [Accepted: 11/01/2022] [Indexed: 09/14/2023] Open
Abstract
Asthma, which is a chronic inflammatory disease of the airways, is usually caused by allergens in which various structures and immune cells are involved. Ephedra sinica, the most commonly used Chinese medicine, has significant clinical effects on asthma, but its components are complex and the mechanism of action has not been fully elucidated. Among its components, we identified an amide alkaloid (EB-A) and investigated its anti-asthmatic activity and the underlying mechanisms. In this study, we replicated an OVA-sensitized/challenged allergic asthma mouse model, and divided the mice into a model (OVA) group, positive drug (Y, 0.5 mg/kg/day) group, and EB-A treatment with low (Low, 10 mg/kg/day) and high dose (High, 20 mg/kg/day) groups. Asthma-related features were analyzed through the airway hyperresponsiveness (AHR), cough and wheeze indexes, allergen-specific IgE, prostaglandin D2 (PDG2), and lung histology in mice. The levels of apoptosis and reactive oxygen species (ROS) in the primary lung cells, cytokines in the serum and broncho-alveolar lavage fluid (BALF), and proteinase-activated receptor-2 (PAR2) pathway activation in the lung tissue were measured to evaluate the inflammatory injury and lung epithelial barrier damage in the mice. Dendritic cell (DC) maturation and mast cell (MC) activation were verified in vitro and in vivo. Furthermore, the effect of a PAR2 activation in lung epithelial cells on the maturation of DCs was evaluated by the co-culture system of (human bronchial epithelial cell lines) 16HBE and bone marrow-derived dendritic cells (BMDCs). The results showed that EB-A inhibited the typical asthmatic phenotypes, as well as lung injury and inflammation, MC activation and degranulation, and DC maturation in the OVA-sensitized/challenged BALB/c mice. In addition, EB-A inhibited the expression of PAR2 in the lung epithelial cells and significantly interfered with the maturation of DCs after inhibiting PAR2. Taken together, our study firstly demonstrated that EB-A could ameliorate OVA-induced allergic asthma by inhibiting MC activation and DC maturation, and the molecular mechanism of EB-A's anti-asthmatic activity might be mediated by inhibiting PAR2. Our data provide a molecular justification for the use of EB-A in the treatment of allergic asthma.
Collapse
Affiliation(s)
- Jufang Jia
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Mengnan Zeng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Denghui Zhu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Xinmian Jiao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Beibei Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Ruolan Yang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Weisheng Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| | - Xiaoke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
- The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Zhengzhou 450046, China
| |
Collapse
|
6
|
Pagel CN, Kularathna PK, Sanaei R, Young ND, Hooper JD, Mackie EJ. Protease-activated receptor-2 dependent and independent responses of bone cells to prostate cancer cell secretory products. Prostate 2022; 82:723-739. [PMID: 35167724 DOI: 10.1002/pros.24316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 08/25/2021] [Accepted: 09/27/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Metastatic prostate cancer lesions in the skeleton are frequently characterized by excessive formation of bone. Prostate cancer cells secrete factors, including serine proteases, that are capable of influencing the behavior of surrounding cells. Some of these proteases activate protease-activated receptor-2 (PAR2 ), which is expressed by osteoblasts (bone-forming cells) and precursors of osteoclasts (bone-resorbing cells). The aim of the current study was to investigate a possible role for PAR2 in regulating the behavior of bone cells exposed to metastatic prostate cancer cells. METHODS The effect of medium conditioned by the PC3, DU145, and MDA-PCa-2b prostate cancer cell lines was investigated in assays of bone cell function using cells isolated from wildtype and PAR2 -null mice. Osteoclast differentiation was assessed by counting tartrate-resistant acid phosphatase-positive multinucleate cells in bone marrow cultured in osteoclastogenic medium. Osteoblasts were isolated from calvariae of neonatal mice, and BrdU incorporation was used to assess their proliferation. Assays of alkaline phosphatase activity and quantitative PCR analysis of osteoblastic gene expression were used to assess osteoblast differentiation. Responses of osteoblasts to medium conditioned by MDA-PCa-2b cells were analyzed by RNAseq. RESULTS Conditioned medium (CM) from all three cell lines inhibited osteoclast differentiation independently of PAR2 . Media from PC3 and DU145 cells had no effect on assays of osteoblast function. Medium conditioned by MDA-PCa-2b cells stimulated BrdU incorporation in both wildtype and PAR2 -null osteoblasts but increased alkaline phosphatase activity and Runx2 and Col1a1 expression in wildtype but not PAR2 -null cells. Functional enrichment analysis of RNAseq data identified enrichment of multiple gene ontology terms associated with lysosomal function in both wildtype and PAR2 -null cells in response to MDA-PCa-2b-CM. Analysis of individual genes identified osteogenesis-associated genes that were either upregulated by MDA-PCa-2b-CM selectively in wildtype cells or downregulated selectively in PAR2 -null cells. CONCLUSIONS Factors secreted by prostate cancer cells influence bone cell behavior through both PAR2 -dependent and -independent mechanisms. Both PAR2 -independent suppression of osteoclast differentiation and PAR2 -dependent stimulation of osteogenesis are likely to determine the nature of prostate cancer metastases in bone.
Collapse
Affiliation(s)
- Charles N Pagel
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Pamu K Kularathna
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Reza Sanaei
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Neil D Young
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - John D Hooper
- Mater Research Institute, Translational Research Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Eleanor J Mackie
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
7
|
Sanaei R, Kularathna P, Taghavi N, Hooper J, Pagel C, Mackie E. Protease-activated receptor-2 promotes osteogenesis in skeletal mesenchymal stem cells at the expense of adipogenesis: Involvement of interleukin-6. Bone Rep 2021; 15:101113. [PMID: 34430676 PMCID: PMC8365448 DOI: 10.1016/j.bonr.2021.101113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 10/27/2022] Open
Abstract
Bone marrow mesenchymal stem cells (MSCs) give rise to osteoblasts and adipocytes, with an inverse relationship between the two. The MSCs from protease-activated receptor-2 knockout (PAR2 KO) mice have a reduced capacity to generate osteoblasts. Here we describe the observation that PAR2 KO osteoblastic cultures generate more adipocytes than wildtype (WT) cultures. Osteoblasts from PAR2 KO mice expressed lower levels of osteoblastic genes (Runx2, Col1a1 and Bglap), and higher levels of the adipocytic gene Pparg than WT osteoblasts. Bone marrow stromal cells from PAR2 KO mice generated fewer osteoblastic colonies (assessed by staining for alkaline phosphatase activity and mineral deposition) and more adipocytic (Oil Red-O positive) colonies than cultures from WT mice. Similarly, cultures of the bone marrow stromal cell line (Kusa 4b10) in which PAR2 was knocked down (F2rl1 KD), were less osteoblastic and more adipocytic than vector control cells. Putative regulators of PAR2-mediated osteogenesis and suppression of adipogenesis were identified in an RNA-sequencing (RNA-seq) investigation; these include C1qtnf3, Gpr35, Grem1, Snorc and Tcea3, which were more highly expressed, and Cnr1, Enpep, Hmgn5, Il6 and Ramp3 which were expressed at lower levels, in control than in F2rl1 KD cells. Interleukin-6 (IL-6) levels were higher in medium harvested from F2rl1 KD cells than from control cells, and a neutralising anti-IL-6 antibody reduced the number of adipocytes in F2rl1 KD cultures to that of control cultures. Thus, PAR2 appears to be a mediator of the reciprocal relationship between osteogenesis and adipogenesis, with IL-6 having a regulatory role in these PAR2-mediated effects.
Collapse
Affiliation(s)
- R. Sanaei
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - P.K. Kularathna
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - N. Taghavi
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - J.D. Hooper
- Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - C.N. Pagel
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - E.J. Mackie
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
8
|
Xue M, Lin H, Liang HPH, McKelvey K, Zhao R, March L, Jackson C. Deficiency of protease-activated receptor (PAR) 1 and PAR2 exacerbates collagen-induced arthritis in mice via differing mechanisms. Rheumatology (Oxford) 2021; 60:2990-3003. [PMID: 33823532 DOI: 10.1093/rheumatology/keaa701] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 09/17/2020] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVES Protease-activated receptor (PAR) 1 and PAR2 have been implicated in RA, however their exact role is unclear. Here, we detailed the mechanistic impact of these receptors on the onset and development of inflammatory arthritis in murine CIA and antigen-induced arthritis (AIA) models. METHODS CIA or AIA was induced in PAR1 or PAR2 gene knockout (KO) and matched wild type mice. The onset and development of arthritis was monitored clinically and histologically. Immune cells, cytokines and MMPs were detected by ELISA, zymography, flow cytometry, western blot or immunohistochemistry. RESULTS In CIA, PAR1KO and PAR2KO exacerbated arthritis, in opposition to their effects in AIA. These deficient mice had high plasma levels of IL-17, IFN-γ, TGF-β1 and MMP-13, and lower levels of TNF-α; T cells and B cells were higher in both KO spleen and thymus, and myeloid-derived suppressor cells were lower only in PAR1KO spleen, when compared with wild type cells. Th1, Th2 and Th17 cells were lower in PAR1KO spleens cells, whereas Th1 and Th2 cells were lower and Th17 cells higher in both KO thymus cells, when compared with wild type cells. PAR1KO synovial fibroblasts proliferated faster and produced the most abundant MMP-9 amongst three type cells in the control, lipopolysaccharides or TNF stimulated conditions. CONCLUSION This is the first study demonstrated that deficiency of PAR1 or PAR2 aggravates inflammatory arthritis in CIA. Furthermore, the protective functions of PAR1 and PAR2 in CIA likely occur via differing mechanisms involving immune cell differentiation and cytokines/MMPs.
Collapse
Affiliation(s)
- Meilang Xue
- Sutton Arthritis Research Laboratory, Institute of Bone and Joint Research, Sydney, NSW, Australia
| | - Haiyan Lin
- Sutton Arthritis Research Laboratory, Institute of Bone and Joint Research, Sydney, NSW, Australia
| | - Hai Po Helena Liang
- Sutton Arthritis Research Laboratory, Institute of Bone and Joint Research, Sydney, NSW, Australia
| | - Kelly McKelvey
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Ruilong Zhao
- Sutton Arthritis Research Laboratory, Institute of Bone and Joint Research, Sydney, NSW, Australia
| | - Lyn March
- Sutton Arthritis Research Laboratory, Institute of Bone and Joint Research, Sydney, NSW, Australia
| | - Christopher Jackson
- Sutton Arthritis Research Laboratory, Institute of Bone and Joint Research, Sydney, NSW, Australia
| |
Collapse
|
9
|
The development of proteinase-activated receptor-2 modulators and the challenges involved. Biochem Soc Trans 2021; 48:2525-2537. [PMID: 33242065 PMCID: PMC7752072 DOI: 10.1042/bst20200191] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/13/2020] [Accepted: 11/02/2020] [Indexed: 11/30/2022]
Abstract
Protease-activated receptor-2 (PAR2) has been extensively studied since its discovery in the mid-1990. Despite the advances in understanding PAR2 pharmacology, it has taken almost 25 years for the first inhibitor to reach clinical trials, and so far, no PAR2 antagonist has been approved for human use. Research has employed classical approaches to develop a wide array of PAR2 agonists and antagonists, consisting of peptides, peptoids and antibodies to name a few, with a surge in patent applications over this period. Recent breakthroughs in PAR2 structure determination has provided a unique insight into proposed PAR2 ligand binding sites. Publication of the first crystal structures of PAR2 resolved in complex with two novel non-peptide small molecule antagonists (AZ8838 and AZ3451) revealed two distinct binding pockets, originally presumed to be allosteric sites, with a PAR2 antibody (Fab3949) used to block tethered ligand engagement with the peptide-binding domain of the receptor. Further studies have proposed orthosteric site occupancy for AZ8838 as a competitive antagonist. One company has taken the first PAR2 antibody (MEDI0618) into phase I clinical trial (NCT04198558). While this first-in-human trial is at the early stages of the assessment of safety, other research into the structural characterisation of PAR2 is still ongoing in an attempt to identify new ways to target receptor activity. This review will focus on the development of novel PAR2 modulators developed to date, with an emphasis placed upon the advances made in the pharmacological targeting of PAR2 activity as a strategy to limit chronic inflammatory disease.
Collapse
|
10
|
Bang E, Kim DH, Chung HY. Protease-activated receptor 2 induces ROS-mediated inflammation through Akt-mediated NF-κB and FoxO6 modulation during skin photoaging. Redox Biol 2021; 44:102022. [PMID: 34082382 PMCID: PMC8182111 DOI: 10.1016/j.redox.2021.102022] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 12/15/2022] Open
Abstract
Long-term exposure to ultraviolet irradiation to skin leads to deleterious intracellular effects, including reactive oxygen species (ROS) production and inflammatory responses, causing accelerated skin aging. Previous studies have demonstrated that increased expression and activation of protease-activated receptor 2 (PAR2) and Akt is observed in keratinocyte proliferation, suggesting their potential regulatory role in skin photoaging. However, the specific underlying molecular mechanism of PAR2 and the Akt/NF-κB/FoxO6-mediated signaling pathway is not clearly defined. In this study, we first used the UVB-irradiated photoaged skin of hairless mice and observed an increase in PAR2 and Gαq expression and PI3-kinase/Akt, NF-κB, and suppressed FoxO6. Consequently, increased levels of proinflammatory cytokines and decreased levels of antioxidant MnSOD was observed. Next, to investigate PAR2-specific roles in inflammation and oxidative stress, we used photoaged hairless mice topically applied with PAR2 antagonist GB83 and photoaged PAR2 knockout mice. PAR2 inhibition and deletion significantly suppressed inflammatory and oxidative stress levels, which were associated with decreased IL-6 and IL-1β levels and increased MnSOD levels, respectively. Furthermore, NF-κB phosphorylation and decreased FoxO6 was reduced by PAR2 inhibition and deletion in vivo. To confirm the in vivo results, we conducted PAR2 knockdown and overexpression in UVB-irradiated HaCaT cells. In PAR2 knockdown cells by si-PAR2 treatment, it suppressed Akt/NF-κB and increased FoxO6, whereas PAR2 overexpression reversed these effects and subsequently modulated proinflammatory target genes. Collectively, our data define that PAR2 induces oxidative stress and inflammation through Akt-mediated phosphorylation of NF-κB (Ser536) and FoxO6 (Ser184), which could be a critical upstream regulatory mechanism in ROS-mediated inflammatory response.
Collapse
Affiliation(s)
- EunJin Bang
- Department of Pharmacy, College of Pharmacy, Pusan National University, Gumjung-gu, Busan, 46241, South Korea
| | - Dae Hyun Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, Gumjung-gu, Busan, 46241, South Korea.
| | - Hae Young Chung
- Department of Pharmacy, College of Pharmacy, Pusan National University, Gumjung-gu, Busan, 46241, South Korea.
| |
Collapse
|
11
|
Bhatt LK, Selokar I, Raut D, Hussain T. Novel Targets for Hypertension Drug Discovery. Curr Hypertens Rep 2021; 23:19. [PMID: 33783647 DOI: 10.1007/s11906-021-01137-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2021] [Indexed: 01/21/2023]
Abstract
PURPOSE OF REVIEW Despite the availability of various medications and prescribing combination therapies, uncontrolled blood pressure and resistance are observed in more than 40% of patients. The purpose of this review is to discuss emerging novel approaches for the treatment of hypertension and propose future research and clinical directions. RECENT FINDINGS Hypertension is a common disease of the cardiovascular system which may arise solely or as a comorbidity of other disorders. It is a crucial risk factor for cardiovascular diseases such as coronary artery disease, myocardial infarction, congestive heart failure, renal failure, and stroke. The results from current literature regarding the novel approaches showed several targets that could be explored as potential therapeutic options. These include toll-like receptor 4, a critical regulator of angiotensin II-induced hypertension; protease-activated receptor 2, which promotes collagen deposition and inflammatory responses; chemerin, which causes metabolic and obesity-associated hypertension; apelin receptor; transient receptor potential melastatin; urotensin-II; and Tie2 receptor. This review discusses various targets and pathways that could be emerging pharmacological therapies for hypertension.
Collapse
Affiliation(s)
- Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's DR. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India.
| | - Ishant Selokar
- Department of Pharmacology, SVKM's DR. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Dezaree Raut
- Department of Pharmacology, SVKM's DR. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Tahir Hussain
- College of Pharmacy, University of Houston, Houston, TX, USA
| |
Collapse
|
12
|
Experimentally induced spine osteoarthritis in rats leads to neurogenic inflammation within neurosegmentally linked myotomes. Exp Gerontol 2021; 149:111311. [PMID: 33744392 DOI: 10.1016/j.exger.2021.111311] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 09/13/2020] [Accepted: 03/04/2021] [Indexed: 12/19/2022]
Abstract
Naturally occurring spine osteoarthritis is clinically associated with the manifestation of chronic inflammatory muscle (myofascial) disease. The purpose of this study was to investigate the causal association between experimentally induced spine osteoarthritis and neurogenic inflammatory responses within neurosegmentally linked myotomes. Wistar Kyoto rats were randomly assigned to spine facet compression surgery (L4-L6) or sham surgery. Animals exposed to facet compression surgery demonstrated radiographic signs of facet-osteoarthritis (L4-L6 spinal levels) and sensory changes (allodynia, thermal hyperalgesia) at 7, 14 and 21 days post-intervention, consistent with the induction of central sensitization; no radiologic or sensory changes were observed after sham surgery. Increased levels of proinflammatory biomarkers including substance P (SP), calcitonin gene related peptide (CGRP), protease-activated receptor-2 (PAR2) and calcium/calmodulin dependent protein kinase II (CaMKII) were observed post-surgery within neurosegmentally-linked rectus femoris (L2-L5) muscle when compared to the non-segmentally linked biceps brachii (C4-C7) muscle; no differences were observed between muscles in the sham surgery group. These findings offer novel insight into the potential role of spine osteoarthritis and neurogenic inflammatory mechanisms in the pathophysiology of chronic inflammatory muscle (myofascial) disease.
Collapse
|
13
|
Ichikawa H, Shimada M, Narita M, Narita I, Kimura Y, Tanaka M, Osanai T, Okumura K, Tomita H. Rivaroxaban, a Direct Factor Xa Inhibitor, Ameliorates Hypertensive Renal Damage Through Inhibition of the Inflammatory Response Mediated by Protease-Activated Receptor Pathway. J Am Heart Assoc 2020; 8:e012195. [PMID: 30957622 PMCID: PMC6507187 DOI: 10.1161/jaha.119.012195] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Background An enhanced renin‐angiotensin system causes hypertensive renal damage. Factor Xa not only functions in the coagulation cascade but also activates intracellular signaling through protease‐activated receptors (PAR). We investigated the effects of rivaroxaban, a factor Xa inhibitor, on hypertensive renal damage in hypertensive mice overexpressing renin (Ren‐TG). Methods and Results The 12‐ to 16‐week‐old Ren‐TG and wild‐type mice were orally administered with or without 6 or 12 mg/kg of rivaroxaban for 1 or 4 months. Plasma factor Xa was significantly increased in the Ren‐TG compared with the wild‐type mice and was reduced by 12 mg/kg of rivaroxaban (P<0.05). Urinary albumin excretion (UAE) was higher in the nontreated 8‐month‐old Ren‐TG than in the wild‐type mice (69.6±29 versus 20.1±8.2 μg/day; P<0.01). Treatment with 12 mg/kg of rivaroxaban for 4 months decreased the UAE to 38.1±13.2 μg/day (P<0.01). Moreover, rivaroxaban treatment attenuated histologic changes of glomerular hypertrophy, mesangial matrix expansion, effacement of the podocyte foot process, and thickened glomerular basement membrane in the Ren‐TG. The renal expression of PAR‐2 was increased in the Ren‐TG, but was inhibited with rivaroxaban treatment. In vitro study using the human podocytes showed that the expressions of PAR‐2 and inflammatory genes and nuclear factor–‐κB activation were induced by angiotensin II stimulation, but were inhibited by rivaroxaban. PAR‐2 knockdown by small interfering RNA also attenuated the PAR‐2‐related inflammatory gene expressions. Conclusions These findings indicate that rivaroxaban exerts protective effects against angiotensin II–induced renal damage, partly through inhibition of the PAR‐2 signaling‐mediated inflammatory response.
Collapse
Affiliation(s)
- Hiroaki Ichikawa
- 1 Department of Cardiology and Nephrology Hirosaki University Graduate School of Medicine Hirosaki Japan
| | - Michiko Shimada
- 1 Department of Cardiology and Nephrology Hirosaki University Graduate School of Medicine Hirosaki Japan
| | - Masato Narita
- 1 Department of Cardiology and Nephrology Hirosaki University Graduate School of Medicine Hirosaki Japan
| | - Ikuyo Narita
- 1 Department of Cardiology and Nephrology Hirosaki University Graduate School of Medicine Hirosaki Japan
| | - Yoshihiro Kimura
- 1 Department of Cardiology and Nephrology Hirosaki University Graduate School of Medicine Hirosaki Japan
| | - Makoto Tanaka
- 2 Department of Stroke and Cerebrovascular Medicine Hirosaki University Graduate School of Medicine Hirosaki Japan
| | - Tomohiro Osanai
- 3 Department of Nursing Science Hirosaki University Graduate School of Health Sciences Hirosaki Japan
| | - Ken Okumura
- 4 Division of Cardiology Saiseikai Kumamoto Hospital Kumamoto Japan
| | - Hirofumi Tomita
- 1 Department of Cardiology and Nephrology Hirosaki University Graduate School of Medicine Hirosaki Japan.,2 Department of Stroke and Cerebrovascular Medicine Hirosaki University Graduate School of Medicine Hirosaki Japan
| |
Collapse
|
14
|
Kawaguchi M, Yamamoto K, Kataoka H, Izumi A, Yamashita F, Kiwaki T, Nishida T, Camerer E, Fukushima T. Protease-activated receptor-2 accelerates intestinal tumor formation through activation of nuclear factor-κB signaling and tumor angiogenesis in Apc Min/+ mice. Cancer Sci 2020; 111:1193-1202. [PMID: 31997435 PMCID: PMC7156842 DOI: 10.1111/cas.14335] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 01/19/2020] [Accepted: 01/23/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatocyte growth factor activator inhibitor‐1 (HAI‐1), encoded by the SPINT1 gene, is a membrane‐bound protease inhibitor expressed on the surface of epithelial cells. Hepatocyte growth factor activator inhibitor‐1 regulates type II transmembrane serine proteases that activate protease‐activated receptor‐2 (PAR‐2). We previously reported that deletion of Spint1 in ApcMin/+ mice resulted in accelerated formation of intestinal tumors, possibly through enhanced nuclear factor‐κB signaling. In this study, we examined the role of PAR‐2 in accelerating tumor formation in the ApcMin/+ model in the presence or absence of Spint1. We observed that knockout of the F2rl1 gene, encoding PAR‐2, not only eliminated the enhanced formation of intestinal tumors caused by Spint1 deletion, but also reduced tumor formation in the presence of Spint1. Exacerbation of anemia and weight loss associated with HAI‐1 deficiency was also normalized by compound deficiency of PAR‐2. Mechanistically, signaling triggered by deregulated protease activities increased nuclear translocation of RelA/p65, vascular endothelial growth factor expression, and vascular density in ApcMin/+‐induced intestinal tumors. These results suggest that serine proteases promote intestinal carcinogenesis through activation of PAR‐2, and that HAI‐1 plays a critical tumor suppressor role as an inhibitor of matriptase, kallikreins, and other PAR‐2 activating proteases.
Collapse
Affiliation(s)
- Makiko Kawaguchi
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Koji Yamamoto
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Hiroaki Kataoka
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Aya Izumi
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Fumiki Yamashita
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Takumi Kiwaki
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Takahiro Nishida
- Department of Pathology, University of Miyazaki, Miyazaki, Japan
| | - Eric Camerer
- Inserm U970, Paris Cardiovascular Research Center, Université de Paris, Paris, France
| | | |
Collapse
|
15
|
Wettschureck N, Strilic B, Offermanns S. Passing the Vascular Barrier: Endothelial Signaling Processes Controlling Extravasation. Physiol Rev 2019; 99:1467-1525. [PMID: 31140373 DOI: 10.1152/physrev.00037.2018] [Citation(s) in RCA: 195] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A central function of the vascular endothelium is to serve as a barrier between the blood and the surrounding tissue of the body. At the same time, solutes and cells have to pass the endothelium to leave or to enter the bloodstream to maintain homeostasis. Under pathological conditions, for example, inflammation, permeability for fluid and cells is largely increased in the affected area, thereby facilitating host defense. To appropriately function as a regulated permeability filter, the endothelium uses various mechanisms to allow solutes and cells to pass the endothelial layer. These include transcellular and paracellular pathways of which the latter requires remodeling of intercellular junctions for its regulation. This review provides an overview on endothelial barrier regulation and focuses on the endothelial signaling mechanisms controlling the opening and closing of paracellular pathways for solutes and cells such as leukocytes and metastasizing tumor cells.
Collapse
Affiliation(s)
- Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Boris Strilic
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| |
Collapse
|
16
|
Noguerol J, Roustan PJ, N'Taye M, Delcombel L, Rolland C, Guiraud L, Sagnat D, Edir A, Bonnart C, Denadai-Souza A, Deraison C, Vergnolle N, Racaud-Sultan C. Sexual dimorphism in PAR 2-dependent regulation of primitive colonic cells. Biol Sex Differ 2019; 10:47. [PMID: 31492202 PMCID: PMC6731565 DOI: 10.1186/s13293-019-0262-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/26/2019] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Sexual dimorphism in biological responses is a critical knowledge for therapeutic proposals. However, gender differences in intestinal stem cell physiology have been poorly studied. Given the important role of the protease-activated receptor PAR2 in the control of colon epithelial primitive cells and cell cycle genes, we have performed a sex-based comparison of its expression and of the effects of PAR2 activation or knockout on cell proliferation and survival functions. METHODS Epithelial primitive cells isolated from colons from male and female mice were cultured as colonoids, and their number and size were measured. PAR2 activation was triggered by the addition of SLIGRL agonist peptide in the culture medium. PAR2-deficient mice were used to study the impact of PAR2 expression on colon epithelial cell culture and gene expression. RESULTS Colonoids from female mice were more abundant and larger compared to males, and these differences were further increased after PAR2 activation by specific PAR2 agonist peptide. The proliferation of male epithelial cells was lower compared to females but was specifically increased in PAR2 knockout male cells. PAR2 expression was higher in male colon cells compared to females and controlled the gene expression and activation of key negative signals of the primitive cell proliferation. This PAR2-dependent brake on the proliferation of male colon primitive cells was correlated with stress resistance. CONCLUSIONS Altogether, these data demonstrate that there is a sexual dimorphism in the PAR2-dependent regulation of primitive cells of the colon crypt.
Collapse
Affiliation(s)
- Julie Noguerol
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, CHU Purpan, place du Dr Baylac, 31024 Toulouse Cedex 3, Toulouse, France
| | - Pierre-Jean Roustan
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, CHU Purpan, place du Dr Baylac, 31024 Toulouse Cedex 3, Toulouse, France
| | - Mikael N'Taye
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, CHU Purpan, place du Dr Baylac, 31024 Toulouse Cedex 3, Toulouse, France
| | - Léo Delcombel
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, CHU Purpan, place du Dr Baylac, 31024 Toulouse Cedex 3, Toulouse, France
| | - Corinne Rolland
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, CHU Purpan, place du Dr Baylac, 31024 Toulouse Cedex 3, Toulouse, France
| | - Laura Guiraud
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, CHU Purpan, place du Dr Baylac, 31024 Toulouse Cedex 3, Toulouse, France
| | - David Sagnat
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, CHU Purpan, place du Dr Baylac, 31024 Toulouse Cedex 3, Toulouse, France
| | - Anissa Edir
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, CHU Purpan, place du Dr Baylac, 31024 Toulouse Cedex 3, Toulouse, France
| | - Chrystelle Bonnart
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, CHU Purpan, place du Dr Baylac, 31024 Toulouse Cedex 3, Toulouse, France
| | - Alexandre Denadai-Souza
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, CHU Purpan, place du Dr Baylac, 31024 Toulouse Cedex 3, Toulouse, France
| | - Céline Deraison
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, CHU Purpan, place du Dr Baylac, 31024 Toulouse Cedex 3, Toulouse, France
| | - Nathalie Vergnolle
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, CHU Purpan, place du Dr Baylac, 31024 Toulouse Cedex 3, Toulouse, France
| | - Claire Racaud-Sultan
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, CHU Purpan, place du Dr Baylac, 31024 Toulouse Cedex 3, Toulouse, France.
| |
Collapse
|
17
|
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
|
18
|
Opneja A, Kapoor S, Stavrou EX. Contribution of platelets, the coagulation and fibrinolytic systems to cutaneous wound healing. Thromb Res 2019; 179:56-63. [PMID: 31078121 DOI: 10.1016/j.thromres.2019.05.001] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/14/2019] [Accepted: 05/01/2019] [Indexed: 12/15/2022]
Abstract
Wound healing is a complex process that consists of multiple phases, each of which are indispensable for adequate repair. Timely initiation and resolution of each of these phases namely, hemostasis, inflammation, proliferation and tissue remodeling, is critical for promoting healing and avoiding excess scar formation. While platelets have long been known to influence the healing process, other components of blood particularly coagulation factors and the fibrinolytic system also contribute to efficient wound repair. This review aims to summarize our current understanding of the role of platelets, the coagulation and fibrinolytic systems in cutaneous wound healing, with a focus on how these components communicate with immune and non-immune cells in the wound microenvironment. We also outline current and potential therapeutic strategies to improve the management of chronic, non-healing wounds.
Collapse
Affiliation(s)
- Aman Opneja
- Department of Medicine, Hematology and Oncology Division, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Sargam Kapoor
- Department of Medicine, Hematology and Oncology Division, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Evi X Stavrou
- Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Medicine, Louis Stokes Veterans Administration Medical Center, VA Northeast Ohio Healthcare System, Cleveland, OH, USA.
| |
Collapse
|
19
|
Duarte FCK, Hurtig M, Clark A, Simpson J, Srbely JZ. Association between naturally occurring spine osteoarthritis in geriatric rats and neurogenic inflammation within neurosegmentally linked skeletal muscle. Exp Gerontol 2019; 118:31-38. [PMID: 30615897 DOI: 10.1016/j.exger.2019.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 12/12/2018] [Accepted: 01/02/2019] [Indexed: 12/15/2022]
Abstract
OBJECTIVE This study aimed to investigate the association between naturally occurring spinal osteoarthritis (OA) (L3-L5), the expression of substance P (SP) centrally (L4-L5) and the presence of neurogenic inflammation within the neurosegmentally linked quadriceps (L2-L5) in elderly rats versus young controls. DESIGN Eight aged (27 ± 3.2 months) and six young (4 ± 0.0 months) male Wistar Kyoto rats were euthanized and submitted to micro-computerized tomography for determination of spine OA. SP expression (% area) at the dorsal horn of the spinal cord as well as the relative expression of SP and protease-activated receptor 2 (PAR2) to alpha-tubulin within quadriceps muscle were determined by immunohistochemistry and Western Blot. RESULTS Spine osteoarthritis was confirmed in all aged rats but no young controls. Aged rats expressed significant increase of SP protein expression within the dorsal horn (MD = 0.086; 95% CI [0.026 to 0.145]; p = 0.0094) and quadriceps (MD = 1.209; 95% CI [0.239 to 2.179]; p = 0.0191) and PAR2 (MD = 0.797; 95% CI [0.160 to 1.435]; p = 0.0187) compared to young controls. CONCLUSION These observations provide novel insight into the potential role of neurogenic inflammation in the pathophysiology of myofascial pain syndrome in the naturally occurring spinal OA in elderly population.
Collapse
Affiliation(s)
- Felipe C K Duarte
- Department of Human Health and Nutritional Science, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada.
| | - Mark Hurtig
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, 50 McGilvray Lane, Guelph, ON N1G 2W1, Canada.
| | - Andrea Clark
- Department of Human Health and Nutritional Science, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada.
| | - Jeremy Simpson
- Department of Human Health and Nutritional Science, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada.
| | - John Z Srbely
- Department of Human Health and Nutritional Science, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada.
| |
Collapse
|
20
|
Francis N, Ayodele BA, O'Brien-Simpson NM, Birchmeier W, Pike RN, Pagel CN, Mackie EJ. Keratinocyte-specific ablation of protease-activated receptor 2 prevents gingival inflammation and bone loss in a mouse model of periodontal disease. Cell Microbiol 2018; 20:e12891. [PMID: 30009515 DOI: 10.1111/cmi.12891] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/13/2018] [Accepted: 07/06/2018] [Indexed: 01/10/2023]
Abstract
Chronic periodontitis is characterised by gingival inflammation and alveolar bone loss. A major aetiological agent is Porphyromonas gingivalis, which secretes proteases that activate protease-activated receptor 2 (PAR2 ). PAR2 expressed on oral keratinocytes is activated by proteases released by P. gingivalis, inducing secretion of interleukin 6 (IL-6), and global knockout of PAR2 prevents bone loss and inflammation in a periodontal disease model in mice. To test the hypothesis that PAR2 expressed on gingival keratinocytes is required for periodontal disease pathology, keratinocyte-specific PAR2 -null mice were generated using K14-Cre targeted deletion of the PAR2 gene (F2rl1). These mice were subjected to a model of periodontitis involving placement of a ligature around a tooth, combined with P. gingivalis infection ("Lig + Inf"). The intervention caused a significant 44% decrease in alveolar bone volume (assessed by microcomputed tomography) in wildtype (K14-Cre:F2rl1wt/wt ), but not littermate keratinocyte-specific PAR2 -null (K14-Cre:F2rl1fl/fl ) mice. Keratinocyte-specific ablation of PAR2 prevented the significant Lig + Inf-induced increase (2.8-fold) in the number of osteoclasts in alveolar bone and the significant up-regulation (2.4-4-fold) of the inflammatory markers IL-6, IL-1β, interferon-γ, myeloperoxidase, and CD11b in gingival tissue. These data suggest that PAR2 expressed on oral epithelial cells is a critical regulator of periodontitis-induced bone loss and will help in designing novel therapies with which to treat the disease.
Collapse
Affiliation(s)
- Nidhish Francis
- Department of Veterinary Biosciences, Melbourne Veterinary School, University of Melbourne, Parkville, Victoria, Australia
| | - Babatunde A Ayodele
- Department of Veterinary Biosciences, Melbourne Veterinary School, University of Melbourne, Parkville, Victoria, Australia
| | - Neil M O'Brien-Simpson
- Melbourne Dental School, Bio21 Institute, University of Melbourne, Parkville, Victoria, Australia
| | | | - Robert N Pike
- La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Charles N Pagel
- Department of Veterinary Biosciences, Melbourne Veterinary School, University of Melbourne, Parkville, Victoria, Australia
| | | |
Collapse
|
21
|
Asaduzzaman M, Davidson C, Nahirney D, Fiteih Y, Puttagunta L, Vliagoftis H. Proteinase-activated receptor-2 blockade inhibits changes seen in a chronic murine asthma model. Allergy 2018; 73:416-420. [PMID: 28940559 DOI: 10.1111/all.13313] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Proteinase-Activated Receptor-2 (PAR2 ) is a G protein-coupled receptor activated by serine proteinases. We have shown that PAR2 activation in the airways is involved in the development of allergic inflammation and airway hyperresponsiveness (AHR) in acute murine models. We hypothesized that functional inhibition of PAR2 prevents allergic inflammation, AHR and airway remodeling in chronic allergic airway inflammation models. MATERIAL AND METHODS We developed and used a 12 week model of cockroach extract (CE)-mediated AHR, airway inflammation and remodeling in BALB/c mice. RESULTS Mice sensitized and challenged with CE for 12 weeks exhibit AHR, increased numbers of eosinophils in bronchoalveolar lavage (BAL) and increased collagen content in the lung tissue compared to saline controls. Administration of an anti-PAR2 antibody, SAM-11, after the initial development of airway inflammation significantly inhibited all these parameters. CONCLUSIONS Our data demonstrate that PAR2 signaling plays a key role in CE-induced AHR and airway inflammation/remodeling in long term models of allergic airway inflammation. Targeting PAR2 activation may be a successful therapeutic strategy for allergic asthma.
Collapse
Affiliation(s)
- M. Asaduzzaman
- Department of Medicine; Pulmonary Research Group; University of Alberta; Edmonton AB Canada
| | - C. Davidson
- Department of Medicine; Pulmonary Research Group; University of Alberta; Edmonton AB Canada
| | - D. Nahirney
- Department of Medicine; Pulmonary Research Group; University of Alberta; Edmonton AB Canada
| | - Y. Fiteih
- Department of Medicine; Pulmonary Research Group; University of Alberta; Edmonton AB Canada
| | - L. Puttagunta
- Department of Laboratory Medicine and Pathology; University of Alberta; Edmonton AB Canada
| | - H. Vliagoftis
- Department of Medicine; Pulmonary Research Group; University of Alberta; Edmonton AB Canada
| |
Collapse
|
22
|
A T cell-specific knockout reveals an important role for protease-activated receptor 2 in lymphocyte development. Int J Biochem Cell Biol 2017; 92:95-103. [DOI: 10.1016/j.biocel.2017.09.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 09/19/2017] [Accepted: 09/21/2017] [Indexed: 01/25/2023]
|
23
|
Bonnart C, Feuillet G, Vasseur V, Cenac N, Vergnolle N, Blanchard N. Protease-activated receptor 2 contributes to Toxoplasma gondii
-mediated gut inflammation. Parasite Immunol 2017; 39. [DOI: 10.1111/pim.12489] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 09/01/2017] [Indexed: 02/02/2023]
Affiliation(s)
- C. Bonnart
- Institut de Recherche en Santé Digestive (IRSD); Université de Toulouse; INSERM, INRA, ENVT, UPS; Toulouse France
| | - G. Feuillet
- Institut de Recherche en Santé Digestive (IRSD); Université de Toulouse; INSERM, INRA, ENVT, UPS; Toulouse France
| | - V. Vasseur
- Centre de Physiopathologie de Toulouse Purpan (CPTP); Université de Toulouse; INSERM, CNRS, UPS; Toulouse France
| | - N. Cenac
- Institut de Recherche en Santé Digestive (IRSD); Université de Toulouse; INSERM, INRA, ENVT, UPS; Toulouse France
| | - N. Vergnolle
- Institut de Recherche en Santé Digestive (IRSD); Université de Toulouse; INSERM, INRA, ENVT, UPS; Toulouse France
| | - N. Blanchard
- Centre de Physiopathologie de Toulouse Purpan (CPTP); Université de Toulouse; INSERM, CNRS, UPS; Toulouse France
| |
Collapse
|
24
|
Muley MM, Krustev E, Reid AR, McDougall JJ. Prophylactic inhibition of neutrophil elastase prevents the development of chronic neuropathic pain in osteoarthritic mice. J Neuroinflammation 2017; 14:168. [PMID: 28835277 PMCID: PMC5569523 DOI: 10.1186/s12974-017-0944-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 08/16/2017] [Indexed: 12/15/2022] Open
Abstract
Background A subset of osteoarthritis (OA) patients experience joint pain with neuropathic characteristics. Mediators such as neutrophil elastase, a serine proteinase, may be released during acute OA inflammatory flares. We have previously shown that local administration of neutrophil elastase causes joint inflammation and pain via activation of proteinase-activated receptor-2 (PAR2). The aim of this study was to examine the contribution of endogenous neutrophil elastase and PAR2 to the development of joint inflammation, pain, and neuropathy associated with monoiodoacetate (MIA)-induced experimental OA. Methods MIA (0.3 mg/10 μl) was injected into the right knee joint of male C57BL/6 mice (20–34 g). Joint inflammation (edema, leukocyte kinetics), neutrophil elastase proteolytic activity, tactile allodynia, and saphenous nerve demyelination were assessed over 14 days post-injection. The effects of inhibiting neutrophil elastase during the early inflammatory phase of MIA (days 0 to 3) were determined using sivelestat (50 mg/kg i.p.) and serpinA1 (10 μg i.p.). Involvement of PAR2 in the development of MIA-induced joint inflammation and pain was studied using the PAR2 antagonist GB83 (5 μg i.p. days 0 to 1) and PAR2 knockout animals. Results MIA caused an increase in neutrophil elastase proteolytic activity on day 1 (P < 0.0001), but not on day 14. MIA also generated a transient inflammatory response which peaked on day 1 (P < 0.01) then subsided over the 2-week time course. Joint pain appeared on day 1 and persisted to day 14 (P < 0.0001). By day 14, the saphenous nerve showed signs of demyelination. Early treatment with sivelestat and serpinA1 blocked the proteolytic activity of neutrophil elastase on day 1 (P < 0.001), and caused lasting improvements in joint inflammation, pain, and saphenous nerve damage (P < 0.05). MIA-induced synovitis was reversed by early treatment with GB83 and attenuated in PAR2 knockout mice (P < 0.05). PAR2 knockout mice also showed reduced MIA-induced joint pain (P < 0.0001) and less nerve demyelination (P = 0.81 compared to saline control). Conclusions Neutrophil elastase and PAR2 contribute significantly to the development of joint inflammation, pain, and peripheral neuropathy associated with experimental OA, suggesting their potential as therapeutic targets.
Collapse
Affiliation(s)
- Milind M Muley
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, 5850 College Street, PO Box 15000, Halifax, Nova Scotia, B3H 4R2, Canada
| | - Eugene Krustev
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, 5850 College Street, PO Box 15000, Halifax, Nova Scotia, B3H 4R2, Canada
| | - Allison R Reid
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, 5850 College Street, PO Box 15000, Halifax, Nova Scotia, B3H 4R2, Canada
| | - Jason J McDougall
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, 5850 College Street, PO Box 15000, Halifax, Nova Scotia, B3H 4R2, Canada.
| |
Collapse
|
25
|
Gao S, Zhu H, Yang H, Zhang H, Li Q, Luo H. The role and mechanism of cathepsin G in dermatomyositis. Biomed Pharmacother 2017; 94:697-704. [PMID: 28797985 DOI: 10.1016/j.biopha.2017.07.088] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 07/18/2017] [Accepted: 07/19/2017] [Indexed: 11/20/2022] Open
Abstract
Dermatomyositis (DM) is an idiopathic inflammatory myopathy characterized by CD4+ T cells and B cells infiltration in perivascular and muscle tissue. Although the infiltration of inflammatory cells plays a key role in muscle damage, the exact mechanism is not clear. Cathepsin G (CTSG) is a member of the serine proteases family and can increase the permeability of vascular endothelial cells and the chemotaxis of inflammatory cells. In this study, we found that the expression of CTSG was increased in peripheral blood mononuclear cells and muscle tissues of DM patients. The activity of CTSG was significantly increased in DM patients and correlated with disease activity. Serum CTSG induced the expression of protease activated receptor 2 (PAR2) and altered the cytoskeleton of human dermal microvascular endothelial cells. Our studies indicate, for the first time, that CTSG may play an important role in muscle inflammatory cells infiltration by increasing the permeability of vascular endothelial cells.
Collapse
Affiliation(s)
- Siming Gao
- Department of Rheumatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| | - Honglin Zhu
- Department of Rheumatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| | - Huan Yang
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| | - Huali Zhang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| | - Qiuxiang Li
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China
| | - Hui Luo
- Department of Rheumatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, People's Republic of China.
| |
Collapse
|
26
|
Abstract
As the largest receptor gene family in the human genome, with >800 members, the signal-transducing G protein-coupled receptors (GPCRs) play critical roles in nearly all conceivable physiological processes, ranging from the sensing of photons and odorants to metabolic homeostasis and migration of leukocytes. Unfortunately, an exhaustive review of the several hundred GPCRs expressed by myeloid cells/macrophages (P.J. Groot-Kormelink, L .Fawcett, P.D. Wright, M. Gosling, and T.C. Kent, BMC Immunol 12:57, 2012, doi:10.1186/1471-2172-13-57) is beyond the scope of this chapter; however, we will endeavor to cover the GPCRs that contribute to the major facets of macrophage biology, i.e., those whose expression is restricted to macrophages and the GPCRs involved in macrophage differentiation/polarization, microbial elimination, inflammation and resolution, and macrophage-mediated pathology. The chemokine receptors, a major group of myeloid GPCRs, will not be extensively covered as they are comprehensively reviewed elsewhere.
Collapse
|
27
|
Rovai ES, Holzhausen M. The Role of Proteinase-Activated Receptors 1 and 2 in the Regulation of Periodontal Tissue Metabolism and Disease. J Immunol Res 2017; 2017:5193572. [PMID: 28503577 PMCID: PMC5414592 DOI: 10.1155/2017/5193572] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/13/2017] [Accepted: 03/05/2017] [Indexed: 01/13/2023] Open
Abstract
Proteinase-activated receptors 1 (PAR1) and 2 (PAR2) are the most highly expressed members of the PAR family in the periodontium. These receptors regulate periodontal inflammatory and repair processes through their activation by endogenous and bacterial enzymes. PAR1 is expressed by the periodontal cells such as human gingival fibroblasts, gingival epithelial cells, periodontal ligament cells, osteoblasts, and monocytic cells and can be activated by thrombin, matrix metalloproteinase 1 (MMP-1), MMP-13, fibrin, and gingipains from Porphyromonas gingivalis. PAR2 is expressed by neutrophils, osteoblasts, oral epithelial cells, and human gingival fibroblasts, and its possible activators in the periodontium are gingipains, neutrophil proteinase 3, and mast cell tryptase. The mechanisms through which PARs can respond to periodontal enzymes and result in appropriate immune responses have until recently been poorly understood. This review discusses recent findings that are beginning to identify a cardinal role for PAR1 and PAR2 on periodontal tissue metabolism.
Collapse
Affiliation(s)
- E. S. Rovai
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, SP, Brazil
| | - M. Holzhausen
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
28
|
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
|
29
|
Kanemaru A, Yamamoto K, Kawaguchi M, Fukushima T, Lin CY, Johnson MD, Camerer E, Kataoka H. Deregulated matriptase activity in oral squamous cell carcinoma promotes the infiltration of cancer-associated fibroblasts by paracrine activation of protease-activated receptor 2. Int J Cancer 2016; 140:130-141. [PMID: 27615543 DOI: 10.1002/ijc.30426] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 08/24/2016] [Accepted: 09/05/2016] [Indexed: 12/29/2022]
Abstract
Cancer-associated fibroblasts (CAFs) are known to contribute to cancer progression. We have reported that cell surface expression of hepatocyte growth factor activator inhibitor 1 (HAI-1) is decreased in invasive oral squamous cell carcinoma (OSCC) cells. This study examined if HAI-1-insufficiency contributes to CAF recruitment in OSCC. Serum-free conditioned medium (SFCM) from a human OSCC line (SAS) stimulated the migration of 3 human fibroblast cell lines, NB1RGB, MRC5 and KD. SFCM from HAI-1-knockdown SAS showed an additive effect on the migration of NB1RGB and MRC5, but not KD. SAS SFCM induced protease-activated receptor-2 (PAR-2) expression in NB1RGB and MRC5, but not in KD, and a PAR-2 antagonist blocked the stimulatory effect of HAI-1 knockdown on migration of the PAR-2 expressing cell lines. Moreover, HAI-1-deficient SFCM showed additive stimulatory effects on the migration of wild-type but not PAR-2-deficient mouse fibroblasts. Therefore, the enhanced migration induced by HAI-1-insufficiency was mediated by PAR-2 activation in fibroblasts. This activation resulted from the deregulation of the activity of matriptase, a PAR-2 agonist protease. HAI-1 may thus prevent CAF recruitment to OSCC by controlling matriptase activity. When HAI-1 expression is reduced on OSCC, matriptase may contribute to CAF accumulation by paracrine activation of fibroblast PAR-2. Immunohistochemical analysis of resected OSCC revealed increased PAR2-positive CAFs in 35% (33/95) of the cases studied. The increased PAR-2 positive CAFs tended to correlate with infiltrative histology of the invasion front and shorter disease-free survival of the patients.
Collapse
Affiliation(s)
- Ai Kanemaru
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Japan
| | - Koji Yamamoto
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Japan
| | - Makiko Kawaguchi
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Japan
| | - Tsuyoshi Fukushima
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Japan
| | - Chen-Yong Lin
- School of Medicine, Lambardi Comprehensive Cancer Centre, Georgetown University, Washington, DC
| | - Michael D Johnson
- School of Medicine, Lambardi Comprehensive Cancer Centre, Georgetown University, Washington, DC
| | - Eric Camerer
- INSERM U970, Paris Cardiovascular Research Centre, Paris, France
| | - Hiroaki Kataoka
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Japan
| |
Collapse
|
30
|
Asaduzzaman M, Nadeem A, Arizmendi N, Davidson C, Nichols HL, Abel M, Ionescu LI, Puttagunta L, Thebaud B, Gordon J, DeFea K, Hollenberg MD, Vliagoftis H. Functional inhibition of PAR2 alleviates allergen-induced airway hyperresponsiveness and inflammation. Clin Exp Allergy 2016; 45:1844-55. [PMID: 26312432 DOI: 10.1111/cea.12628] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 05/28/2015] [Accepted: 06/07/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND Proteinase-activated receptor 2 (PAR2 ) is a G protein-coupled receptor activated by trypsin-like serine proteinases. PAR2 activation has been associated with inflammation including allergic airway inflammation. We have also shown that PAR2 activation in the airways leads to allergic sensitization. The exact contribution of PAR2 in the development of eosinophilic inflammation and airway hyperresponsiveness (AHR) in sensitized individuals is not clear. OBJECTIVE To investigate whether functional inhibition of PAR2 during allergen challenge of allergic mice would inhibit allergen-induced AHR and inflammation in mouse models of asthma. METHODS Mice were sensitized and challenged with ovalbumin (OVA) or cockroach extract (CE). To investigate the role of PAR2 in the development of AHR and airway inflammation, we administered blocking anti-PAR2 antibodies, or a cell permeable peptide inhibitor of PAR2 signalling, pepducin, i.n. before allergen challenges and then assessed AHR and airway inflammation. RESULTS Administration of anti-PAR2 antibodies significantly inhibited OVA- and CE-induced AHR and airway inflammation. In particular, two anti-PAR2 antibodies, the monoclonal SAM-11 and polyclonal B5, inhibited AHR, airway eosinophilia, the increase of cytokines in the lung tissue and antigen-specific T cell proliferation, but had no effect on antigen-specific IgG and IgE levels. Pepducin was also effective in inhibiting AHR and airway inflammation in an OVA model of allergic airway inflammation. CONCLUSIONS AND CLINICAL RELEVANCE Functional blockade of PAR2 in the airways during allergen challenge improves allergen-induced AHR and inflammation in mice. Therefore, topical PAR2 blockade in the airways, through anti-PAR2 antibodies or molecules that interrupt PAR2 signalling, has the potential to be used as a therapeutic option in allergic asthma.
Collapse
Affiliation(s)
- M Asaduzzaman
- Pulmonary Research Group, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - A Nadeem
- Pulmonary Research Group, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - N Arizmendi
- Pulmonary Research Group, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - C Davidson
- Pulmonary Research Group, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - H L Nichols
- Division of Biomedical Sciences and Cell, Molecular and Developmental Biology, University of California, Riverside, CA, USA
| | - M Abel
- Pulmonary Research Group, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - L I Ionescu
- Department of Physiology, Women and Children Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - L Puttagunta
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - B Thebaud
- Department of Physiology, Women and Children Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - J Gordon
- Immunology Research Group, University of Saskatchewan, Saskatoon, SK, Canada
| | - K DeFea
- Division of Biomedical Sciences and Cell, Molecular and Developmental Biology, University of California, Riverside, CA, USA
| | - M D Hollenberg
- Department of Pharmacology and Therapeutics, University of Calgary, Calgary, AB, Canada
| | - H Vliagoftis
- Pulmonary Research Group, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
31
|
Lieu T, Savage E, Zhao P, Edgington-Mitchell L, Barlow N, Bron R, Poole DP, McLean P, Lohman RJ, Fairlie DP, Bunnett NW. Antagonism of the proinflammatory and pronociceptive actions of canonical and biased agonists of protease-activated receptor-2. Br J Pharmacol 2016; 173:2752-65. [PMID: 27423137 PMCID: PMC4995288 DOI: 10.1111/bph.13554] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/05/2016] [Accepted: 07/05/2016] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Diverse proteases cleave protease-activated receptor-2 (PAR2) on primary sensory neurons and epithelial cells to evoke pain and inflammation. Trypsin and tryptase activate PAR2 by a canonical mechanism that entails cleavage within the extracellular N-terminus revealing a tethered ligand that activates the cleaved receptor. Cathepsin-S and elastase are biased agonists that cleave PAR2 at different sites to activate distinct signalling pathways. Although PAR2 is a therapeutic target for inflammatory and painful diseases, the divergent mechanisms of proteolytic activation complicate the development of therapeutically useful antagonists. EXPERIMENTAL APPROACH We investigated whether the PAR2 antagonist GB88 inhibits protease-evoked activation of nociceptors and protease-stimulated oedema and hyperalgesia in rodents. KEY RESULTS Intraplantar injection of trypsin, cathespsin-S or elastase stimulated mechanical and thermal hyperalgesia and oedema in mice. Oral GB88 or par2 deletion inhibited the algesic and proinflammatory actions of all three proteases, but did not affect basal responses. GB88 also prevented pronociceptive and proinflammatory effects of the PAR2-selective agonists 2-furoyl-LIGRLO-NH2 and AC264613. GB88 did not affect capsaicin-evoked hyperalgesia or inflammation. Trypsin, cathepsin-S and elastase increased [Ca(2+) ]i in rat nociceptors, which expressed PAR2. GB88 inhibited this activation of nociceptors by all three proteases, but did not affect capsaicin-evoked activation of nociceptors or inhibit the catalytic activity of the three proteases. CONCLUSIONS AND IMPLICATIONS GB88 inhibits the capacity of canonical and biased protease agonists of PAR2 to cause nociception and inflammation.
Collapse
Affiliation(s)
- T Lieu
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Australia
| | - E Savage
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Australia
| | - P Zhao
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Australia
| | - L Edgington-Mitchell
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Australia
| | - N Barlow
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Australia
| | - R Bron
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Australia
| | - D P Poole
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Australia
- Departments of Anatomy and Neuroscience, University of Melbourne, Melbourne, Australia
| | - P McLean
- Takeda Pharmaceuticals, Zurich, Switzerland
| | - R-J Lohman
- Centre for Inflammation and Disease Research and Centre for Pain Research, Institute of Molecular Bioscience, University of Queensland, Brisbane, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Queensland, Brisbane, Australia
| | - D P Fairlie
- Centre for Inflammation and Disease Research and Centre for Pain Research, Institute of Molecular Bioscience, University of Queensland, Brisbane, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Queensland, Brisbane, Australia
| | - N W Bunnett
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Australia
- Departments of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Australia
| |
Collapse
|
32
|
Nasri I, Bonnet D, Zwarycz B, d'Aldebert E, Khou S, Mezghani-Jarraya R, Quaranta M, Rolland C, Bonnart C, Mas E, Ferrand A, Cenac N, Magness S, Van Landeghem L, Vergnolle N, Racaud-Sultan C. PAR2-dependent activation of GSK3β regulates the survival of colon stem/progenitor cells. Am J Physiol Gastrointest Liver Physiol 2016; 311:G221-36. [PMID: 27313176 PMCID: PMC5007290 DOI: 10.1152/ajpgi.00328.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 05/16/2016] [Indexed: 01/31/2023]
Abstract
Protease-activated receptors PAR1 and PAR2 play an important role in the control of epithelial cell proliferation and migration. However, the survival of normal and tumor intestinal stem/progenitor cells promoted by proinflammatory mediators may be critical in oncogenesis. The glycogen synthase kinase-3β (GSK3β) pathway is overactivated in colon cancer cells and promotes their survival and drug resistance. We thus aimed to determine PAR1 and PAR2 effects on normal and tumor intestinal stem/progenitor cells and whether they involved GSK3β. First, PAR1 and PAR2 were identified in colon stem/progenitor cells by immunofluorescence. In three-dimensional cultures of murine crypt units or single tumor Caco-2 cells, PAR2 activation decreased numbers and size of normal or cancerous spheroids, and PAR2-deficient spheroids showed increased proliferation, indicating that PAR2 represses proliferation. PAR2-stimulated normal cells were more resistant to stress (serum starvation or spheroid passaging), suggesting prosurvival effects of PAR2 Accordingly, active caspase-3 was strongly increased in PAR2-deficient normal spheroids. PAR2 but not PAR1 triggered GSK3β activation through serine-9 dephosphorylation in normal and tumor cells. The PAR2-triggered GSK3β activation implicates an arrestin/PP2A/GSK3β complex that is dependent on the Rho kinase activity. Loss of PAR2 was associated with high levels of GSK3β nonactive form, strengthening the role of PAR2 in GSK3β activation. GSK3 pharmacological inhibition impaired the survival of PAR2-stimulated spheroids and serum-starved cells. Altogether our data identify PAR2/GSK3β as a novel pathway that plays a critical role in the regulation of stem/progenitor cell survival and proliferation in normal colon crypts and colon cancer.
Collapse
Affiliation(s)
- Imen Nasri
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France; ,2Laboratoire de Chimie des Substances Naturelles, Faculté des Sciences de Sfax, Université de Sfax, Sfax, Tunisia;
| | - Delphine Bonnet
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France; ,3Service de Médecine Interne, Fédération Digestive, Centre Hospitalier Universitaire Purpan, Toulouse, France;
| | - Bailey Zwarycz
- 4Departments of Medicine and Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina;
| | - Emilie d'Aldebert
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Sokchea Khou
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Raoudha Mezghani-Jarraya
- 2Laboratoire de Chimie des Substances Naturelles, Faculté des Sciences de Sfax, Université de Sfax, Sfax, Tunisia;
| | - Muriel Quaranta
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Corinne Rolland
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Chrystelle Bonnart
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Emmanuel Mas
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France; ,5Service de Gastroentérologie, Hépatologie et Nutrition, Hôpital des Enfants, Toulouse, France; and
| | - Audrey Ferrand
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Nicolas Cenac
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Scott Magness
- 4Departments of Medicine and Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina;
| | - Laurianne Van Landeghem
- 6Institut National de la Santé et de la Recherche Médicale U913, Université de Nantes, Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - Nathalie Vergnolle
- 1Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| | - Claire Racaud-Sultan
- Institut de Recherche en Santé Digestive, Université de Toulouse, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, Ecole Nationale Vétérinaire de Toulouse, Université Paul Sabatier, Toulouse, France;
| |
Collapse
|
33
|
Bar-Shavit R, Maoz M, Kancharla A, Jaber M, Agranovich D, Grisaru-Granovsky S, Uziely B. Protease-activated receptors (PARs) in cancer: Novel biased signaling and targets for therapy. Methods Cell Biol 2015; 132:341-58. [PMID: 26928551 DOI: 10.1016/bs.mcb.2015.11.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite the fact that G protein-coupled receptors (GPCRs) mediate numerous physiological processes and represent targets for therapeutics for a vast array of diseases, their role in tumor biology is under appreciated. Protease-activated receptors (PARs) form a family which belongs to GPCR class A. PAR1&2 emerge with a central role in epithelial malignancies. Although the part of PAR1&2 in cancer is on the rise, their underlying signaling events are poorly understood. We review hereby past, present, and future cancer-associated PAR biology. Mainly, their role in physiological (placenta-cytotophobalst) and patho-physiological invasion processes. The identification and characterization of signal pleckstrin homology (PH)-domain-binding motifs established critical sites for breast cancer growth in PAR1&2. Among the proteins found to harbor important PH-domains and are involved in PAR biology are Akt/PKB as also Etk/Bmx and Vav3. A point mutation in PAR2, H349A, but not R352A, abrogated PH-protein association and is sufficient to markedly reduce PAR2-instigated breast tumor growth in vivo as also placental extravillous trophoblast (EVT) invasion in vitro is markedly reduced. Similarly, the PAR1 mutant hPar1-7A, which is unable to bind PH-domain, inhibits mammary tumors and EVT invasion, endowing these motifs with physiological significance and underscoring the importance of these previously unknown PAR1 and PAR2 PH-domain-binding motifs in both pathological and physiological invasion processes.
Collapse
Affiliation(s)
- R Bar-Shavit
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - M Maoz
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - A Kancharla
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - M Jaber
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - D Agranovich
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | - B Uziely
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
34
|
Zhang D, Li S, Hu L, Sheng L, Chen L. Modulation of protease-activated receptor expression by Porphyromonas gingivalis in human gingival epithelial cells. BMC Oral Health 2015; 15:128. [PMID: 26476532 PMCID: PMC4609475 DOI: 10.1186/s12903-015-0105-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 10/05/2015] [Indexed: 02/07/2023] Open
Abstract
Background Protease-activated receptors (PARs) are G-protein-coupled receptors with an active role in mediating inflammation, pain and other functions. The oral pathogen Porphyromonas gingivalis (P. gingivalis) secretes proteases that activate PARs. The aim of this study was to elucidate the role of PARs in the pathogenesis of chronic periodontitis by expression analysis of PARs in human gingival epithelial cells (GECs) before and after P. gingivalis supernatants treatment. Methods GECs were isolated from healthy human gingival tissue samples. The expression of PARs in GECs was determined by reverse transcription-polymerase chain reaction (RT-PCR) and flow cytometry. The effect of P. gingivalis proteases was investigated by quantitative real-time reverse transcription polymerase chain reaction (QRT-PCR) and flow cytometry. Results PAR-1, PAR-2, and PAR-3 were expressed in GECs. PAR-4 was not found by both RT-PCR and flow cytometry. Analysis of gene expression using QRT-PCR showed an up-regulation of PAR-2 mRNA in comparison to the untreated control cells (P < 0.05). In contrast, the mRNA expressions of PAR-1 and PAR-3 were significantly down-regulated (P > 0.05) in response to P. gingivalis supernatant compared to that in unstimulated control cells. This effect was abrogated by the protease inhibitor TLCK (P < 0.05). The results of flow cytometry indicated PARs protein levels consistent with mRNA levels in the results of QRT-PCR. Conclusions Our study shows that PAR-1, PAR-2 and PAR-3 are expressed in GECs. P. gingivalis proteases play a role in the regulation of innate immune responses in GECs. GECs use PARs to recognize P. gingivalis and mediate cell responses involved in innate immunity.
Collapse
Affiliation(s)
- Diya Zhang
- Dental Department, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
| | - Shenglai Li
- Department of Oral and Maxillofacial Surgery, Stomatology Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China.
| | - Lingjing Hu
- Department of Oral Medicine, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
| | - Lieping Sheng
- Dental Department, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
| | - Lili Chen
- Department of Oral Medicine, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
| |
Collapse
|
35
|
Nadeem A, Alharbi NO, Vliagoftis H, Tyagi M, Ahmad SF, Sayed-Ahmed MM. Proteinase activated receptor-2-mediated dual oxidase-2 up-regulation is involved in enhanced airway reactivity and inflammation in a mouse model of allergic asthma. Immunology 2015; 145:391-403. [PMID: 25684443 DOI: 10.1111/imm.12453] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 01/27/2015] [Accepted: 02/09/2015] [Indexed: 12/31/2022] Open
Abstract
Airway epithelial cells (AECs) express a variety of receptors, which sense danger signals from various aeroallergens/pathogens being inhaled constantly. Proteinase-activated receptor 2 (PAR-2) is one such receptor and is activated by cockroach allergens, which have intrinsic serine proteinase activity. Recently, dual oxidases (DUOX), especially DUOX-2, have been shown to be involved in airway inflammation in response to Toll-like receptor activation. However, the association between PAR-2 and DUOX-2 has not been explored in airways of allergic mice. Therefore, this study investigated the contribution of DUOX-2/reactive oxygen species (ROS) signalling in airway reactivity and inflammation after PAR-2 activation. Mice were sensitized intraperitoneally with intact cockroach allergen extract (CE) in the presence of aluminium hydroxide followed by intranasal challenge with CE. Mice were then assessed for airway reactivity, inflammation, oxidative stress (DUOX-2, ROS, inducible nitric oxide synthase, nitrite, nitrotyrosine and protein carbonyls) and apoptosis (Bax, Bcl-2, caspase-3). Challenge with CE led to up-regulation of DUOX-2 and ROS in AECs with concomitant increases in airway reactivity/inflammation and parameters of oxidative stress, and apoptosis. All of these changes were significantly inhibited by intranasal administration of ENMD-1068, a small molecule antagonist of PAR-2 in allergic mice. Administration of diphenyliodonium to allergic mice also led to improvement of allergic airway responses via inhibition of the DUOX-2/ROS pathway; however, these effects were less pronounced than PAR-2 antagonism. The current study suggests that PAR-2 activation leads to up-regulation of the DUOX-2/ROS pathway in AECs, which is involved in regulation of airway reactivity and inflammation via oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Ahmed Nadeem
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Naif O Alharbi
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Harissios Vliagoftis
- Pulmonary Research Group, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Manoj Tyagi
- Pulmonary Medicine and Critical Care, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sheikh F Ahmad
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed M Sayed-Ahmed
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
36
|
LI MENGMENG, YANG XIAOMING, ZHANG YADONG, CHEN LIANG, LU HANYU, LI XIAOBO, YIN LIANHUA, ZHI XIULING. Activation of protease-activated receptor-2 is associated with increased expression of inflammatory factors in the adipose tissues of obese mice. Mol Med Rep 2015; 12:6227-34. [DOI: 10.3892/mmr.2015.4179] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 07/03/2015] [Indexed: 11/05/2022] Open
|
37
|
Muley MM, Reid AR, Botz B, Bölcskei K, Helyes Z, McDougall JJ. Neutrophil elastase induces inflammation and pain in mouse knee joints via activation of proteinase-activated receptor-2. Br J Pharmacol 2015; 173:766-77. [PMID: 26140667 DOI: 10.1111/bph.13237] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 06/05/2015] [Accepted: 06/30/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Neutrophil elastase plays a crucial role in arthritis. Here, its potential in triggering joint inflammation and pain was assessed, and whether these effects were mediated by proteinase-activated receptor-2 (PAR2). EXPERIMENTAL APPROACH Neutrophil elastase (5 μg) was injected into the knee joints of mice and changes in blood perfusion, leukocyte kinetics and paw withdrawal threshold were assessed. Similar experiments were performed in animals pretreated with the neutrophil elastase inhibitor sivelestat, the PAR2 antagonist GB83, the p44/42 MAPK inhibitor U0126 and in PAR2 receptor knockout (KO) mice. Neutrophil elastase activity was also evaluated in arthritic joints by fluorescent imaging and sivelestat was assessed for anti-inflammatory and analgesic properties. KEY RESULTS Intra-articular injection of neutrophil elastase caused an increase in blood perfusion, leukocyte kinetics and a decrease in paw withdrawal threshold. Sivelestat treatment suppressed this effect. The PAR2 antagonist GB83 reversed neutrophil elastase-induced synovitis and pain and these responses were also attenuated in PAR2 KO mice. The MAPK inhibitor U0126 also blocked neutrophil elastase-induced inflammation and pain. Active neutrophil elastase was increased in acutely inflamed knees as shown by an activatable fluorescent probe. Sivelestat appeared to reduce neutrophil elastase activity, but had only a moderate anti-inflammatory effect in this model. CONCLUSIONS AND IMPLICATIONS Neutrophil elastase induced acute inflammation and pain in knee joints of mice. These changes are PAR2-dependent and appear to involve activation of a p44/42 MAPK pathway. Blocking neutrophil elastase, PAR2 and p44/42 MAPK activity can reduce inflammation and pain, suggesting their utility as therapeutic targets.
Collapse
Affiliation(s)
- Milind M Muley
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Allison R Reid
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Bálint Botz
- Department of Pharmacology and Pharmacotherapy, University of Pécs, School of Medicine, Pécs, Hungary.,János Szentágothai Research Centre, University of Pécs, Molecular Pharmacology Research Team, Pécs, Hungary
| | - Kata Bölcskei
- Department of Pharmacology and Pharmacotherapy, University of Pécs, School of Medicine, Pécs, Hungary.,János Szentágothai Research Centre, University of Pécs, Molecular Pharmacology Research Team, Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, University of Pécs, School of Medicine, Pécs, Hungary.,MTA NAP B Pain Research Group, University of Pécs, School of Medicine, Pécs, Hungary.,János Szentágothai Research Centre, University of Pécs, Molecular Pharmacology Research Team, Pécs, Hungary
| | - Jason J McDougall
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada.,Department of Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
38
|
Marki A, Esko JD, Pries AR, Ley K. Role of the endothelial surface layer in neutrophil recruitment. J Leukoc Biol 2015; 98:503-15. [PMID: 25979432 DOI: 10.1189/jlb.3mr0115-011r] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 03/25/2015] [Indexed: 12/15/2022] Open
Abstract
Neutrophil recruitment in most tissues is limited to postcapillary venules, where E- and P-selectins are inducibly expressed by venular endothelial cells. These molecules support neutrophil rolling via binding of PSGL-1 and other ligands on neutrophils. Selectins extend ≤ 38 nm above the endothelial plasma membrane, and PSGL-1 extends to 50 nm above the neutrophil plasma membrane. However, endothelial cells are covered with an ESL composed of glycosaminoglycans that is ≥ 500 nm thick and has measurable resistance against compression. The neutrophil surface is also covered with a surface layer. These surface layers would be expected to completely shield adhesion molecules; thus, neutrophils should not be able to roll and adhere. However, in the cremaster muscle and in many other models investigated using intravital microscopy, neutrophils clearly roll, and their rolling is easily and quickly induced. This conundrum was thought to be resolved by the observation that the induction of selectins is accompanied by ESL shedding; however, ESL shedding only partially reduces the ESL thickness (to 200 nm) and thus is insufficient to expose adhesion molecules. In addition to its antiadhesive functions, the ESL also presents neutrophil arrest-inducing chemokines. ESL heparan sulfate can also bind L-selectin expressed by the neutrophils, which contributes to rolling and arrest. We conclude that ESL has both proadhesive and antiadhesive functions. However, most previous studies considered either only the proadhesive or only the antiadhesive effects of the ESL. An integrated model for the role of the ESL in neutrophil rolling, arrest, and transmigration is needed.
Collapse
Affiliation(s)
- Alex Marki
- *Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, USA; and Department of Physiology and Center for Cardiovascular Research, Charite, Berlin, Germany
| | - Jeffrey D Esko
- *Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, USA; and Department of Physiology and Center for Cardiovascular Research, Charite, Berlin, Germany
| | - Axel R Pries
- *Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, USA; and Department of Physiology and Center for Cardiovascular Research, Charite, Berlin, Germany
| | - Klaus Ley
- *Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, USA; and Department of Physiology and Center for Cardiovascular Research, Charite, Berlin, Germany
| |
Collapse
|
39
|
Zhao P, Lieu T, Barlow N, Sostegni S, Haerteis S, Korbmacher C, Liedtke W, Jimenez-Vargas NN, Vanner SJ, Bunnett NW. Neutrophil Elastase Activates Protease-activated Receptor-2 (PAR2) and Transient Receptor Potential Vanilloid 4 (TRPV4) to Cause Inflammation and Pain. J Biol Chem 2015; 290:13875-87. [PMID: 25878251 DOI: 10.1074/jbc.m115.642736] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Indexed: 12/25/2022] Open
Abstract
Proteases that cleave protease-activated receptor-2 (PAR(2)) at Arg(36)↓Ser(37) reveal a tethered ligand that binds to the cleaved receptor. PAR(2) activates transient receptor potential (TRP) channels of nociceptive neurons to induce neurogenic inflammation and pain. Although proteases that cleave PAR(2) at non-canonical sites can trigger distinct signaling cascades, the functional importance of the PAR(2)-biased agonism is uncertain. We investigated whether neutrophil elastase, a biased agonist of PAR(2), causes inflammation and pain by activating PAR2 and TRP vanilloid 4 (TRPV4). Elastase cleaved human PAR(2) at Ala(66)↓Ser(67) and Ser(67)↓Val(68). Elastase stimulated PAR(2)-dependent cAMP accumulation and ERK1/2 activation, but not Ca(2+) mobilization, in KNRK cells. Elastase induced PAR(2) coupling to Gαs but not Gαq in HEK293 cells. Although elastase did not promote recruitment of G protein-coupled receptor kinase-2 (GRK(2)) or β-arrestin to PAR(2), consistent with its inability to promote receptor endocytosis, elastase did stimulate GRK6 recruitment. Elastase caused PAR(2)-dependent sensitization of TRPV4 currents in Xenopus laevis oocytes by adenylyl cyclase- and protein kinase A (PKA)-dependent mechanisms. Elastase stimulated PAR(2)-dependent cAMP formation and ERK1/2 phosphorylation, and a PAR(2)- and TRPV4-mediated influx of extracellular Ca(2+) in mouse nociceptors. Adenylyl cyclase and PKA-mediated elastase-induced activation of TRPV4 and hyperexcitability of nociceptors. Intraplantar injection of elastase to mice caused edema and mechanical hyperalgesia by PAR(2)- and TRPV4-mediated mechanisms. Thus, the elastase-biased agonism of PAR(2) causes Gαs-dependent activation of adenylyl cyclase and PKA, which activates TRPV4 and sensitizes nociceptors to cause inflammation and pain. Our results identify a novel mechanism of elastase-induced activation of TRPV4 and expand the role of PAR(2) as a mediator of protease-driven inflammation and pain.
Collapse
Affiliation(s)
- Peishen Zhao
- From the Monash Institute of Pharmaceutical Sciences and
| | - TinaMarie Lieu
- From the Monash Institute of Pharmaceutical Sciences and
| | | | - Silvia Sostegni
- the Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Silke Haerteis
- the Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Christoph Korbmacher
- the Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Wolfgang Liedtke
- the Department of Neurology, School of Medicine, Duke University, Durham, North Carolina 27710
| | - Nestor N Jimenez-Vargas
- the Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario N7L 3N6, Canada, and
| | - Stephen J Vanner
- the Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario N7L 3N6, Canada, and
| | - Nigel W Bunnett
- From the Monash Institute of Pharmaceutical Sciences and the ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville 3052, Australia, the Department of Pharmacology, University of Melbourne, Melbourne 3010, Australia
| |
Collapse
|
40
|
Cattaruzza F, Amadesi S, Carlsson JF, Murphy JE, Lyo V, Kirkwood K, Cottrell GS, Bogyo M, Knecht W, Bunnett NW. Serine proteases and protease-activated receptor 2 mediate the proinflammatory and algesic actions of diverse stimulants. Br J Pharmacol 2015; 171:3814-26. [PMID: 24749982 DOI: 10.1111/bph.12738] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 03/26/2014] [Accepted: 04/11/2014] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Although serine proteases and agonists of protease-activated receptor 2 (PAR2) cause inflammation and pain, the spectrum of proteases that are activated by proinflammatory and algesic stimuli and their contribution to inflammatory pain are uncertain. EXPERIMENTAL APPROACH Enzymic assays and selective inhibitors were used to characterize protease activity in mice after intraplantar injections of formalin, bradykinin, PAR2 activating peptide (AP) or vehicle. The capacity of these proteases and of recombinant mouse trypsin 4 to cleave fragments of PAR2 and to activate PAR2 in cell lines was determined. Protease inhibitors and par2 (-/-) mice were used to assess the contributions of proteases and PAR2 to pain and inflammation. KEY RESULTS Intraplantar injection of formalin, bradykinin or PAR2-AP led to the activation of proteases that were susceptible to the serine protease inhibitor melagatran but resistant to soybean trypsin inhibitor (SBTI). Melagatran inhibited mouse trypsin 4, which degraded SBTI. Proteases generated in inflamed tissues cleaved PAR2-derived peptides. These proteases and trypsin 4 increased [Ca(2+) ]i in PAR2-transfected but not in untransfected cells, and melagatran suppressed this activity. Melagatran or PAR2 deletion suppressed oedema and mechanical hypersensitivity induced by intraplantar formalin, bradykinin and PAR2-AP, but had no effect on capsaicin-induced pain. CONCLUSIONS AND IMPLICATIONS Diverse proinflammatory and algesic agents activate melagatran-sensitive serine proteases that cause inflammation and pain by a PAR2-mediated mechanism. By inducing self-activating proteases, PAR2 amplifies and sustains inflammation and pain. Serine protease inhibitors can attenuate the inflammatory and algesic effects of diverse stimuli, representing a useful therapeutic strategy.
Collapse
Affiliation(s)
- F Cattaruzza
- Department of Surgery, University of California, San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
EPCR-dependent PAR2 activation by the blood coagulation initiation complex regulates LPS-triggered interferon responses in mice. Blood 2015; 125:2845-54. [PMID: 25733582 DOI: 10.1182/blood-2014-11-610717] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 02/23/2015] [Indexed: 01/14/2023] Open
Abstract
Infection and inflammation are invariably associated with activation of the blood coagulation mechanism, secondary to the inflammation-induced expression of the coagulation initiator tissue factor (TF) on innate immune cells. By investigating the role of cell-surface receptors for coagulation factors in mouse endotoxemia, we found that the protein C receptor (ProcR; EPCR) was required for the normal in vivo and in vitro induction of lipopolysaccharide (LPS)-regulated gene expression. In cultured bone marrow-derived myeloid cells and in monocytic RAW264.7 cells, the LPS-induced expression of functionally active TF, assembly of the ternary TF-VIIa-Xa initiation complex of blood coagulation, and the EPCR-dependent activation of protease-activated receptor 2 (PAR2) by the ternary TF-VIIa-Xa complex were required for the normal LPS induction of messenger RNAs encoding the TLR3/4 signaling adaptor protein Pellino-1 and the transcription factor interferon regulatory factor 8. In response to in vivo challenge with LPS, mice lacking EPCR or PAR2 failed to fully initiate an interferon-regulated gene expression program that included the Irf8 target genes Lif, Iigp1, Gbp2, Gbp3, and Gbp6. The inflammation-induced expression of TF and crosstalk with EPCR, PAR2, and TLR4 therefore appear necessary for the normal evolution of interferon-regulated host responses.
Collapse
|
42
|
Jackson MT, Moradi B, Zaki S, Smith MM, McCracken S, Smith SM, Jackson CJ, Little CB. Depletion of protease-activated receptor 2 but not protease-activated receptor 1 may confer protection against osteoarthritis in mice through extracartilaginous mechanisms. Arthritis Rheumatol 2015; 66:3337-48. [PMID: 25200274 DOI: 10.1002/art.38876] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 09/04/2014] [Indexed: 01/03/2023]
Abstract
OBJECTIVE To explore the involvement of protease-activated receptor 1 (PAR-1) and PAR-2 in the pathologic processes of osteoarthritis (OA) and to identify the cells/tissues primarily affected by ablation of PAR-1 or PAR-2 in mice. METHODS OA was induced in the joints of wild-type (WT), PAR-1(+/+) , PAR-1(-/-) , and PAR-2(-/-) mice by destabilization of the medial meniscus (DMM), and scores of histologic features (cartilage aggrecan loss and erosion, subchondral bone sclerosis, osteophytes, and synovitis) were compared at 1, 4, and 8 weeks post-DMM. The effects of PAR ablation on cartilage degradation and chondrocyte metalloproteinase expression/activity were studied in cultures of mouse femoral head tissue with or without interleukin-1α (IL-1α). At 1 week post-DMM, synovial expression of cytokines and metalloproteinase genes was measured by reverse transcription-polymerase chain reaction, and populations of inflammatory cells were quantified by flow cytometry. RESULTS Deletion of PAR-2, but not that of PAR-1, in mice significantly delayed the progression of cartilage damage and inhibited subchondral bone sclerosis following DMM. There was no inhibitory effect of PAR-1 or PAR-2 ablation on IL-1α-induced cartilage degradation or chondrocyte metalloproteinase expression/activation. A low but significant level of synovitis persisted in mice subjected to DMM compared to that in control mice subjected to sham surgery, but no differences between the genotypes were seen 4 or 8 weeks post-DMM. One week after DMM, increased synovial expression of proinflammatory cytokines and metalloproteinase genes, along with increased levels of CD4+ T cells, inflammatory monocytes, and activated macrophages, were seen in all genotypes. However, there was a significant reduction in the percentage of activated macrophages in PAR-2(-/-) mice compared to PAR-1(-/-) and WT mice. CONCLUSION Deletion of PAR-2, but not that of PAR-1, results in a significant decrease in DMM-induced cartilage damage. The chondroprotection in PAR-2(-/-) mice appears to occur indirectly through modulation of extracartilaginous events such as subchondral bone remodeling and synovial macrophage activation, rather than through alteration of chondrocyte catabolic responses.
Collapse
Affiliation(s)
- Miriam T Jackson
- Kolling Institute of Medical Research and the University of Sydney at Royal North Shore Hospital, St. Leonards, New South Wales, Australia
| | | | | | | | | | | | | | | |
Collapse
|
43
|
White MJV, Galvis-Carvajal E, Gomer RH. A brief exposure to tryptase or thrombin potentiates fibrocyte differentiation in the presence of serum or serum amyloid p. THE JOURNAL OF IMMUNOLOGY 2014; 194:142-50. [PMID: 25429068 DOI: 10.4049/jimmunol.1401777] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A key question in both wound healing and fibrosis is the trigger for the initial formation of scar tissue. To help form scar tissue, circulating monocytes enter the tissue and differentiate into fibroblast-like cells called fibrocytes, but fibrocyte differentiation is strongly inhibited by the plasma protein serum amyloid P (SAP), and healthy tissues contain very few fibrocytes. In wounds and fibrotic lesions, mast cells degranulate to release tryptase, and thrombin mediates blood clotting in early wounds. Tryptase and thrombin are upregulated in wound healing and fibrotic lesions, and inhibition of these proteases attenuates fibrosis. We report that tryptase and thrombin potentiate human fibrocyte differentiation at biologically relevant concentrations and exposure times, even in the presence of concentrations of serum and SAP that normally completely inhibit fibrocyte differentiation. Fibrocyte potentiation by thrombin and tryptase is mediated by protease-activated receptors 1 and 2, respectively. Together, these results suggest that tryptase and thrombin may be an initial trigger to override SAP inhibition of fibrocyte differentiation to initiate scar tissue formation.
Collapse
Affiliation(s)
- Michael J V White
- Department of Biology, Texas A&M University, College Station, TX 77843-3474
| | | | - Richard H Gomer
- Department of Biology, Texas A&M University, College Station, TX 77843-3474
| |
Collapse
|
44
|
Crisante G, Battista L, Iwaszkiewicz J, Nesca V, Mérillat AM, Sergi C, Zoete V, Frateschi S, Hummler E. The CAP1/Prss8 catalytic triad is not involved in PAR2 activation and protease nexin-1 (PN-1) inhibition. FASEB J 2014; 28:4792-805. [PMID: 25138159 DOI: 10.1096/fj.14-253781] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Serine proteases, serine protease inhibitors, and protease-activated receptors (PARs) are responsible for several human skin disorders characterized by impaired epidermal permeability barrier function, desquamation, and inflammation. In this study, we addressed the consequences of a catalytically dead serine protease on epidermal homeostasis, the activation of PAR2 and the inhibition by the serine protease inhibitor nexin-1. The catalytically inactive serine protease CAP1/Prss8, when ectopically expressed in the mouse, retained the ability to induce skin disorders as well as its catalytically active counterpart (75%, n=81). Moreover, this phenotype was completely normalized in a PAR2-null background, indicating that the effects mediated by the catalytically inactive CAP1/Prss8 depend on PAR2 (95%, n=131). Finally, nexin-1 displayed analogous inhibitory capacity on both wild-type and inactive mutant CAP1/Prss8 in vitro and in vivo (64% n=151 vs. 89% n=109, respectively), indicating that the catalytic site of CAP1/Prss8 is dispensable for nexin-1 inhibition. Our results demonstrate a novel inhibitory interaction between CAP1/Prss8 and nexin-1, opening the search for specific CAP1/Prss8 antagonists that are independent of its catalytic activity.
Collapse
Affiliation(s)
| | | | - Justyna Iwaszkiewicz
- Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland
| | | | | | - Chloé Sergi
- Department of Pharmacology and Toxicology and
| | - Vincent Zoete
- Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland
| | | | | |
Collapse
|
45
|
Zhao P, Lieu T, Barlow N, Metcalf M, Veldhuis NA, Jensen DD, Kocan M, Sostegni S, Haerteis S, Baraznenok V, Henderson I, Lindström E, Guerrero-Alba R, Valdez-Morales EE, Liedtke W, McIntyre P, Vanner SJ, Korbmacher C, Bunnett NW. Cathepsin S causes inflammatory pain via biased agonism of PAR2 and TRPV4. J Biol Chem 2014; 289:27215-27234. [PMID: 25118282 DOI: 10.1074/jbc.m114.599712] [Citation(s) in RCA: 147] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Serine proteases such as trypsin and mast cell tryptase cleave protease-activated receptor-2 (PAR2) at R(36)↓S(37) and reveal a tethered ligand that excites nociceptors, causing neurogenic inflammation and pain. Whether proteases that cleave PAR2 at distinct sites are biased agonists that also induce inflammation and pain is unexplored. Cathepsin S (Cat-S) is a lysosomal cysteine protease of antigen-presenting cells that is secreted during inflammation and which retains activity at extracellular pH. We observed that Cat-S cleaved PAR2 at E(56)↓T(57), which removed the canonical tethered ligand and prevented trypsin activation. In HEK and KNRK cell lines and in nociceptive neurons of mouse dorsal root ganglia, Cat-S and a decapeptide mimicking the Cat-S-revealed tethered ligand-stimulated PAR2 coupling to Gαs and formation of cAMP. In contrast to trypsin, Cat-S did not mobilize intracellular Ca(2+), activate ERK1/2, recruit β-arrestins, or induce PAR2 endocytosis. Cat-S caused PAR2-dependent activation of transient receptor potential vanilloid 4 (TRPV4) in Xenopus laevis oocytes, HEK cells and nociceptive neurons, and stimulated neuronal hyperexcitability by adenylyl cyclase and protein kinase A-dependent mechanisms. Intraplantar injection of Cat-S caused inflammation and hyperalgesia in mice that was attenuated by PAR2 or TRPV4 deletion and adenylyl cyclase inhibition. Cat-S and PAR2 antagonists suppressed formalin-induced inflammation and pain, which implicates endogenous Cat-S and PAR2 in inflammatory pain. Our results identify Cat-S as a biased agonist of PAR2 that causes PAR2- and TRPV4-dependent inflammation and pain. They expand the role of PAR2 as a mediator of protease-driven inflammatory pain.
Collapse
Affiliation(s)
- Peishen Zhao
- Monash Institute of Pharmaceutical Sciences, Parkville 3052, Australia
| | - TinaMarie Lieu
- Monash Institute of Pharmaceutical Sciences, Parkville 3052, Australia
| | - Nicholas Barlow
- Monash Institute of Pharmaceutical Sciences, Parkville 3052, Australia
| | - Matthew Metcalf
- Monash Institute of Pharmaceutical Sciences, Parkville 3052, Australia
| | | | - Dane D Jensen
- Monash Institute of Pharmaceutical Sciences, Parkville 3052, Australia
| | - Martina Kocan
- Monash Institute of Pharmaceutical Sciences, Parkville 3052, Australia
| | - Silvia Sostegni
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Silke Haerteis
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | | | | | | | - Raquel Guerrero-Alba
- Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario N7L 3N6, Canada
| | - Eduardo E Valdez-Morales
- Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario N7L 3N6, Canada
| | - Wolfgang Liedtke
- Division of Neurology, Department of Medicine, Duke University, Durham, North Carolina 27710
| | - Peter McIntyre
- School of Medical Sciences and Health Innovations Research Institute, RMIT University, Bundoora 3083, Australia
| | - Stephen J Vanner
- Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario N7L 3N6, Canada
| | - Christoph Korbmacher
- Institut für Zelluläre und Molekulare Physiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Nigel W Bunnett
- Monash Institute of Pharmaceutical Sciences, Parkville 3052, Australia,; Department of Pharmacology, University of Melbourne, Melbourne 3010, Australia, and; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville 3052, Australia.
| |
Collapse
|
46
|
Kakarala KK, Jamil K, Devaraji V. Structure and putative signaling mechanism of Protease activated receptor 2 (PAR2) - a promising target for breast cancer. J Mol Graph Model 2014; 53:179-199. [PMID: 25173751 DOI: 10.1016/j.jmgm.2014.07.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 07/16/2014] [Accepted: 07/21/2014] [Indexed: 12/12/2022]
Abstract
Experimental evidences have observed enhanced expression of protease activated receptor 2 (PAR2) in breast cancer consistently. However, it is not yet recognized as an important therapeutic target for breast cancer as the primary molecular mechanisms of its activation are not yet well-defined. Nevertheless, recent reports on the mechanism of GPCR activation and signaling have given new insights to GPCR functioning. In the light of these details, we attempted to understand PAR2 structure & function using molecular modeling techniques. In this work, we generated averaged representative stable models of PAR2, using protease activated receptor 1 (PAR1) as a template and selected conformation based on their binding affinity with PAR2 specific agonist, GB110. Further, the selected model was used for studying the binding affinity of putative ligands. The selected ligands were based on a recent publication on phylogenetic analysis of Class A rhodopsin family of GPCRs. This study reports putative ligands, their interacting residues, binding affinity and molecular dynamics simulation studies on PAR2-ligand complexes. The results reported from this study would be useful for researchers and academicians to investigate PAR2 function as its physiological role is still hypothetical. Further, this information may provide a novel therapeutic scheme to manage breast cancer.
Collapse
Affiliation(s)
- Kavita Kumari Kakarala
- Centre for Biotechnology and Bioinformatics (CBB), School of Life Sciences, Jawaharlal Nehru Institute of Advanced Studies (JNIAS), 6th Floor, Buddha Bhawan, M.G. Road, Secunderabad 500003, Andhra Pradesh, India.
| | - Kaiser Jamil
- Centre for Biotechnology and Bioinformatics (CBB), School of Life Sciences, Jawaharlal Nehru Institute of Advanced Studies (JNIAS), 6th Floor, Buddha Bhawan, M.G. Road, Secunderabad 500003, Andhra Pradesh, India
| | - Vinod Devaraji
- College of Pharmacy, Madras Medical College, E.V.R. Periyar Salai, Chennai 600003, India
| |
Collapse
|
47
|
Role of protease-activated receptors for the innate immune response of the heart. Trends Cardiovasc Med 2014; 24:249-55. [PMID: 25066486 DOI: 10.1016/j.tcm.2014.06.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Revised: 06/04/2014] [Accepted: 06/21/2014] [Indexed: 02/07/2023]
Abstract
Protease-activated receptors (PARs) are a family of G-protein-coupled receptors with a unique activation mechanism via cleavage by the serine proteases of the coagulation cascade, immune cell-released proteases, and proteases from pathogens. Pathogens, such as viruses and bacteria, cause myocarditis and heart failure and PAR1 was shown to positively regulate the anti-viral innate immune response via interferon β during virus-induced myocarditis. In contrast, PAR2 negatively regulated the innate immune response and inhibited the interferon β expression. Thus, PARs play a central role for the innate immune response in the heart.
Collapse
|
48
|
Sales KU, Friis S, Konkel JE, Godiksen S, Hatakeyama M, Hansen KK, Rogatto SR, Szabo R, Vogel LK, Chen W, Gutkind JS, Bugge TH. Non-hematopoietic PAR-2 is essential for matriptase-driven pre-malignant progression and potentiation of ras-mediated squamous cell carcinogenesis. Oncogene 2014; 34:346-56. [PMID: 24469043 PMCID: PMC4112178 DOI: 10.1038/onc.2013.563] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 12/06/2013] [Accepted: 12/10/2013] [Indexed: 01/27/2023]
Abstract
The membrane-anchored serine protease, matriptase, is consistently dysregulated in a range of human carcinomas, and high matriptase activity correlates with poor prognosis. Furthermore, matriptase is unique among tumor-associated proteases in that epithelial stem cell expression of the protease suffices to induce malignant transformation. Here, we use genetic epistasis analysis to identify proteinase-activated receptor (PAR)-2-dependent inflammatory signaling as an essential component of matriptase-mediated oncogenesis. In cell-based assays, matriptase was a potent activator of PAR-2, and PAR-2 activation by matriptase caused robust induction of nuclear factor (NF)κB through Gαi. Importantly, genetic elimination of PAR-2 from mice completely prevented matriptase-induced pre-malignant progression, including inflammatory cytokine production, inflammatory cell recruitment, epidermal hyperplasia and dermal fibrosis. Selective ablation of PAR-2 from bone marrow-derived cells did not prevent matriptase-driven pre-malignant progression, indicating that matriptase activates keratinocyte stem cell PAR-2 to elicit its pro-inflammatory and pro-tumorigenic effects. When combined with previous studies, our data suggest that dual induction of PAR-2-NFκB inflammatory signaling and PI3K-Akt-mTor survival/proliferative signaling underlies the transforming potential of matriptase and may contribute to pro-tumorigenic signaling in human epithelial carcinogenesis.
Collapse
Affiliation(s)
- K U Sales
- 1] Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA [2] Clinical Research Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - S Friis
- 1] Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA [2] Department of Cellular and Molecular Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - J E Konkel
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - S Godiksen
- 1] Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA [2] Department of Cellular and Molecular Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark [3] Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - M Hatakeyama
- 1] Department of Urology, Faculty of Medicine, Sao Paulo State University (UNESP), Botucatu, Sao Paulo, Brazil [2] AC Camargo Cancer Center, Sao Paulo, Brazil
| | - K K Hansen
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - S R Rogatto
- 1] Department of Urology, Faculty of Medicine, Sao Paulo State University (UNESP), Botucatu, Sao Paulo, Brazil [2] AC Camargo Cancer Center, Sao Paulo, Brazil
| | - R Szabo
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - L K Vogel
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - W Chen
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - J S Gutkind
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - T H Bugge
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
49
|
Chung H, Ramachandran R, Hollenberg MD, Muruve DA. Proteinase-activated receptor-2 transactivation of epidermal growth factor receptor and transforming growth factor-β receptor signaling pathways contributes to renal fibrosis. J Biol Chem 2013; 288:37319-31. [PMID: 24253040 DOI: 10.1074/jbc.m113.492793] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Chronic kidney diseases cause significant morbidity and mortality in the population. During renal injury, kidney-localized proteinases can signal by cleaving and activating proteinase-activated receptor-2 (PAR2), a G-protein-coupled receptor involved in inflammation and fibrosis that is highly expressed in renal tubular cells. Following unilateral ureteric obstruction, PAR2-deficient mice displayed reduced renal tubular injury, fibrosis, collagen synthesis, connective tissue growth factor (CTGF), and α-smooth muscle actin gene expression at 7 days, compared with wild-type controls. In human proximal tubular epithelial cells in vitro, PAR2 stimulation with PAR2-activating peptide (PAR2-AP) alone significantly up-regulated the expression of CTGF, a potent profibrotic cytokine. The induction of CTGF by PAR2-AP was synergistically increased when combined with transforming growth factor-β (TGF-β). Consistent with these findings, treating human proximal tubular epithelial cells with PAR2-AP induced Smad2/3 phosphorylation in the canonical TGF-β signaling pathway. The Smad2 phosphorylation and CTGF induction required signaling via both the TGFβ-receptor and EGF receptor suggesting that PAR2 utilizes transactivation mechanisms to initiate fibrogenic signaling. Taken together, our data support the hypothesis that PAR2 synergizes with the TGFβ signaling pathway to contribute to renal injury and fibrosis.
Collapse
|
50
|
Periodontal treatment downregulates protease-activated receptor 2 in human gingival crevicular fluid cells. Infect Immun 2013; 81:4399-407. [PMID: 24042113 DOI: 10.1128/iai.01107-13] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Protease-activated receptor 2 (PAR2) is implicated in the pathogenesis of chronic inflammatory diseases, including periodontitis; it can be activated by gingipain and produced by Porphyromonas gingivalis and by neutrophil protease 3 (P3). PAR2 activation plays a relevant role in inflammatory processes by inducing the release of important inflammatory mediators associated with periodontal breakdown. The effects of periodontal treatment on PAR2 expression and its association with levels of proinflammatory mediators and activating proteases were investigated in chronic periodontitis patients. Positive staining for PAR2 was observed in gingival crevicular fluid cells and was reflective of tissue destruction. Overexpression of PAR2 was positively associated with inflammatory clinical parameters and with the levels of interleukin-6 (IL-6), IL-8, tumor necrosis factor alpha, matrix metalloprotease 2 (MMP-2), MMP-8, hepatocyte growth factor, and vascular endothelial growth factor. Elevated levels of gingipain and P3 and decreased levels of dentilisin and the protease inhibitors secretory leukocyte protease inhibitor and elafin were also associated with PAR2 overexpression. Healthy periodontal sites from individuals with chronic periodontitis showed diminished expression of PAR2 mRNA and the PAR2 protein (P < 0.05). Furthermore, periodontal treatment resulted in decreased PAR2 expression and correlated with decreased expression of inflammatory mediators and activating proteases. We concluded that periodontal treatment resulted in decreased levels of proteases and that proinflammatory mediators are associated with decreased PAR2 expression, suggesting that PAR2 expression is influenced by the presence of periodontal infection and is not a constitutive characteristic favoring periodontal inflammation.
Collapse
|