1
|
Gress C, Litzenburger T, Schmid R, Xiao K, Heissig F, Muller M, Gupta A, Hohlfeld JM. Transcriptomic characterization of the human segmental endotoxin challenge model. Sci Rep 2024; 14:1721. [PMID: 38242945 PMCID: PMC10798985 DOI: 10.1038/s41598-024-51547-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/06/2024] [Indexed: 01/21/2024] Open
Abstract
Segmental instillation of lipopolysaccharide (LPS) by bronchoscopy safely induces transient airway inflammation in human lungs. This model enables investigation of pulmonary inflammatory mechanisms as well as pharmacodynamic analysis of investigational drugs. The aim of this work was to describe the transcriptomic profile of human segmental LPS challenge with contextualization to major respiratory diseases. Pre-challenge bronchoalveolar lavage (BAL) fluid and biopsies were sampled from 28 smoking, healthy participants, followed by segmental instillation of LPS and saline as control. Twenty-four hours post instillation, BAL and biopsies were collected from challenged lung segments. Total RNA of cells from BAL and biopsy samples were sequenced and analysed for differentially expressed genes (DEGs). After challenge with LPS compared with saline, 6316 DEGs were upregulated and 241 were downregulated in BAL, but only one DEG was downregulated in biopsy samples. Upregulated DEGs in BAL were related to molecular functions such as "Inflammatory response" or "chemokine receptor activity", and upregulated pro-inflammatory pathways such as "Wnt-"/"Ras-"/"JAK-STAT" "-signaling pathway". Furthermore, the segmental LPS challenge model resembled aspects of the five most prevalent respiratory diseases chronic obstructive pulmonary disease (COPD), asthma, pneumonia, tuberculosis and lung cancer and featured similarities with acute exacerbations in COPD (AECOPD) and community-acquired pneumonia. Overall, our study provides extensive information about the transcriptomic profile from BAL cells and mucosal biopsies following LPS challenge in healthy smokers. It expands the knowledge about the LPS challenge model providing potential overlap with respiratory diseases in general and infection-triggered respiratory insults such as AECOPD in particular.
Collapse
Affiliation(s)
- Christina Gress
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Clinical Airway Research, 30625, Hannover, Germany
- German Center for Lung Research (DZL-BREATH), Hannover, Germany
| | | | - Ramona Schmid
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Ke Xiao
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Clinical Airway Research, 30625, Hannover, Germany
| | - Florian Heissig
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Meike Muller
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Clinical Airway Research, 30625, Hannover, Germany
- German Center for Lung Research (DZL-BREATH), Hannover, Germany
| | - Abhya Gupta
- Boehringer Ingelheim International GmbH, Biberach an der Riss, Germany
| | - Jens M Hohlfeld
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Clinical Airway Research, 30625, Hannover, Germany.
- German Center for Lung Research (DZL-BREATH), Hannover, Germany.
- Hannover Medical School, Department of Respiratory Medicine and Infectious Disease, Hannover, Germany.
| |
Collapse
|
2
|
Bruserud Ø, Mosevoll KA, Bruserud Ø, Reikvam H, Wendelbo Ø. The Regulation of Neutrophil Migration in Patients with Sepsis: The Complexity of the Molecular Mechanisms and Their Modulation in Sepsis and the Heterogeneity of Sepsis Patients. Cells 2023; 12:cells12071003. [PMID: 37048076 PMCID: PMC10093057 DOI: 10.3390/cells12071003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Common causes include gram-negative and gram-positive bacteria as well as fungi. Neutrophils are among the first cells to arrive at an infection site where they function as important effector cells of the innate immune system and as regulators of the host immune response. The regulation of neutrophil migration is therefore important both for the infection-directed host response and for the development of organ dysfunctions in sepsis. Downregulation of CXCR4/CXCL12 stimulates neutrophil migration from the bone marrow. This is followed by transmigration/extravasation across the endothelial cell barrier at the infection site; this process is directed by adhesion molecules and various chemotactic gradients created by chemotactic cytokines, lipid mediators, bacterial peptides, and peptides from damaged cells. These mechanisms of neutrophil migration are modulated by sepsis, leading to reduced neutrophil migration and even reversed migration that contributes to distant organ failure. The sepsis-induced modulation seems to differ between neutrophil subsets. Furthermore, sepsis patients should be regarded as heterogeneous because neutrophil migration will possibly be further modulated by the infecting microorganisms, antimicrobial treatment, patient age/frailty/sex, other diseases (e.g., hematological malignancies and stem cell transplantation), and the metabolic status. The present review describes molecular mechanisms involved in the regulation of neutrophil migration; how these mechanisms are altered during sepsis; and how bacteria/fungi, antimicrobial treatment, and aging/frailty/comorbidity influence the regulation of neutrophil migration.
Collapse
Affiliation(s)
- Øystein Bruserud
- Leukemia Research Group, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
- Correspondence:
| | - Knut Anders Mosevoll
- Section for Infectious Diseases, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
- Section for Infectious Diseases, Department of Clinical Research, University of Bergen, 5021 Bergen, Norway
| | - Øyvind Bruserud
- Department for Anesthesiology and Intensive Care, Haukeland University Hospital, 5021 Bergen, Norway
| | - Håkon Reikvam
- Leukemia Research Group, Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| | - Øystein Wendelbo
- Section for Infectious Diseases, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
- Faculty of Health, VID Specialized University, Ulriksdal 10, 5009 Bergen, Norway
| |
Collapse
|
3
|
Yakupu A, Zhang D, Guan H, Jiang M, Dong J, Niu Y, Tang J, Liu Y, Ma X, Lu S. Single-cell analysis reveals melanocytes may promote inflammation in chronic wounds through cathepsin G. Front Genet 2023; 14:1072995. [PMID: 36755572 PMCID: PMC9900029 DOI: 10.3389/fgene.2023.1072995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
During acute wound (AW) healing, a series of proper communications will occur between different epidermal cells at precise temporal stages to restore the integrity of the skin. However, it is still unclear what variation happened in epidermal cell interaction in the chronic wound environment. To provide new insights into chronic wound healing, we reconstructed the variations in the epidermal cell-cell communication network that occur in chronic wound healing via single-cell RNA-seq (scRNA-seq) data analysis. We found that the intricate cellular and molecular interactions increased in pressure ulcer (PU) compared to AW, especially the PARs signaling pathways were significantly upregulated. It shows that the PARs signaling pathways' main source was melanocytes and the CTSG-F2RL1 ligand-receptor pairs were its main contributor. Cathepsin G (CatG or CTSG) is a serine protease mainly with trypsin- and chymotrypsin-like specificity. It is synthesized and secreted by some immune or non-immune cells. Whereas, it has not been reported that melanocytes can synthesize and secrete the CTSG. F2R Like Trypsin Receptor 1 (F2RL1) is a member of proteinase-activated receptors (PARs) that are irreversibly activated by proteolytic cleavage and its stimulation can promote inflammation and inflammatory cell infiltration. In this study, we found that melanocytes increased in pressure ulcers, melanocytes can synthesize and secrete the CTSG and may promote inflammation in chronic wounds through CTSG-F2RL1 pairs, which may be a novel potential target and a therapeutic strategy in the treatment of chronic wounds.
Collapse
Affiliation(s)
- Aobuliaximu Yakupu
- Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,Wound Healing Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Di Zhang
- Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,Wound Healing Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Haonan Guan
- Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,Wound Healing Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Minfei Jiang
- Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,Wound Healing Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiaoyun Dong
- Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,Wound Healing Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yiwen Niu
- Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,Wound Healing Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiajun Tang
- Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,Wound Healing Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yingkai Liu
- Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,Wound Healing Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xian Ma
- Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,Wound Healing Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,*Correspondence: Xian Ma, ; Shuliang Lu,
| | - Shuliang Lu
- Department of Burn, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,Wound Healing Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China,*Correspondence: Xian Ma, ; Shuliang Lu,
| |
Collapse
|
4
|
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
|
5
|
Kim YS, Go G, Yun CW, Yea JH, Yoon S, Han SY, Lee G, Lee MY, Lee SH. Topical Administration of Melatonin-Loaded Extracellular Vesicle-Mimetic Nanovesicles Improves 2,4-Dinitrofluorobenzene-Induced Atopic Dermatitis. Biomolecules 2021; 11:1450. [PMID: 34680082 PMCID: PMC8533309 DOI: 10.3390/biom11101450] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/25/2021] [Accepted: 10/01/2021] [Indexed: 12/18/2022] Open
Abstract
Atopic dermatitis (AD) is caused by multiple factors that trigger chronic skin inflammation, including a defective skin barrier, immune cell activation, and microbial exposure. Although melatonin has an excellent biosafety profile and a potential to treat AD, there is limited clinical evidence from controlled trials that support the use of melatonin as an AD treatment. The delivery of melatonin via the transdermal delivery system is also a challenge in designing melatonin-based AD treatments. In this study, we generated melatonin-loaded extracellular vesicle-mimetic nanoparticles (MelaNVs) to improve the transdermal delivery of melatonin and to evaluate their therapeutic potential in AD. The MelaNVs were spherical nanoparticles with an average size of 100 nm, which is the optimal size for the transdermal delivery of drugs. MelaNVs showed anti-inflammatory effects by suppressing the release of TNF-α and β-hexosaminidase in LPS-treated RAW264.7 cells and compound 48/80-treated RBL-2H3 cells, respectively. MelaNVs showed a superior suppressive effect compared to an equivalent concentration of free melatonin. Treating a 2,4-dinitrofluorobenzene (DNCB)-induced AD-like mouse model with MelaNVs improved AD by suppressing local inflammation, mast cell infiltration, and fibrosis. In addition, MelaNVs effectively suppressed serum IgE levels and regulated serum IFN-γ and IL-4 levels. Taken together, these results suggest that MelaNVs are novel and efficient transdermal delivery systems of melatonin and that MelaNVs can be used as a treatment to improve AD.
Collapse
Affiliation(s)
- Yoon Seon Kim
- Department of Medical Science, Soonchunhyang University, Asan-si 31538, Korea;
| | - Gyeongyun Go
- Department of Biochemistry, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea; (G.G.); (J.-H.Y.); (G.L.)
- Department of Biochemistry, BK21FOUR Project2, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea
| | - Chul-Won Yun
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul 04401, Korea;
| | - Ji-Hye Yea
- Department of Biochemistry, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea; (G.G.); (J.-H.Y.); (G.L.)
| | - Sungtae Yoon
- Stembio Ltd., Entrepreneur 306, Asan-si 31538, Korea; (S.Y.); (S.-Y.H.)
| | - Su-Yeon Han
- Stembio Ltd., Entrepreneur 306, Asan-si 31538, Korea; (S.Y.); (S.-Y.H.)
| | - Gaeun Lee
- Department of Biochemistry, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea; (G.G.); (J.-H.Y.); (G.L.)
- Department of Biochemistry, BK21FOUR Project2, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea
| | - Mi-Young Lee
- Department of Medical Science, Soonchunhyang University, Asan-si 31538, Korea;
- Department of Medical Biotechnology, Soonchunhyang University, Asan-si 31538, Korea
| | - Sang Hun Lee
- Department of Biochemistry, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea; (G.G.); (J.-H.Y.); (G.L.)
- Department of Biochemistry, BK21FOUR Project2, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul 04401, Korea;
- Stembio Ltd., Entrepreneur 306, Asan-si 31538, Korea; (S.Y.); (S.-Y.H.)
| |
Collapse
|
6
|
Macrophage protease-activated receptor 2 regulates fetal liver erythropoiesis in mice. Blood Adv 2021; 4:5810-5824. [PMID: 33232477 DOI: 10.1182/bloodadvances.2020003299] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/14/2020] [Indexed: 12/23/2022] Open
Abstract
Deficiencies in many coagulation factors and protease-activated receptors (PARs) affect embryonic development. We describe a defect in definitive erythropoiesis in PAR2-deficient mice. Embryonic PAR2 deficiency increases embryonic death associated with variably severe anemia in comparison with PAR2-expressing embryos. PAR2-deficient fetal livers display reduced macrophage densities, erythroblastic island areas, and messenger RNA expression levels of markers for erythropoiesis and macrophages. Coagulation factor synthesis in the liver coincides with expanding fetal liver hematopoiesis during midgestation, and embryonic factor VII (FVII) deficiency impairs liver macrophage development. Cleavage-insensitive PAR2-mutant mice recapitulate the hematopoiesis defect of PAR2-deficient embryos, and macrophage-expressed PAR2 directly supports erythroblastic island function and the differentiation of red blood cells in the fetal liver. Conditional deletion of PAR2 in macrophages impairs erythropoiesis, as well as increases inflammatory stress, as evidenced by upregulation of interferon-regulated hepcidin antimicrobial peptide. In contrast, postnatal macrophage PAR2 deficiency does not have any effect on steady-state Kupffer cells, bone marrow macrophage numbers, or erythropoiesis, but erythropoiesis in macrophages from PAR2-deficient mice is impaired following hemolysis. These data identify a novel function for macrophage PAR2 signaling in adapting to rapid increases in blood demand during gestational development and postnatal erythropoiesis under stress conditions.
Collapse
|
7
|
Buhl T, Ikoma A, Kempkes C, Cevikbas F, Sulk M, Buddenkotte J, Akiyama T, Crumrine D, Camerer E, Carstens E, Schön MP, Elias P, Coughlin SR, Steinhoff M. Protease-Activated Receptor-2 Regulates Neuro-Epidermal Communication in Atopic Dermatitis. Front Immunol 2020; 11:1740. [PMID: 32903402 PMCID: PMC7435019 DOI: 10.3389/fimmu.2020.01740] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 06/29/2020] [Indexed: 01/01/2023] Open
Abstract
Background: Activation of protease-activated receptor-2 (PAR2) has been implicated in inflammation, pruritus, and skin barrier regulation, all characteristics of atopic dermatitis (AD), as well as Netherton syndrome which has similar characteristics. However, understanding the precise role of PAR2 on neuro-immune communication in AD has been hampered by the lack of appropriate animal models. Methods: We used a recently established mouse model with epidermal overexpression of PAR2 (PAR2OE) and littermate WT mice to study the impact of increased PAR2 expression in epidermal cells on spontaneous and house dust mite (HDM)-induced skin inflammation, itch, and barrier dysfunction in AD, in vivo and ex vivo. Results: PAR2OE newborns displayed no overt abnormalities, but spontaneously developed dry skin, severe pruritus, and eczema. Dermatological, neurophysiological, and immunological analyses revealed the hallmarks of AD-like skin disease. Skin barrier defects were observed before onset of skin lesions. Application of HDM onto PAR2OE mice triggered pruritus and the skin phenotype. PAR2OE mice displayed an increased density of nerve fibers, increased nerve growth factor and endothelin-1 expression levels, alloknesis, enhanced scratching (hyperknesis), and responses of dorsal root ganglion cells to non-histaminergic pruritogens. Conclusion: PAR2 in keratinocytes, activated by exogenous and endogenous proteases, is sufficient to drive barrier dysfunction, inflammation, and pruritus and sensitize skin to the effects of HDM in a mouse model that mimics human AD. PAR2 signaling in keratinocytes appears to be sufficient to drive several levels of neuro-epidermal communication, another feature of human AD.
Collapse
Affiliation(s)
- Timo Buhl
- Department of Dermatology and Surgery, University of California, San Francisco, San Francisco, CA, United States.,Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| | - Akihiko Ikoma
- Department of Dermatology and Surgery, University of California, San Francisco, San Francisco, CA, United States.,Department of Dermatology and UCD Charles Institute for Translational Dermatology, University College Dublin, Dublin, Ireland
| | - Cordula Kempkes
- Department of Dermatology and Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Ferda Cevikbas
- Department of Dermatology and Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Mathias Sulk
- Department of Dermatology and Surgery, University of California, San Francisco, San Francisco, CA, United States.,Department of Dermatology, University Hospital Münster, Münster, Germany
| | - Joerg Buddenkotte
- Department of Dermatology and Venerology, Hamad Medical Corporation, Doha, Qatar.,Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Tasuku Akiyama
- Department of Dermatology, Anatomy and Cell Biology, Temple Itch Center, Temple University, Philadelphia, PA, United States.,Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, United States
| | - Debbie Crumrine
- Department of Dermatology and Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Eric Camerer
- INSERM U970, Paris Cardiovascular Research Centre, Paris, France
| | - Earl Carstens
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA, United States
| | - Michael P Schön
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| | - Peter Elias
- Department of Dermatology and Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Shaun R Coughlin
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Martin Steinhoff
- Department of Dermatology and Surgery, University of California, San Francisco, San Francisco, CA, United States.,Department of Dermatology and UCD Charles Institute for Translational Dermatology, University College Dublin, Dublin, Ireland.,Department of Dermatology and Venerology, Hamad Medical Corporation, Doha, Qatar.,Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.,Department of Dermatology, Medical School, University of Qatar, Doha, Qatar.,School of Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar.,Department of Dermatology, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
8
|
Transcriptome Analysis and Emerging Driver Identification of CD8+ T Cells in Patients with Vitiligo. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2503924. [PMID: 31885781 PMCID: PMC6899274 DOI: 10.1155/2019/2503924] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/09/2019] [Indexed: 12/12/2022]
Abstract
Activated CD8+ T cells play important roles in the pathogenesis of vitiligo. However, driving factors about the activation and migration of CD8+ T cells remain obscure. In this study, we aim to identify differentially expressed genes (DEGs) and uncover potential factors that drive the disease in melanocyte-specific CD8+ T cells in vitiligo. A total of 1147 DEGs were found through transcriptome sequencing in CD8+ T cells from lesional skin of vitiligo patients and normal controls. Based on KEGG pathway enrichment analysis and PPI, 16 upregulated and 23 downregulated genes were identified. Ultimately, 3 genes were figured out after RT-qPCR verification. The mRNA and protein expression levels of PIK3CB, HIF-1α, and F2RL1 were all elevated in CD8+ T cells from peripheral blood in vitiligo. HIF-1α and PIK3CB were significantly increased in lesional skin of vitiligo. Two CpG sites of the HIF-1α promoter were hypomethylated in vitiligo CD8+ T cells. In conclusion, HIF-1α, F2RL1, and PIK3CB may act as novel drivers for vitiligo, which are all closely associated with reactive oxygen species and possibly contribute to the activation and/or migration of melanocyte-specific CD8+ T cells in vitiligo. In addition, we uncovered a potential role for DNA hypomethylation of HIF-1α in CD8+ T cells of vitiligo.
Collapse
|
9
|
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
|
10
|
Fukushima H, Alves VTE, Carvalho VFD, Ambrósio LMB, Eichler RADS, Carvalho MHCD, Saraiva L, Holzhausen M. PAR-2 expression in the gingival crevicular fluid reflects chronic periodontitis severity. Braz Oral Res 2017; 31:e16. [PMID: 28146220 DOI: 10.1590/1807-3107bor-2017.vol31.0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 12/08/2016] [Indexed: 11/22/2022] Open
Abstract
Recent studies investigating protease-activated receptor type 2 (PAR-2) suggest an association between the receptor and periodontal inflammation. It is known that gingipain, a bacterial protease secreted by the important periodontopathogen Porphyromonas gingivalis can activate PAR-2. Previous studies by our group found that PAR-2 is overexpressed in the gingival crevicular fluid (GCF) of patients with moderate chronic periodontitis (MP). The present study aimed at evaluating whether PAR-2 expression is associated with chronic periodontitis severity. GCF samples and clinical parameters, including plaque and bleeding on probing indices, probing pocket depth and clinical attachment level, were collected from the control group (n = 19) at baseline, and from MP patients (n = 19) and severe chronic periodontitis (SP) (n = 19) patients before and 6 weeks after periodontal non-surgical treatment. PAR-2 and gingipain messenger RNA (mRNA) in the GCF of 4 periodontal sites per patient were evaluated by Reverse Transcription Polymerase Chain Reaction (RT-qPCR). PAR-2 and gingipain expressions were greater in periodontitis patients than in control group patients. In addition, the SP group presented increased PAR-2 and gingipain mRNA levels, compared with the MP group. Furthermore, periodontal treatment significantly reduced (p <0.05) PAR-2 expression in patients with periodontitis. In conclusion, PAR-2 is associated with chronic periodontitis severity and with gingipain levels in the periodontal pocket, thus suggesting that PAR-2 expression in the GCF reflects the severity of destruction during periodontal infection.
Collapse
Affiliation(s)
- Henrique Fukushima
- Universidade de São Paulo - USP, School of Dentistry, Department of Stomatology, São Paulo, SP, Brazil
| | | | | | | | | | | | - Luciana Saraiva
- Universidade de São Paulo - USP, School of Dentistry, Department of Stomatology, São Paulo, SP, Brazil
| | - Marinella Holzhausen
- Universidade de São Paulo - USP, School of Dentistry, Department of Stomatology, São Paulo, SP, Brazil
| |
Collapse
|
11
|
Jayaprakash K, Demirel I, Khalaf H, Bengtsson T. The role of phagocytosis, oxidative burst and neutrophil extracellular traps in the interaction between neutrophils and the periodontal pathogenPorphyromonas gingivalis. Mol Oral Microbiol 2015; 30:361-75. [DOI: 10.1111/omi.12099] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2015] [Indexed: 01/11/2023]
Affiliation(s)
- K. Jayaprakash
- School of Health and Medical Sciences; Örebro University; Örebro Sweden
| | - I. Demirel
- School of Health and Medical Sciences; Örebro University; Örebro Sweden
| | - H. Khalaf
- School of Health and Medical Sciences; Örebro University; Örebro Sweden
| | - T. Bengtsson
- School of Health and Medical Sciences; Örebro University; Örebro Sweden
| |
Collapse
|
12
|
Wronkowitz N, Görgens SW, Romacho T, Villalobos LA, Sánchez-Ferrer CF, Peiró C, Sell H, Eckel J. Soluble DPP4 induces inflammation and proliferation of human smooth muscle cells via protease-activated receptor 2. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1613-21. [PMID: 24928308 DOI: 10.1016/j.bbadis.2014.06.004] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 05/27/2014] [Accepted: 06/02/2014] [Indexed: 12/27/2022]
Abstract
DPP4 is an ubiquitously expressed cell-surface protease that is shedded to the circulation as soluble DPP4 (sDPP4). We recently identified sDPP4 as a novel adipokine potentially linking obesity to the metabolic syndrome. The aim of this study was to investigate direct effects of sDPP4 on human vascular smooth muscle cells (hVSMCs) and to identify responsible signaling pathways. Using physiological concentrations of sDPP4, we could observe a concentration-dependent activation of ERK1/2 (3-fold) after 6h, which remained stable for up to 24h. Additionally, sDPP4 treatment induced a 1.5-fold phosphorylation of the NF-κB subunit p65. In accordance with sDPP4-induced stress and inflammatory signaling, sDPP4 also stimulates hVSMC proliferation. Furthermore we could observe an increased expression and secretion of pro-inflammatory cytokines like interleukin (IL)-6, IL-8 and MCP-1 (2.5-, 2.4- and 1.5-fold, respectively) by the sDPP4 treatment. All direct effects of sDPP4 on signaling, proliferation and inflammation could completely be prevented by DPP4 inhibition. Bioinformatic analysis and signaling signature induced by sDPP4 suggest that sDPP4 might be an agonist for PAR2. After the silencing of PAR2, the sDPP4-induced ERK activation as well as the proliferation was totally abolished. Additionally, the sDPP4-induced upregulation of IL-6 and IL-8 could completely be prevented by the PAR2 silencing. In conclusion, we show for the first time that sDPP4 directly activates the MAPK and NF-κB signaling cascade involving PAR2 and resulting in the induction of inflammation and proliferation of hVSMC. Thus, our in vitro data might extend the current view of sDPP4 action and shed light on cardiovascular effects of DPP4-inhibitors.
Collapse
Affiliation(s)
- Nina Wronkowitz
- Paul-Langerhans-Group for Integrative Physiology, German Diabetes Center, 40225 Düsseldorf, Germany
| | - Sven W Görgens
- Paul-Langerhans-Group for Integrative Physiology, German Diabetes Center, 40225 Düsseldorf, Germany
| | - Tania Romacho
- Paul-Langerhans-Group for Integrative Physiology, German Diabetes Center, 40225 Düsseldorf, Germany
| | - Laura A Villalobos
- Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Carlos F Sánchez-Ferrer
- Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Concepción Peiró
- Department of Pharmacology and Therapeutics, School of Medicine, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Henrike Sell
- Paul-Langerhans-Group for Integrative Physiology, German Diabetes Center, 40225 Düsseldorf, Germany.
| | - Jürgen Eckel
- Paul-Langerhans-Group for Integrative Physiology, German Diabetes Center, 40225 Düsseldorf, Germany
| |
Collapse
|
13
|
Proteinase-activated receptor-2 agonist activates anti-influenza mechanisms and modulates IFNγ-induced antiviral pathways in human neutrophils. BIOMED RESEARCH INTERNATIONAL 2013; 2013:879080. [PMID: 24171176 PMCID: PMC3793289 DOI: 10.1155/2013/879080] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 08/12/2013] [Indexed: 01/31/2023]
Abstract
Proteinase-activated receptor-2 (PAR2) is expressed by human leukocytes and participates in the development of inflammatory diseases. Recent studies demonstrated an ability of PAR2 agonist to enhance IFNγ-induced antiviral responses of human leukocytes. However, the precise cellular antiviral defense mechanisms triggered in leukocytes after stimulation with IFNγ and/or PAR2 agonist remain elusive. Therefore, we aimed to identify neutrophil defense mechanisms involved in antiviral resistance. Here we demonstrated that PAR2 agonist enhanced IFNγ-related reduction of influenza A virus (IAV) replication in human neutrophils. PAR2-mediated decrease in IAV replication was associated with reduced NS-1 transcription. Moreover, PAR2-dependent neutrophil activation resulted in enhanced myeloperoxidase degranulation and extracellular myeloperoxidase disrupted IAV. The production of ROS was elevated in response to PAR2 activation. Interestingly, IFNγ did not influence both effects: PAR2 agonist-triggered myeloperoxidase (MPO) release and reactive oxygen species (ROS) production, which are known to limit IAV infections. In contrast, orthomyxovirus resistance gene A (MxA) protein expression was synergistically elevated through PAR2 agonist and IFNγ in neutrophils. Altogether, these findings emphasize two PAR2-controlled antiviral mechanisms that are independent of or modulated by IFNγ.
Collapse
|
14
|
Thirkettle S, Decock J, Arnold H, Pennington CJ, Jaworski DM, Edwards DR. Matrix metalloproteinase 8 (collagenase 2) induces the expression of interleukins 6 and 8 in breast cancer cells. J Biol Chem 2013; 288:16282-16294. [PMID: 23632023 PMCID: PMC3675567 DOI: 10.1074/jbc.m113.464230] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Matrix metalloproteinase 8 (MMP-8) is a tumor-suppressive protease that cleaves numerous substrates, including matrix proteins and chemokines. In particular, MMP-8 proteolytically activates IL-8 and, thereby, regulates neutrophil chemotaxis in vivo. We explored the effects of expression of either a WT or catalytically inactive (E198A) mutant version of MMP-8 in human breast cancer cell lines. Analysis of serum-free conditioned media from three breast cancer cell lines (MCF-7, SK-BR-3, and MDA-MB-231) expressing WT MMP-8 revealed elevated levels of IL-6 and IL-8. This increase was mirrored at the mRNA level and was dependent on MMP-8 catalytic activity. However, sustained expression of WT MMP-8 by breast cancer cells was non-permissive for long-term growth, as shown by reduced colony formation compared with cells expressing either control vector or E198A mutant MMP-8. In long-term culture of transfected MDA-MB-231 cells, expression of WT but not E198A mutant MMP-8 was lost, with IL-6 and IL-8 levels returning to base line. Rare clonal isolates of MDA-MB-231 cells expressing WT MMP-8 were generated, and these showed constitutively high levels of IL-6 and IL-8, although production of the interleukins was no longer dependent upon MMP-8 activity. These studies support a causal connection between MMP-8 activity and the IL-6/IL-8 network, with an acute response to MMP-8 involving induction of the proinflammatory mediators, which may in part serve to compensate for the deleterious effects of MMP-8 on breast cancer cell growth. This axis may be relevant to the recognized ability of MMP-8 to orchestrate the innate immune system in inflammation in vivo.
Collapse
Affiliation(s)
- Sally Thirkettle
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Julie Decock
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Hugh Arnold
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Caroline J Pennington
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom
| | - Diane M Jaworski
- Department of Neurological Sciences, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Dylan R Edwards
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, United Kingdom.
| |
Collapse
|
15
|
Eijkelkamp N, Linley J, Torres J, Bee L, Dickenson A, Gringhuis M, Minett M, Hong G, Lee E, Oh U, Ishikawa Y, Zwartkuis F, Cox J, Wood J. A role for Piezo2 in EPAC1-dependent mechanical allodynia. Nat Commun 2013; 4:1682. [PMID: 23575686 PMCID: PMC3644070 DOI: 10.1038/ncomms2673] [Citation(s) in RCA: 168] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 02/27/2013] [Indexed: 02/07/2023] Open
Abstract
Aberrant mechanosensation has an important role in different pain states. Here we show that Epac1 (cyclic AMP sensor) potentiation of Piezo2-mediated mechanotransduction contributes to mechanical allodynia. Dorsal root ganglia Epac1 mRNA levels increase during neuropathic pain, and nerve damage-induced allodynia is reduced in Epac1-/- mice. The Epac-selective cAMP analogue 8-pCPT sensitizes mechanically evoked currents in sensory neurons. Human Piezo2 produces large mechanically gated currents that are enhanced by the activation of the cAMP-sensor Epac1 or cytosolic calcium but are unaffected by protein kinase C or protein kinase A and depend on the integrity of the cytoskeleton. In vivo, 8-pCPT induces long-lasting allodynia that is prevented by the knockdown of Epac1 and attenuated by mouse Piezo2 knockdown. Piezo2 knockdown also enhanced thresholds for light touch. Finally, 8-pCPT sensitizes responses to innocuous mechanical stimuli without changing the electrical excitability of sensory fibres. These data indicate that the Epac1-Piezo2 axis has a role in the development of mechanical allodynia during neuropathic pain.
Collapse
Affiliation(s)
- N Eijkelkamp
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht 3584 EA, The Netherlands
- There authors shared first authorship
| | - J.E. Linley
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
- There authors shared first authorship
| | - J.M. Torres
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
- Department of Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Granada, Granada 18012, Spain
| | - L. Bee
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
- Research Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - A.H. Dickenson
- Research Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - M. Gringhuis
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - M.S. Minett
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - G.S. Hong
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
- Department of Molecular Medicine and Biopharmaceutical Sciences, World Class University Program, Seoul National University, Seoul 151-742, South Korea
| | - E. Lee
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
- Department of Molecular Medicine and Biopharmaceutical Sciences, World Class University Program, Seoul National University, Seoul 151-742, South Korea
| | - U. Oh
- Department of Molecular Medicine and Biopharmaceutical Sciences, World Class University Program, Seoul National University, Seoul 151-742, South Korea
| | - Y. Ishikawa
- Cardiovascular Research Institute, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - F.J. Zwartkuis
- Department of Physiological Chemistry, University Medical Center Utrecht, Center for Biomedical Genetics and Cancer Genomics Center, Utrecht 3584 CG, The Netherlands
| | - J.J. Cox
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - J.N. Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
- Department of Molecular Medicine and Biopharmaceutical Sciences, World Class University Program, Seoul National University, Seoul 151-742, South Korea
| |
Collapse
|
16
|
Lohman RJ, Cotterell AJ, Barry GD, Liu L, Suen JY, Vesey DA, Fairlie DP. An antagonist of human protease activated receptor-2 attenuates PAR2 signaling, macrophage activation, mast cell degranulation, and collagen-induced arthritis in rats. FASEB J 2012; 26:2877-87. [PMID: 22467762 DOI: 10.1096/fj.11-201004] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Multiple serine proteases exert proinflammatory actions by signaling through protease-activated receptor-2 (PAR2) on the cell surface. Although inhibitors of individual proteases are anti-inflammatory, we sought to discover whether the first potent antagonist of their common target PAR2 might be beneficial in treating chronic arthritis-like inflammatory disease. Using a fluorescence assay, a novel compound, GB88, was shown to antagonize PAR2-induced intracellular Ca(2+) release in human monocyte-derived macrophages, being 1000 times more potent than a control compound, ENMD-1068 (IC(50) 1.6 ± 0.5 μM vs. 1.2 ± 0.4 mM, respectively). In Wistar rats, GB88 was orally bioavailable (F=55%, T(max) 4 h, C(max) 1.7 μM, 10 mg/kg). GB88 inhibited the acute paw edema induced in Wistar rats by intraplantar λ-carrageenan or PAR2 agonists 2-furoyl-LIGRLO-NH(2) or mast cell β-tryptase, without inhibiting proteolytic activity of tryptase in vitro. In the chronic collagen-induced model of arthritis in rats, GB88 (10 mg/kg) was disease modifying and ameliorated pathological and histopathological changes (edema, pannus formation, synovial hyperplasia, collagen degradation, macrophage invasion, mast cell degranulation) compared to untreated arthritic controls. The results suggest that an orally active PAR2 antagonist is effective in treating chronic arthritis in rats through inhibiting macrophage infiltration, mast cell degranulation, and β-tryptase-PAR2 signaling in joint inflammation.
Collapse
Affiliation(s)
- Rink-Jan Lohman
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | | | | | | | | | | | | |
Collapse
|
17
|
Pan HY, Yano M, Kido H. Effects of inhibitors of Toll-like receptors, protease-activated receptor-2 signalings and trypsin on influenza A virus replication and upregulation of cellular factors in cardiomyocytes. THE JOURNAL OF MEDICAL INVESTIGATION 2011; 58:19-28. [PMID: 21372483 DOI: 10.2152/jmi.58.19] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Severe influenza sometimes causes myocarditis. We recently found that influenza A virus (IAV) infection induces various cellular factors, such as proinflammatory cytokines IL-6, IL-1β and TNF-α, matrix metalloproteinases (MMPs) and ectopic trypsin in mice hearts and in H9c2 cardiomyocytes. The induction of these cellular factors in turn promotes viral replication, myocardial inflammation and cellular damage through their intracellular signal transductions in cooperation with the IAV-induced Toll-like receptors (TLRs) and proteinase-activated receptor-2 (PAR-2) signalings, although the precise nature of these interactions remain obscure. By using specific inhibitors of TLRs and PAR-2 signalings and trypsin inhibitor aprotinin, we analyzed the role of TLR signaling and PAR-2 signaling in the IAV-induced pathological changes in cardiomyocytes. Inhibitors of TLR7/8-Myeloid Differentiation factor 88-nuclear factor-κB signaling and aprotinin effectively suppressed IAV-induced upregulation of proinflammatory cytokines, MMPs, trypsinogen and viral replication. Inhibitor of TLR3-Toll/interleukin-1 receptor domain-containing adaptor inducing interferons-dependent signaling predominantly suppressed the upregulation of interferon-β, a key intracellular host immune response factor. In contrast to the suppressive effect of trypsin inhibitor aprotinin on IAV replication, PAR-2 inhibitor FSY-NH(2), induced marginal upregulation of trypsinogen and subsequent stimulation of IAV replication.
Collapse
Affiliation(s)
- Hai-Yan Pan
- Division of Enzyme Chemistry, Institute for Enzyme Research, University of Tokushima, Tokushima, Japan
| | | | | |
Collapse
|
18
|
Shpacovitch VM, Feld M, Holzinger D, Kido M, Hollenberg MD, Levi-Schaffer F, Vergnolle N, Ludwig S, Roth J, Luger T, Steinhoff M. Role of proteinase-activated receptor-2 in anti-bacterial and immunomodulatory effects of interferon-γ on human neutrophils and monocytes. Immunology 2011; 133:329-39. [PMID: 21501162 DOI: 10.1111/j.1365-2567.2011.03443.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Recent studies show that proteinase-activated receptor-2 (PAR(2)) contributes to the development of inflammatory responses. However, investigations into the precise role of PAR(2) activation in the anti-microbial defence of human leucocytes are just beginning. We therefore evaluated the contribution of PAR(2) to the anti-microbial response of isolated human innate immune cells. We found that PAR(2) agonist, acting alone, enhances phagocytosis of Staphylococcus aureus and killing of Escherichia coli by human leucocytes, and that the magnitude of the effect is similar to that of interferon-γ (IFN-γ). However, co-application of PAR(2) -cAP and IFN-γ did not enhance the phagocytic and bacteria-killing activity of leucocytes beyond that triggered by either agonist alone. On the other hand, IFN-γ enhances PAR(2) agonist-induced monocyte chemoattractant protein 1 (MCP-1) secretion by human neutrophils and monocytes. Furthermore, phosphoinositide-3 kinase and janus kinase molecules are involved in the synergistic effect of PAR(2) agonist and IFN-γ on MCP-1 secretion. Our findings suggest a potentially protective role of PAR(2) agonists in the anti-microbial defence established by human monocytes and neutrophils.
Collapse
Affiliation(s)
- Victoria M Shpacovitch
- Department of Dermatology, IZKF Münster, Boltzmann Institute for Immunobiology of the Skin, University of Münster, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Cevikbas F, Seeliger S, Fastrich M, Hinte H, Metze D, Kempkes C, Homey B, Steinhoff M. Role of protease-activated receptors in human skin fibrosis and scleroderma. Exp Dermatol 2010; 20:69-71. [DOI: 10.1111/j.1600-0625.2010.01184.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
20
|
Hyun E, Andrade-Gordon P, Steinhoff M, Beck PL, Vergnolle N. Contribution of bone marrow-derived cells to the pro-inflammatory effects of protease-activated receptor-2 in colitis. Inflamm Res 2010; 59:699-709. [PMID: 20339899 PMCID: PMC2917702 DOI: 10.1007/s00011-010-0181-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Revised: 01/18/2010] [Accepted: 02/15/2010] [Indexed: 12/30/2022] Open
Abstract
Objective Our aim was to determine the contribution of proteinase-activated receptor-2 (PAR2)-expressing bone marrow-derived cells on the development of colonic inflammation. Materials Chimeric mice were generated by injecting bone marrow cells from wildtype (PAR2+/+) or PAR2 knockout mice (PAR2−/−) into irradiated PAR2+/+ or PAR2−/− mice. Treatments: Colitis was induced by giving 2.5% dextran sodium sulfate (DSS) solution for 7 days or by a single intracolonic administration of trinitrobenzene sulphonic acid (TNBS, 2 mg dissolved in 40% ethanol). Methods Seven days after the induction of colitis, bowel thickness, inflammatory parameters [myeloperoxidase (MPO) activity, macroscopic/microscopic damage scores], and leukocyte trafficking (visualized via intravital microscopy) were assessed. Results Total deficiency of PAR2 resulted in a marked reduction in severity of both TNBS and DSS induced colitis as assessed by MPO activity, macroscopic damage, bowel thickness, and leukocyte adherence. Colitis was attenuated in all chimeric lines in which there was loss of PAR2 in the host, non-bone marrow-derived tissue, independent of the status of PAR expression by bone marrow-derived cells. Interestingly, TNBS colitis was attenuated in PAR2+/+ chimeric mice with PAR2−/− derived bone marrow but these animals were not protected from DSS colitis. Conclusions Expression of PAR2 by host-derived tissues plays a dominant role in regulating colonic inflammation. PAR2 expression by bone marrow-derived cells appears to play a role in TNBS colitis but not in DSS induced injury. Electronic supplementary material The online version of this article (doi:10.1007/s00011-010-0181-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Eric Hyun
- Department of Physiology and Pharmacology, University of Calgary, 3330 Hospital Drive, Calgary, AB T2N 4N1 Canada
| | - Patricia Andrade-Gordon
- Johnson & Johnson Pharmaceutical Research & Development, Welsh Rd and Mckean Rd, Spring House, PA 19477 USA
| | - Martin Steinhoff
- Department of Dermatology, Interdisciplinary Center for Clinical Research, University of Munster, Von-Esmarch-Strasse 58, 48149 Münster, Germany
| | - Paul L. Beck
- Department of Physiology and Pharmacology, University of Calgary, 3330 Hospital Drive, Calgary, AB T2N 4N1 Canada
| | - Nathalie Vergnolle
- Department of Physiology and Pharmacology, University of Calgary, 3330 Hospital Drive, Calgary, AB T2N 4N1 Canada
- Centre de Physiopathologie de Toulouse Purpan, CHU Purpan, INSERM U563, 31000 Toulouse, France
- Université Toulouse III Paul Sabatier, route de Narbonne, 31000 Toulouse, France
- CHU Purpan, INSERM U563, BP 3028, 31024 Toulouse Cedex, France
| |
Collapse
|
21
|
St-Onge M, Lagarde S, Laflamme C, Rollet-Labelle E, Marois L, Naccache PH, Pouliot M. Proteinase-activated receptor-2 up-regulation by Fcgamma-receptor activation in human neutrophils. FASEB J 2010; 24:2116-25. [PMID: 20154268 DOI: 10.1096/fj.09-146167] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We shed new light on the expression and function of the proteinase-activated receptor (PAR) family, associated with inflammation and hyperalgesia, in human granulocytes. Resting cells expressed constitutive levels of PAR-2 and PAR-3 mRNA but not PAR-1 or PAR-4. Based on flow cytometry, stimulation with opsonized bacteria (Bop) specifically up-regulated cell surface expression of PAR-2 in a concentration-dependent and time-dependent manner, independent of transcription or de novo protein synthesis. Primary granules were identified as a source of preformed PAR-2 that can readily be mobilized at the surface on fusion with the plasma membrane. Cellular response to PAR-2 activation, measured as changes in intracellular calcium concentration, was enhanced in PAR-2 up-regulated cells. Increase of cell-surface PAR-2 and of cell responsiveness were dependent specifically on the engagement of immunoglobulin (Ig)-binding receptors. Together, our results reveal that mobilization of intracellular granules, in response to Ig-receptor activation, up-regulates PAR-2 surface expression and makes neutrophils more responsive to proteinase activity. This enhanced response to PAR-2 activation indicates that molecular communication between pain and inflammation may be more important than previously believed.
Collapse
Affiliation(s)
- Mireille St-Onge
- Centre de Recherche en Rhumatologie et Immunologie du Centre Hospitalier Universitaire de Québec, Department of Microbiology-Infectiology and Immunology, Faculty of Medicine, Laval University, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
22
|
Peripheral relays in stress-induced activation of visceral afferents in the gut. Auton Neurosci 2010; 153:99-105. [DOI: 10.1016/j.autneu.2009.07.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2009] [Revised: 07/06/2009] [Accepted: 07/07/2009] [Indexed: 12/23/2022]
|
23
|
McIntosh K, Cunningham MR, Cadalbert L, Lockhart J, Boyd G, Ferrell W, Plevin R. Proteinase-activated receptor-2 mediated inhibition of TNFalpha-stimulated JNK activation - A novel paradigm for G(q/11) linked GPCRs. Cell Signal 2010; 22:265-73. [PMID: 19781631 PMCID: PMC2806525 DOI: 10.1016/j.cellsig.2009.09.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2009] [Accepted: 09/14/2009] [Indexed: 12/14/2022]
Abstract
In this study we examined the potential for PAR(2) and TNFalpha to synergise at the level of MAP kinase signalling in PAR(2) expressing NCTC2544 cells. However, to our surprise we found that activation of PAR(2) by trypsin or the specific activating peptide SLIGKV-OH strongly inhibited both the phosphorylation and activity of JNK. In contrast neither p38 MAP kinase nor ERK activation was affected although TNFalpha stimulated IkappaBalpha loss was partially reversed. The inhibitory effect was not observed in parental cells nor in cells expressing PAR(4), however inhibition was reversed by pre-incubation with the novel PAR(2) antagonist K14585, suggesting that the effect is specific for PAR(2) activation. SLIGKV-OH was found to be more potent in inhibiting TNFalpha-induced JNK activation than in stimulating JNK alone, suggesting agonist-directed signalling. The PKC activator PMA, also mimicked the inhibitory effect of SLIGKV-OH, and the effects of both agents were reversed by pre-treatment with the PKC inhibitor, GF109203X. Furthermore, incubation with the novel G(q/11) inhibitor YM25480 also reversed PAR(2) mediated inhibition. Activation of PAR(2) was found to disrupt TNFR1 binding to RIP and TRADD and this was reversed by both GF109203X and YM25480. A similar mode of inhibition observed in HUVECs through PAR(2) or P2Y2 receptors demonstrates the potential of a novel paradigm for GPCRs linked to G(q/11), in mediating inhibition of TNFalpha-stimulated JNK activation. This has important implications in assessing the role of GPCRs in inflammation and other conditions.
Collapse
Key Words
- par-2, proteinase-activated receptor-2
- par-2 ap, par-2 activating peptide
- 2f-ligkv-oh, 2-furoyl-ligkv-hydroxyl
- jnk, c-jun n-terminal protein kinase
- nfκb, nuclear factor kappa b
- pkc, protein kinase c
- map kinase, mitogen-activated protein kinase
- pma, phorbol 12-myristate 13-acetate
- tnfα, tumour necrosis factor-alpha, tnfr1, tnf-receptor-1
- gpcr, g-protein-coupled receptors
- rip, receptor interacting protein
- tradd, tnf receptor activated death domain
- traf, tnf receptor activating factor
- nhek, normal human epithelial keratinocytes
- gf109203x, 3-[1-[3-(dimethylamino)propyl]-1h-indol-3-yl]-4-(1h-indol-3-yl)-1h-pyrrole-2,5-dione monohydrochloride
- fadd, fas-associated death domain
- madd, map kinase activating death domain protein
- il-6, interleukin-6
- proteinase-activated receptor 2
- c-jun n-terminal protein kinase
- tnfα
- protein kinase c
Collapse
Affiliation(s)
- Kathryn McIntosh
- Department of Physiology and Pharmacology, University of Strathclyde, Strathclyde Institute for Pharmacy and Biomedical Sciences, 27 Taylor Street, Glasgow, G4 0NR, Scotland, UK
- School of Engineering and Science, University of The West of Scotland, Paisley PA1 2BE, UK
| | - Margaret R. Cunningham
- Department of Physiology and Pharmacology, University of Strathclyde, Strathclyde Institute for Pharmacy and Biomedical Sciences, 27 Taylor Street, Glasgow, G4 0NR, Scotland, UK
| | - Laurence Cadalbert
- Department of Physiology and Pharmacology, University of Strathclyde, Strathclyde Institute for Pharmacy and Biomedical Sciences, 27 Taylor Street, Glasgow, G4 0NR, Scotland, UK
| | - John Lockhart
- School of Engineering and Science, University of The West of Scotland, Paisley PA1 2BE, UK
| | - Gary Boyd
- School of Engineering and Science, University of The West of Scotland, Paisley PA1 2BE, UK
| | - W.R. Ferrell
- Centre for Rheumatic Disease, Royal Infirmary, Glasgow G31 2ER, UK
| | - Robin Plevin
- Department of Physiology and Pharmacology, University of Strathclyde, Strathclyde Institute for Pharmacy and Biomedical Sciences, 27 Taylor Street, Glasgow, G4 0NR, Scotland, UK
| |
Collapse
|
24
|
Borensztajn K, von der Thüsen JH, Peppelenbosch MP, Spek CA. The coagulation factor Xa/protease activated receptor-2 axis in the progression of liver fibrosis: a multifaceted paradigm. J Cell Mol Med 2009; 14:143-53. [PMID: 19968736 PMCID: PMC3837617 DOI: 10.1111/j.1582-4934.2009.00980.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Hepatic fibrosis is a common response to virtually all forms of chronic liver injury independent of the etiologic agent. Despite the relatively large population of patients suffering from hepatic fibrosis and cirrhosis, no efficient and well-tolerated drugs are available for the treatment of this disorder. The lack of efficient treatment options is at least partly because the underlying cellular mechanisms leading to hepatic fibrosis are only partly understood. It is thus of pivotal importance to better understand the cellular processes contributing to the progression of hepatic fibrosis. Interestingly in this perspective, a common feature of fibrotic disease of various organs is the activation of the coagulation cascade and hepatic fibrosis is also accompanied by a local hypercoagulable state. Activated blood coagulation factors directly target liver cells by activating protease-activated receptors (PAR) thereby inducing a plethora of cellular responses like (among others) proliferation, migration and extracellular matrix production. Coagulation factor driven PAR activation thus establishes a potential link between activation of the coagulation cascade and the progression of fibrosis. The current review focuses on blood coagulation factor Xa and summarizes the variety of cellular functions induced by factor Xa-driven PAR-2 activation and the subsequent consequences for tissue repair and hepatic fibrosis.
Collapse
Affiliation(s)
- Keren Borensztajn
- Center for Experimental and Molecular Medicine, Academic Medical Center, Meibergdreef, Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
25
|
Pagano RL, Sampaio SC, Juliano MA, Juliano L, Giorgi R. Involvement of proteinase-activated receptors 1 and 2 in spreading and phagocytosis by murine adherent peritoneal cells: modulation by the C-terminal of S100A9 protein. Eur J Pharmacol 2009; 628:240-6. [PMID: 19941849 DOI: 10.1016/j.ejphar.2009.11.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Revised: 11/02/2009] [Accepted: 11/16/2009] [Indexed: 11/17/2022]
Abstract
Proteinase-activated receptors (PAR) are widely recognized for their modulatory properties in inflammatory and immune responses; however, their direct role on phagocyte effector functions remains unknown. S100A9, a protein secreted during inflammatory responses, deactivates activated peritoneal macrophages, and its C-terminal portion inhibits spreading and phagocytosis of adherent peritoneal cells. Herein, the effect of PAR1 and PAR2 agonists was investigated on spreading and phagocytosis by adherent peritoneal cells, as well as the ability of murine C-terminal of S100A9 peptide (mS100A9p) to modulate this effect. Adherent peritoneal cells obtained from mouse abdominal cavity were incubated with PAR1 and PAR2 agonists and spreading and phagocytosis of Candida albicans particles were evaluated. PAR1 agonists increased both the spreading and the phagocytic activity, but PAR2 agonists only increased the spreading index. mS100A9p reverted both the increased spreading and phagocytosis induced by PAR1 agonists, but no interference in the increased spreading induced by PAR2 agonists was noticed. The shorter homologue peptide to the C-terminal of mS100A9p, corresponding to the H(92)-E(97) region, also reverted the increased spreading and phagocytosis induced by PAR1 agonists. These findings show that proteinase-activated receptors have an important role for spreading and phagocytosis of adherent peritoneal cells, and that the peptide corresponding to the C-terminal of S100A9 protein is a remarkable candidate for use as a novel compound to modulate PAR1 function.
Collapse
Affiliation(s)
- Rosana L Pagano
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av Prof Lineu Prestes, São Paulo, SP, Brazil
| | | | | | | | | |
Collapse
|
26
|
Su X, Matthay MA. Role of protease activated receptor 2 in experimental acute lung injury and lung fibrosis. Anat Rec (Hoboken) 2009; 292:580-6. [PMID: 19226616 DOI: 10.1002/ar.20846] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Protease activated receptor 2 (PAR2) is widely-distributed (lung, liver, kidney, etc.) and expressed by variety of cells (i.e. leukocytes, epithelial cells, endothelial cells, and fibroblast). PAR2 may participate in many pathological processes, such as, inflammation, injury, as well as fibrosis. Therefore, in this study, we tested whether PAR2 would exert a role in acid-induced acute lung injury, E. coli pneumonia, bleomycin-induced acute lung injury and fibrosis. Acid, E. coli, or bleomycin were intratracheally instilled into the lungs of both wildtype and PAR2 knockout mice to detect differences in pulmonary edema, lung vascular permeability, lung fibrosis, and other parameters. Knockout of PAR2 did not affect the extent of pulmonary edema and lung vascular permeability in acid-induced acute lung injury. Also, both activation of PAR2 in the airspaces of the lung and deletion of PAR2 did not alter the magnitude of pulmonary edema and lung vascular permeability in E. coli pneumonia. Finally, PAR2 deficiency did not affect the severity of lung inflammation and lung fibrosis in bleomycin-induced acute lung injury and lung fibrosis models. Thus, PAR2 does not appear to play a critical role in the pathogeneses of experimental acid-induced acute lung injury, E. coli pneumonia, and bleomycin-induced acute lung injury and pulmonary fibrosis in mice.
Collapse
Affiliation(s)
- Xiao Su
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143-0130, USA.
| | | |
Collapse
|
27
|
Lin CB, Chen N, Scarpa R, Guan F, Babiarz-Magee L, Liebel F, Li WH, Kizoulis M, Shapiro S, Seiberg M. LIGR, a protease-activated receptor-2-derived peptide, enhances skin pigmentation without inducing inflammatory processes. Pigment Cell Melanoma Res 2008; 21:172-83. [PMID: 18426410 DOI: 10.1111/j.1755-148x.2008.00441.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The protease-activated receptor-2 (PAR-2) is a seven transmembrane G-protein-coupled receptor that could be activated by serine protease cleavage or by synthetic peptide agonists. We showed earlier that activation of PAR-2 with Ser-Leu-Ile-Gly-Arg-Leu-NH(2) (SLIGRL), a known PAR-2 activating peptide, induces keratinocyte phagocytosis and increases skin pigmentation, indicating that PAR-2 regulates pigmentation by controlling phagocytosis of melanosomes. Here, we show that Leu-Ile-Gly-Arg-NH(2) (LIGR) can also induce skin pigmentation. Both SLIGRL and LIGR increased melanin deposition in vitro and in vivo, and visibly darkened human skins grafted onto severe combined immuno-deficient (SCID) mice. Both SLIGRL and LIGR stimulated Rho-GTP activation resulting in keratinocyte phagocytosis. Interestingly, LIGR activates only a subset of the PAR-2 signaling pathways, and unlike SLIGRL, it does not induce inflammatory processes. LIGR did not affect many PAR-2 signaling pathways, including [Ca(2+)] mobilization, cAMP induction, the induction of cyclooxgenase-2 (COX-2) expression and the secretion of prostaglandin E2, interleukin-6 and -8. PAR-2 siRNA inhibited LIGR-induced phagocytosis, indicating that LIGR signals via PAR-2. Our data suggest that LIGR is a more specific regulator of PAR-2-induced pigmentation relative to SLIGRL. Therefore, enhancing skin pigmentation by topical applications of LIGR may result in a desired tanned-like skin color, without enhancing inflammatory processes, and without the need of UV exposure.
Collapse
Affiliation(s)
- Connie B Lin
- The Johnson & Johnson Skin Research Center, Consumer Product Worldwide, A division of Johnson & Johnson Consumer Companies, Inc., 199 Grandview Rd., Skillman, NJ 08558, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Feld M, Shpacovitch VM, Ehrhardt C, Kerkhoff C, Hollenberg MD, Vergnolle N, Ludwig S, Steinhoff M. Agonists of proteinase-activated receptor-2 enhance IFN-gamma-inducible effects on human monocytes: role in influenza A infection. THE JOURNAL OF IMMUNOLOGY 2008; 180:6903-10. [PMID: 18453611 DOI: 10.4049/jimmunol.180.10.6903] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Proteinase-activated receptor-2 (PAR(2)) is expressed by different types of human leukocytes and involved in the development of inflammatory and infectious diseases. However, its precise role in the regulation of human monocyte and macrophage function during viral infection remains unclear. Also, the ability of PAR(2) agonists to enhance the effects induced by immune mediators during infection or inflammation is still poorly investigated. Therefore, we investigated the ability of a PAR(2) agonist to enhance IFN-gamma-induced suppression of influenza A virus replication in human monocytes. We found that this effect correlates with an increased abundance of IkappaBalpha after costimulation of cells with PAR(2) agonist and IFN-gamma. Remarkably, coapplication of PAR(2) agonist and IFN-gamma also enhances the effects of IFN-gamma on IFN-gamma-inducible protein 10 kDa release, and CD64 and alphaVbeta3 surface expression by human monocytes. Together, these findings indicate a potentially protective role of PAR(2) activation during the progression of influenza A virus infection. This effect could be associated with the ability of PAR(2) agonists to enhance IFN-gamma-induced protective effects on human monocytes.
Collapse
Affiliation(s)
- Micha Feld
- Department of Dermatology and Ludwig-Boltzmann-Institute for Immunobiology of the Skin, University of Münster, Münster, Germany
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Migration, cell-cell interaction and adhesion in the immune system. ERNST SCHERING FOUNDATION SYMPOSIUM PROCEEDINGS 2008:97-137. [PMID: 18510101 DOI: 10.1007/2789_2007_062] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Migration is an essential function of immune cells. It is necessary to lead immune cell precursors from their site of generation to the places of maturation or function. Cells of the adaptive immune system also need to interact physically with each other or with specialized antigen presenting cells in lymphatic tissues in order to become activated. Thereby a complex series of controlled migration events, adhesive interactions and signalling responses is induced. Finally cells must be able to leave the activating tissues and re-enter the bloodstream from which they extravasate into inflamed tissue sites. Cells of the innate immune system can function directly without the need for previous activation. However, these cells have to adapt their function to a panoply of pathogens and environmental niches which can be invaded. The current review highlights the central aspects of cellular dynamics underlying adaptive and innate cellular immunity. Thereby a focus will be put on recent results obtained by microscopic observation of live cells in vitro or by intravital 2-photon microscopy in live animals.
Collapse
|
30
|
Shpacovitch VM, Seeliger S, Huber-Lang M, Balkow S, Feld M, Hollenberg MD, Sarma VJ, Ward PA, Strey A, Gerke V, Sommerhoff CP, Vergnolle N, Steinhoff M. Agonists of proteinase-activated receptor-2 affect transendothelial migration and apoptosis of human neutrophils. Exp Dermatol 2007; 16:799-806. [PMID: 17845211 DOI: 10.1111/j.1600-0625.2007.00605.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Skin is the first barrier preventing microorganism invasion in host. Wounds destroy this defense barrier and, without an appropriate care, may lead to sepsis. Neutrophil activation and immigration plays an important role at the inflammatory stage of wound healing. Neutrophils are known to express proteinase-activated receptors (PARs), which can be activated by serine proteases, also by enzymes involved in wound healing. We previously reported that PAR(2) agonists up-regulate cell adhesion molecule expression and cytokine production by human neutrophils. Here, we demonstrate that PAR(2) agonists (serine proteases as well as synthetic peptides) reduce transendothelial migration of neutrophils and prolong their life in vitro. Synthetic PAR(2) agonist also enhanced protective interferon (IFN)gamma-induced FcgammaRI expression at neutrophil cell surface. Of note, IFNgamma is a cytokine, which was used in clinical trials to reactivate human neutrophil functions during sepsis. Moreover, we observed a significant increase of PAR(2) expression on cell surface of neutrophils from septic patients as compared with healthy volunteers. Together, our results indicate that PAR(2) may be involved in the pathophysiology of neutrophil-endothelial interactions during wound healing or later during sepsis in humans, potentially by affecting neutrophil apoptosis, transendothelial migration and Fcgamma receptor-mediated phagocytosis.
Collapse
Affiliation(s)
- Victoria M Shpacovitch
- Department of Dermatology, IZKF Münster, and Boltzmann Institute for Immunobiology of the Skin, University of Münster, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Proteinases and signalling: pathophysiological and therapeutic implications via PARs and more. Br J Pharmacol 2007; 153 Suppl 1:S263-82. [PMID: 18059329 DOI: 10.1038/sj.bjp.0707507] [Citation(s) in RCA: 210] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Proteinases like thrombin, trypsin and tissue kallikreins are now known to regulate cell signaling by cleaving and activating a novel family of G-protein-coupled proteinase-activated receptors (PARs 1-4) via exposure of a tethered receptor-triggering ligand. On their own, short synthetic PAR-selective PAR-activating peptides (PAR-APs) mimicking the tethered ligand sequences can activate PARs 1, 2 and 4 and cause physiological responses both in vitro and in vivo. Using the PAR-APs as sentinel probes in vivo, it has been found that PAR activation can affect the vascular, renal, respiratory, gastrointestinal, musculoskeletal and nervous systems (both central and peripheral nervous system) and can promote cancer metastasis and invasion. In general, responses triggered by PARs 1, 2 and 4 are in keeping with an innate immune inflammatory response, ranging from vasodilatation to intestinal inflammation, increased cytokine production and increased or decreased nociception. Further, PARs have been implicated in a number of disease states, including cancer and inflammation of the cardiovascular, respiratory, musculoskeletal, gastrointestinal and nervous systems. In addition to activating PARs, proteinases can cause hormone-like effects by other signalling mechanisms, like growth factor receptor activation, that may be as important as the activation of PARs. We, therefore, propose that the PARs themselves, their activating serine proteinases and their associated signalling pathways can be considered as attractive targets for therapeutic drug development. Thus, proteinases in general must now be considered as 'hormone-like' messengers that can signal either via PARs or other mechanisms.
Collapse
|
32
|
Shpacovitch V, Feld M, Bunnett NW, Steinhoff M. Protease-activated receptors: novel PARtners in innate immunity. Trends Immunol 2007; 28:541-50. [PMID: 17977790 DOI: 10.1016/j.it.2007.09.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2007] [Revised: 09/08/2007] [Accepted: 09/19/2007] [Indexed: 11/17/2022]
Abstract
Protease-activated receptors (PARs) belong to a family of G protein-coupled receptors activated by serine proteases via proteolytic cleavage. PARs are expressed on epithelial cells, endothelial cells, and leukocytes, indicating a role in controlling barrier function against external danger. During inflammation, microorganisms as well as host immune cells release various proteases activating PARs. Thus, PARs can be viewed as an integral component of the host antimicrobial alarm system. When stimulated, PARs regulate various functions of leukocytes in vivo and in vitro, revealing a novel pathway by which proteases affect innate immune responses. Understanding protease-immune interactions could lead to novel strategies for the treatment of infectious and immune-related diseases.
Collapse
Affiliation(s)
- V Shpacovitch
- Department of Dermatology and Ludwig Boltzmann Institute for Cell Biology of the Skin, University of Münster, D-48149 Münster, Germany
| | | | | | | |
Collapse
|
33
|
Rudack C, Steinhoff M, Mooren F, Buddenkotte J, Becker K, von Eiff C, Sachse F. PAR-2 activation regulates IL-8 and GRO-alpha synthesis by NF-kappaB, but not RANTES, IL-6, eotaxin or TARC expression in nasal epithelium. Clin Exp Allergy 2007; 37:1009-22. [PMID: 17581194 DOI: 10.1111/j.1365-2222.2007.02686.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND The effects of protease-activated receptor-2 (PAR-2) stimulation on inflammation mechanisms of chronic rhinosinusitis (CRS) are still unknown. METHODS PAR-2 receptor expression was investigated by immunohistochemistry and Taqman mRNA analysis in the mucosa of different rhinosinusitis entities. In primary nasal epithelial cell cultures, the function of PAR-2 and its ability to produce CXC, CC chemokines, and IL-6 were measured by calcium mobilization and stimulation tests. Inhibition tests were performed using cortisone, serine protease inhibitors, cysteine protease inhibitors, Pertussis toxin (PTX) and nuclear transcription factor (NF-kappaB) inhibition (BAY 11-7085). Signal transduction pathways were analysed by electromobility shift assays (EMSA) and NF-kappaB binding studies. RESULTS The expression of PAR-2 was found to be increased in CRS specimens. The activation of PAR by trypsin or PAR-2-specific activating peptide (AP) caused an increase in cytosolic calcium, as well as the release of the CXC chemokines IL-8 and growth-related oncogene (GRO)-alpha, but not the release of CC chemokines or IL-6. AP-induced CXC chemokine was sensitive to PTX and activation of NF-kappaB was inhibited by BAY11-7085. Furthermore, a serine protease inhibitor significantly inhibited chemokine synthesis stimulated by trypsin and culture supernatants of staphylococci, whereas steroids and cysteine protease inhibitors had little effect. CONCLUSION PAR-2 plays a role in serine protease-mediated regulation - staphylococcal and non-staphylococcal origin - of IL-8 and GRO-alpha in nasal epithelial cells, but not in the regulation of CC chemokines. PAR-2 may therefore be involved in the pathophysiology of CRS and NP at different sites of activation, namely (i) proteases, (ii) the PAR-2 receptor itself or (iii) the application of novel agents that block NF-kappaB/IkappaB-alpha signalling.
Collapse
Affiliation(s)
- C Rudack
- Department of Otorhinolaryngology, Head and Neck Surgery, IZK Münster and Boltzmann Institute for Immunobiology of the Skin, Münster, Germany.
| | | | | | | | | | | | | |
Collapse
|
34
|
Cottrell GS, Amadesi S, Pikios S, Camerer E, Willardsen JA, Murphy BR, Caughey GH, Wolters PJ, Coughlin SR, Peterson A, Knecht W, Pothoulakis C, Bunnett NW, Grady EF. Protease-activated receptor 2, dipeptidyl peptidase I, and proteases mediate Clostridium difficile toxin A enteritis. Gastroenterology 2007; 132:2422-37. [PMID: 17570216 PMCID: PMC2366898 DOI: 10.1053/j.gastro.2007.03.101] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2006] [Accepted: 03/15/2007] [Indexed: 12/23/2022]
Abstract
BACKGROUND & AIMS We studied the role of protease-activated receptor 2 (PAR(2)) and its activating enzymes, trypsins and tryptase, in Clostridium difficile toxin A (TxA)-induced enteritis. METHODS We injected TxA into ileal loops in PAR(2) or dipeptidyl peptidase I (DPPI) knockout mice or in wild-type mice pretreated with tryptase inhibitors (FUT-175 or MPI-0442352) or soybean trypsin inhibitor. We examined the effect of TxA on expression and activity of PAR(2) and trypsin IV messenger RNA in the ileum and cultured colonocytes. We injected activating peptide (AP), trypsins, tryptase, and p23 in wild-type mice, some pretreated with the neurokinin 1 receptor antagonist SR140333. RESULTS TxA increased fluid secretion, myeloperoxidase activity in fluid and tissue, and histologic damage. PAR(2) deletion decreased TxA-induced ileitis, reduced luminal fluid secretion by 20%, decreased tissue and fluid myeloperoxidase by 50%, and diminished epithelial damage, edema, and neutrophil infiltration. DPPI deletion reduced secretion by 20% and fluid myeloperoxidase by 55%. In wild-type mice, FUT-175 or MPI-0442352 inhibited secretion by 24%-28% and tissue and fluid myeloperoxidase by 31%-71%. Soybean trypsin inhibitor reduced secretion to background levels and tissue myeloperoxidase by up to 50%. TxA increased expression of PAR(2) and trypsin IV in enterocytes and colonocytes and caused a 2-fold increase in Ca(2+) responses to PAR(2) AP. AP, tryptase, and trypsin isozymes (trypsin I/II, trypsin IV, p23) caused ileitis. SR140333 prevented AP-induced ileitis. CONCLUSIONS PAR(2) and its activators are proinflammatory in TxA-induced enteritis. TxA stimulates existing PAR(2) and up-regulates PAR(2) and activating proteases, and PAR(2) causes inflammation by neurogenic mechanisms.
Collapse
Affiliation(s)
- Graeme S Cottrell
- Center for the Neurobiology of Digestive Disease, Department of Surgery, University of California, San Francisco, San Francisco, California 94143-0660, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Busso N, Frasnelli M, Feifel R, Cenni B, Steinhoff M, Hamilton J, So A. Evaluation of protease-activated receptor 2 in murine models of arthritis. ACTA ACUST UNITED AC 2007; 56:101-7. [PMID: 17195212 DOI: 10.1002/art.22312] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Protease-activated receptor 2 (PAR-2) activation has been linked to pro- and antiinflammatory cellular responses. We undertook this study to explore the importance of PAR-2 activation in 4 murine models of arthritis and to analyze the expression of PAR-2 in human arthritic synovium. METHODS Zymosan-induced arthritis (ZIA), K/BxN serum-induced arthritis, and Freund's complete adjuvant (CFA)-induced arthritis were generated in naive PAR-2(-/-) mice and PAR-2(+/+) littermates. Antigen-induced arthritis (AIA) was generated in immunized mice using methylated bovine serum albumin (mBSA). The severity of arthritis was assessed by clinical scoring, technetium uptake measurement, and histologic analysis. Immune responses to mBSA were also evaluated from AIA. The expression of PAR-2 in synovial tissues from rheumatoid arthritis (RA) and osteoarthritis (OA) patients was compared. RESULTS In AIA, arthritis was significantly decreased in PAR-2-deficient mice and was associated with decreased levels of anti-mBSA IgG antibodies and lymph node cell proliferation. No difference in arthritis severity was seen in mice with ZIA, K/BxN serum-induced arthritis, and CFA-induced arthritis. Synovial biopsy specimens from RA patients demonstrated significantly increased expression of PAR-2 compared with those from OA patients. CONCLUSION PAR-2 deficiency was found to modulate articular inflammation in murine models of arthritis that require prior immunization and was associated with reduced levels of anti-mBSA IgG and lymph node cell proliferation in AIA. Expression of PAR-2 in RA synovium was significantly higher than that in OA synovium, and this suggests that PAR-2 is implicated in the pathogenesis of immune-mediated forms of arthritis.
Collapse
MESH Headings
- Animals
- Arthritis, Experimental/immunology
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/pathology
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Fluorescent Antibody Technique, Direct
- Humans
- Immunoenzyme Techniques
- Mice
- Mice, Knockout
- Osteoarthritis/metabolism
- Osteoarthritis/pathology
- Receptor, PAR-2/deficiency
- Receptor, PAR-2/genetics
- Receptor, PAR-2/immunology
- Synovial Membrane/metabolism
- Synovial Membrane/pathology
Collapse
Affiliation(s)
- Nathalie Busso
- Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
36
|
Behnsen J, Narang P, Hasenberg M, Gunzer F, Bilitewski U, Klippel N, Rohde M, Brock M, Brakhage AA, Gunzer M. Environmental dimensionality controls the interaction of phagocytes with the pathogenic fungi Aspergillus fumigatus and Candida albicans. PLoS Pathog 2007; 3:e13. [PMID: 17274685 PMCID: PMC1790725 DOI: 10.1371/journal.ppat.0030013] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2006] [Accepted: 12/19/2006] [Indexed: 11/19/2022] Open
Abstract
The fungal pathogens Aspergillus fumigatus and Candida albicans are major health threats for immune-compromised patients. Normally, macrophages and neutrophil granulocytes phagocytose inhaled Aspergillus conidia in the two-dimensional (2-D) environment of the alveolar lumen or Candida growing in tissue microabscesses, which are composed of a three-dimensional (3-D) extracellular matrix. However, neither the cellular dynamics, the per-cell efficiency, the outcome of this interaction, nor the environmental impact on this process are known. Live imaging shows that the interaction of phagocytes with Aspergillus or Candida in 2-D liquid cultures or 3-D collagen environments is a dynamic process that includes phagocytosis, dragging, or the mere touching of fungal elements. Neutrophils and alveolar macrophages efficiently phagocytosed or dragged Aspergillus conidia in 2-D, while in 3-D their function was severely impaired. The reverse was found for phagocytosis of Candida. The phagocytosis rate was very low in 2-D, while in 3-D most neutrophils internalized multiple yeasts. In competitive assays, neutrophils primarily incorporated Aspergillus conidia in 2-D and Candida yeasts in 3-D despite frequent touching of the other pathogen. Thus, phagocytes show activity best in the environment where a pathogen is naturally encountered. This could explain why "delocalized" Aspergillus infections such as hematogeneous spread are almost uncontrollable diseases, even in immunocompetent individuals.
Collapse
Affiliation(s)
- Judith Behnsen
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Priyanka Narang
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Mike Hasenberg
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Frank Gunzer
- Department of Physics, The German University of Cairo, New Cairo City, Egypt
| | | | - Nina Klippel
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Manfred Rohde
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Matthias Brock
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Axel A Brakhage
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Matthias Gunzer
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
37
|
Gera L, Varga R, Török L, Kaszaki J, Szabó A, Nagy K, Boros M. Beneficial effects of phosphatidylcholine during hindlimb reperfusion. J Surg Res 2007; 139:45-50. [PMID: 17275842 DOI: 10.1016/j.jss.2006.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2006] [Revised: 07/21/2006] [Accepted: 08/07/2006] [Indexed: 11/28/2022]
Abstract
BACKGROUND Microcirculatory dysfunctions and mast cell (MC) reactions play important roles in hypoxic tissue injuries. The aims of this study were to characterize the effects of hindlimb ischemia-reperfusion (I-R) on the periosteal microcirculation and to define the consequences of systemic phosphatidylcholine (PC) therapy during this condition. MATERIALS AND METHODS Microcirculatory changes were visualized by means of fluorescence intravital videomicroscopy in anesthetized Wistar rats. There was 60 min of complete hindlimb ischemia followed by a 180-min reperfusion in the presence of PC treatment (50 mg/kg i.v.; in the second 10 min of reperfusion) or vehicle. Further two groups served as vehicle- or PC-treated sham-operated controls. The proportion of degranulated MCs and the leukocyte accumulation (myeloperoxidase, MPO assay) were determined in muscle biopsies. RESULTS I-R significantly increased the muscle MPO activity (from 14.94 to 63.45 mU/mg) and the proportion of degranulated MCs (to 82.5%). The periosteal capillary RBC velocity (RBCV) and the functional capillary density (FCD) had decreased, while the primary and secondary leukocyte-endothelial cell interactions had increased by the end of reperfusion (rolling from 20.8 to 40.0%, and firm adherence from 254 to 872 mm(-2)). PC treatment decreased the leukocyte rolling and sticking, preserved the FCD and improved the RBCV. The MC degranulation and MPO activity diminished significantly in the muscle layer. CONCLUSIONS PC administration improves I-R-induced periosteal microcirculatory dysfunctions and ameliorates secondary inflammatory reactions. Systemic PC treatment could offer a potential treatment modality during hypoperfusion or inflammatory conditions of the bones.
Collapse
Affiliation(s)
- László Gera
- Department of Traumatology, Kecskemét County Hospital, Kecskemét, Hungary
| | | | | | | | | | | | | |
Collapse
|
38
|
Seitz I, Hess S, Schulz H, Eckl R, Busch G, Montens HP, Brandl R, Seidl S, Schömig A, Ott I. Membrane-type serine protease-1/matriptase induces interleukin-6 and -8 in endothelial cells by activation of protease-activated receptor-2: potential implications in atherosclerosis. Arterioscler Thromb Vasc Biol 2007; 27:769-75. [PMID: 17255532 DOI: 10.1161/01.atv.0000258862.61067.14] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The serine protease MT-SP1/matriptase plays an important role in cell migration and matrix degradation. Hepatocyte growth factor (HGF), urokinase-type plasminogen activator (uPA), and protease-activated receptor 2 (PAR-2) have been identified as in vitro substrates of MT-SP1/matriptase. Because PAR-2 is expressed in endothelial cells and contributes to inflammatory processes, we sought to investigate the effects of MT-SP1/matriptase on endothelial cytokine expression and analyzed MT-SP1/matriptase expression in vascular cells and atherosclerotic lesions. METHODS AND RESULTS In endothelial cells, recombinant MT-SP1/matriptase dose-dependently induced interleukin (IL)-8 and IL-6 mRNA and protein expression dependent on its proteolytic activity. MT-SP1/matriptase time-dependently induced phosphorylation of p38 MAPK and p42/44 MAPK. Inhibitor experiments revealed that p38 MAPK and PKCalpha were necessary for IL-8 induction. PAR-2 downregulation abolished and PAR-2 overexpression augmented MT-SP1/matriptase-induced IL-8 expression as evidence for PAR-2 signaling. In human atherectomies, MT-SP1/matriptase was expressed in blood cells adherent to the endothelium. Concordantly, basal MT-SP1/matriptase expression was detected in isolated monocytes. Coincubation of monocytes and endothelial cells resulted in an increased IL-8 release, which was reduced after downregulation of endothelial PAR-2 and monocytic MT-SP1/matriptase. CONCLUSION MT-SP1/matriptase induces release of proinflammatory cytokines in endothelial cells through activation of PAR-2. MT-SP1/matriptase is expressed in monocytes, thus, interaction of monocytic MT-SP1/matriptase with endothelial PAR-2 may contribute to atherosclerosis.
Collapse
Affiliation(s)
- Isabell Seitz
- Deutsches Herzzentrum und 1. Medizinische Klinik, Technische Universität München, 80636 München, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Xiang Y, Masuko-Hongo K, Sekine T, Nakamura H, Yudoh K, Nishioka K, Kato T. Expression of proteinase-activated receptors (PAR)-2 in articular chondrocytes is modulated by IL-1beta, TNF-alpha and TGF-beta. Osteoarthritis Cartilage 2006; 14:1163-73. [PMID: 16757188 DOI: 10.1016/j.joca.2006.04.015] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2005] [Accepted: 04/18/2006] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To investigate the modulation of expression of proteinase-activated receptor-2 (PAR-2) in articular chondrocytes by inflammatory cytokines. DESIGN Articular synovium and cartilage tissues were collected from eight patients with osteoarthritis (OA), and three patients without arthropathy ("normal"). Chondrocytes were stimulated with interleukin (IL)-1beta, tumor necrosis factor (TNF)-alpha or transforming growth factor (TGF)-beta1. The expression of PAR-2 was detected using reverse transcriptase-polymerase chain reaction (PCR), Western blotting and immunofluorescence. Quantitative PCR was performed to assess the expression levels of PAR-2 messenger RNA (mRNA). RESULTS The expression of PAR-2 mRNA was demonstrated in both OA and normal chondrocytes as well as in synovial fibroblasts. However, the level of PAR-2 in OA chondrocytes was much higher than in normal chondrocytes. Long-term culture revealed that PAR-2 mRNA expression was maintained up to three passages in OA but not in normal chondrocytes. IL-1beta and TNF-alpha both upregulated PAR-2 expression in normal and OA chondrocytes. In contrast, TGF-beta1 significantly decreased expression of PAR-2 in OA chondrocytes but increased PAR-2 in normal chondrocytes. CONCLUSIONS Overexpression of PAR-2 in OA chondrocytes is upregulated by proinflammatory cytokines IL-1beta and TNF-alpha, and down-regulated by regulatory cytokine TGF-beta1. PAR-2 may be involved in the pathogenesis of OA.
Collapse
Affiliation(s)
- Y Xiang
- Department of Bioregulation and Proteomics, Institute of Medical Science, St. Marianna University School of Medicine, Kawasaki, Japan
| | | | | | | | | | | | | |
Collapse
|
40
|
Ramachandran R, Morice AH, Compton SJ. Proteinase-Activated Receptor2Agonists Upregulate Granulocyte Colony-Stimulating Factor, IL-8, and VCAM-1 Expression in Human Bronchial Fibroblasts. Am J Respir Cell Mol Biol 2006; 35:133-41. [PMID: 16498082 DOI: 10.1165/rcmb.2005-0362oc] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Proteinase-activated receptors (PARs) are a novel family of G-protein-coupled receptors. PAR2 has been implicated in inflammatory airways disease. Although fibroblasts are pathologically important in the airways, the proinflammatory role of PAR2 in these cells remains unknown. We assessed PAR expression and functionality in human primary bronchial fibroblasts (HPBFs) before assessing PAR2-mediated HPBF proliferation, cytokine production, and adhesion molecule expression. RT-PCR and flow cytometry demonstrated that HPBFs express hPAR1, hPAR2, and hPAR3, but not hPAR4. Intracellular calcium signaling in HPBFs in response to PAR agonists showed that only hPAR1 and hPAR2 were functional receptors. We used the MTT assay to assess HPBF proliferation. Of the PAR2 agonist proteinases or selective PAR2-activating peptides (PAR2-APs) tested, none stimulated HPBF proliferation, whereas thrombin was a HPBF growth factor. mRNA for IL-8 and granulocyte colony-stimulating factor (G-CSF) was upregulated after addition of SLIGKV-NH2 when assessed by RT-PCR. No significant increase in G-CSF or IL-8 protein was detected. Trypsin stimulated IL-8 and G-CSF release from HPBF in a time- and dose-dependent manner. Leupeptin and soya trypsin inhibitor abrogated trypsin-stimulated cytokine release, indicating a requirement for trypsin's proteolytic activity. Trypsin and SLIGKV-NH2 stimulated an increase in VCAM-1 expression at 12 h after treatment, which declined thereafter. PAR2-driven upregulation of VCAM-1 cell surface expression and the release of IL-8 and G-CSF from bronchial fibroblasts may be important in promoting neutrophilic airways inflammation.
Collapse
Affiliation(s)
- Rithwik Ramachandran
- Respiratory Medicine, Division of Academic Medicine, Post Graduate Medical Institute of the University of Hull in association with the Hull York Medical School, East Yorkshire, HU16 5JQ, United Kingdom
| | | | | |
Collapse
|
41
|
Chen LW, Huang HL, Lee IT, Hsu CM, Lu PJ. THERMAL INJURY-INDUCED PRIMING EFFECT OF NEUTROPHIL IS TNF-α AND P38 DEPENDENT. Shock 2006; 26:69-76. [PMID: 16783201 DOI: 10.1097/01.shk0000209531.38188.18] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Priming response of neutrophil in clinical-related conditions and its mechanism has not been clarified. This study is to determine if thermal injury-induced priming effect of neutrophil is TNF-alpha and p38 dependent. In Experiment 1, bone marrow neutrophil of wild-type (WT) mice and TNF receptor superfamily, member 1A (Tnfrsf1a-/-) mice were harvested and treated with TNF-alpha, platelet activating factor (PAF) first, then with or without N-formyl-Met-Leu-Phe (fMLP). Reactive oxygen species (ROS) production and p38 phosphorylation were evaluated. In Experiment 2, ROS of neutrophil from WT and Tnfrsf1a-/- mice at 3 or 15 h after thermal injury with or without fMLP treatment were assayed. In Experiment 3, p38 and p44/42 phosphorylation, CXCR2 and macrophage inflammatory protein-2 expression, apoptotic ratio, and activating protein-1 (AP-1) and nuclear factor-kappa B (NF-kappaB) activation of neutrophil from WT and Tnfrsf1a-/- mice at 3 h after thermal injury were tested. FMLP treatment after TNF-alpha or PAF incubation of neutrophil increased ROS of PAF-treated but not TNF-alpha-treated neutrophil. PAF treatment increased ROS of neutrophil in WT and Tnfrsf1a-/- mice. FMLP increased ROS of neutrophil of WT mice at 3 h after thermal but not that of Tnfrsf1a-/- mice. TNF-alpha and PAF increased p38 phosphorylation of neutrophil in WT but not that in Tnfrsf1a-/- mice. Thermal injury increased p38 phosphorylation, NF-kappaB activation, and decreased apoptosis of neutrophil at 3 h after thermal injury in WT but not in Tnfrsf1a-/- mice. Thermal injury also induced AP-1 activation and ROS production on neutrophil at 3 and 15 h after thermal injury, respectively, in WT and Tnfrsf1a-/- mice. Collectively, fMLP stimulates ROS of neutrophil through TNF-alpha signaling; PAF stimulates that of neutrophil through both TNF-alpha-dependent and TNF-alpha-independent pathway. Thermal injury induces a TNF-alpha-dependent priming effect and a TNF-alpha-independent activation effect on neutrophil at 3 and 15 h after thermal injury, respectively. NF-kappaB signaling pathway plays an important role in neutrophil activation. Thermal injury also induces TNF-alpha-dependent delay apoptosis and TNF-alpha-independent AP-1 activation of neutrophil at 3 h after thermal injury. Taken together with the TNF-alpha-dependent p38 and NF-kappaB activation in primed neutrophil, we conclude that thermal injury-induced priming effect of polymorphonuclear neutrophil is TNF-alpha and p38 dependent.
Collapse
Affiliation(s)
- Lee-Wei Chen
- Department of Surgery, Kaohsiung Veterans General Hospital, National Yang-Ming Medical University, Taipei, Taiwan, ROC
| | | | | | | | | |
Collapse
|
42
|
Henry PJ. The protease-activated receptor2 (PAR2)-prostaglandin E2-prostanoid EP receptor axis: a potential bronchoprotective unit in the respiratory tract? Eur J Pharmacol 2006; 533:156-70. [PMID: 16483565 DOI: 10.1016/j.ejphar.2005.12.051] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2005] [Indexed: 01/12/2023]
Abstract
Protease-activated receptor2 (PAR2) is a subtype of G protein-coupled receptor that is widely expressed within the respiratory tract. Stimulation of PAR2 by proteases such as trypsin and tryptase, or by small peptidic activators induces a complex array of effects within the airways. One such PAR2-mediated effect by basal airway epithelial cells is the generation of prostaglandin E2. Prostaglandin E2 produces a raft of anti-inflammatory effects within the airways, principally through the activation of the prostanoid EP2 and EP3 receptor subtypes. This article reviews the PAR2-prostaglandin E2-prostanoid EP receptor axis and discusses approaches through which its activation may provide beneficial effects in respiratory disease.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/pharmacology
- Anti-Inflammatory Agents/therapeutic use
- Dinoprostone/metabolism
- Disease Models, Animal
- Humans
- Ligands
- Pneumonia/metabolism
- Pneumonia/prevention & control
- Receptor, PAR-2/drug effects
- Receptor, PAR-2/metabolism
- Receptors, Prostaglandin E/drug effects
- Receptors, Prostaglandin E/metabolism
- Receptors, Prostaglandin E, EP1 Subtype
- Receptors, Prostaglandin E, EP2 Subtype
- Receptors, Prostaglandin E, EP3 Subtype
- Respiratory Mucosa/drug effects
- Respiratory Mucosa/metabolism
- Respiratory System/drug effects
- Respiratory System/metabolism
Collapse
Affiliation(s)
- Peter J Henry
- School of Medicine and Pharmacology, University of Western Australia, Stirling Highway, Nedlands, Western Australia, 6009, Australia.
| |
Collapse
|
43
|
Iosub R, Klug J, Fijak M, Schneider E, Fröhlich S, Blumbach K, Wennemuth G, Sommerhoff CP, Steinhoff M, Meinhardt A. Development of testicular inflammation in the rat involves activation of proteinase-activated receptor-2. J Pathol 2006; 208:686-98. [PMID: 16450334 DOI: 10.1002/path.1938] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mast cells are involved in early events crucial to inflammation and autoimmune disease. Recently, proteinase-activated receptor-2 (PAR(2)), a G-protein coupled receptor important to injury responses, was shown to be activated by mast cell tryptase. To investigate whether mast cells and PAR(2) are involved in the development and/or aggravation of testicular inflammation, we studied acute and chronic inflammatory models in the rat. In normal testes, PAR(2) was detected immunohistochemically in macrophages, in peritubular cells (PTCs) and in spermatid acrosomes. In experimentally induced autoimmune orchitis (EAO), PAR(2) was strongly upregulated in macrophages and peritubular-like cells, forming concentric layers around granulomas. Mast cells increased 10-fold in number, were more widely distributed throughout the interstitial tissue, and were partially degranulated. Isolated PTCs expressed functional PAR(2), responded to PAR(2) activation by phosphorylating extracellular signal-regulated kinases 1/2 (ERK1/2) and activating protein kinase c, and increased intracellular Ca(2+) concentrations as well as monocyte chemoattractant protein-1 (MCP-1), transforming growth factor beta(2) (TGFbeta(2)), and cyclooxygenase-2 (COX-2) mRNA expression. Expression of these inflammatory mediators, together with iNOS, also increased significantly in testes 50 days after EAO. In vivo, expression of cytokines and inflammatory mediators was upregulated after injection of recombinant tryptase (MCP-1, TGFbeta(2), and COX-2) and a specific PAR(2) peptide agonist (MCP-1, TGFbeta(2)) in the testis after 5 h. These results suggest that PAR(2) activation elicited on PTCs by mast cell tryptase contributes to acute testicular inflammation and that this pathogenetic mechanism may also play a role in autoimmune orchitis.
Collapse
Affiliation(s)
- R Iosub
- Department of Anatomy and Cell Biology, Unit of Reproductive Biology, Justus Liebig University of Giessen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Su X, Camerer E, Hamilton JR, Coughlin SR, Matthay MA. Protease-activated receptor-2 activation induces acute lung inflammation by neuropeptide-dependent mechanisms. THE JOURNAL OF IMMUNOLOGY 2005; 175:2598-605. [PMID: 16081834 DOI: 10.4049/jimmunol.175.4.2598] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Protease-activated receptors (PARs) and tachykinin-immunoreactive fibers are located in the lung as sentries to respond to a variety of pathological stimuli. The effects of PAR activation on the lung have not been adequately studied. We report on the effects of instilling PAR-activating peptides (PAR-APs, including PAR1-, PAR2-, and PAR4-AP) into the lungs of ventilated or spontaneously breathing mice. PAR2-AP, but not PAR1-AP or PAR4-AP, caused a sharp increase in lung endothelial and epithelial permeability to protein, extravascular lung water, and airway tone. No responses to PAR2-AP were detected in PAR2 knockout mice. In bronchoalveolar lavage, PAR2 activation caused 8- and 5-fold increase in MIP-2 and substance P levels, respectively, and a 12-fold increase in the number of neutrophils. Ablation of sensory neurons (by capsaicin) markedly decreased the PAR2-mediated airway constriction, and virtually abolished PAR2-mediated pulmonary inflammation and edema, as did blockade of NK1 or NK2 receptors. Thus, PAR2 activation in the lung induces airway constriction, lung inflammation, and protein-rich pulmonary edema. These effects were either partly or completely neuropeptide dependent, suggesting that PAR2 can cause lung inflammation by a neurogenic mechanism.
Collapse
Affiliation(s)
- Xiao Su
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143, USA
| | | | | | | | | |
Collapse
|
45
|
Hirota Y, Osuga Y, Hirata T, Harada M, Morimoto C, Yoshino O, Koga K, Yano T, Tsutsumi O, Taketani Y. Activation of protease-activated receptor 2 stimulates proliferation and interleukin (IL)-6 and IL-8 secretion of endometriotic stromal cells. Hum Reprod 2005; 20:3547-53. [PMID: 16096323 DOI: 10.1093/humrep/dei255] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Inflammation has been proposed to play essential roles in the pathophysiology of endometriosis, in which neutrophils and mast cells have been suggested to be involved. We studied whether the protease-activated receptor 2 (PAR2), which is activated by enzymes from neutrophils and mast cells, in endometriotic stromal cells (ESC) has any implication in the development of the disease. METHODS Cultured ESC were stimulated with various concentrations of a specific PAR2 agonist peptide. Proliferating activity of the cells was determined using immunostaining of proliferating cell nuclear antigen (a cell proliferation marker), 5-bromo-2'-deoxyuridine incorporation into DNA and cell count. The concentrations of interleukin (IL)-6 and IL-8 were measured using specific enzyme-linked immunosorbent assay kits. The phosphorylation of three mitogen-activated protein kinases (MAPK), i.e. p38 MAPK, p42/44 MAPK and stress-activated protein Kinase/c-jun N terminal Kinase, in ESC was examined with Western blot analysis. RESULTS Activation of PAR2 stimulated the proliferation of ESC and the secretion of IL-6 and IL-8 from ESC in a dose-dependent manner. Activation of PAR2 stimulated the phosphorylation of all three MAPK, and inhibitors of each MAPK suppressed the PAR2 activation-induced proliferation of ESC. CONCLUSIONS The activation of PAR2 in ESC may be involved in the pathophysiology of endometriosis by inducing the growth and inflammation of endometriotic lesions.
Collapse
Affiliation(s)
- Yasushi Hirota
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tokyo, 7-3-1, Hongo, Tokyo, 113-8655, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Steinhoff M, Buddenkotte J, Shpacovitch V, Rattenholl A, Moormann C, Vergnolle N, Luger TA, Hollenberg MD. Proteinase-activated receptors: transducers of proteinase-mediated signaling in inflammation and immune response. Endocr Rev 2005; 26:1-43. [PMID: 15689571 DOI: 10.1210/er.2003-0025] [Citation(s) in RCA: 364] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Serine proteinases such as thrombin, mast cell tryptase, trypsin, or cathepsin G, for example, are highly active mediators with diverse biological activities. So far, proteinases have been considered to act primarily as degradative enzymes in the extracellular space. However, their biological actions in tissues and cells suggest important roles as a part of the body's hormonal communication system during inflammation and immune response. These effects can be attributed to the activation of a new subfamily of G protein-coupled receptors, termed proteinase-activated receptors (PARs). Four members of the PAR family have been cloned so far. Thus, certain proteinases act as signaling molecules that specifically regulate cells by activating PARs. After stimulation, PARs couple to various G proteins and activate signal transduction pathways resulting in the rapid transcription of genes that are involved in inflammation. For example, PARs are widely expressed by cells involved in immune responses and inflammation, regulate endothelial-leukocyte interactions, and modulate the secretion of inflammatory mediators or neuropeptides. Together, the PAR family necessitates a paradigm shift in thinking about hormone action, to include proteinases as key modulators of biological function. Novel compounds that can modulate PAR function may be potent candidates for the treatment of inflammatory or immune diseases.
Collapse
Affiliation(s)
- Martin Steinhoff
- Department of Dermatology and Boltzmann Institute for Immunobiology of the Skin, University of Münster, von-Esmarch-Strasse 58, 48149 Münster, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Morgan MD, Harper L, Lu X, Nash G, Williams J, Savage COS. Can neutrophils be manipulated in vivo? Rheumatology (Oxford) 2004; 44:597-601. [PMID: 15598708 DOI: 10.1093/rheumatology/keh507] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- M D Morgan
- Renal Immunobiology, Division of Immunity and Infection, The School of Medicine, University of Birmingham, Birmingham B15 2TT, UK
| | | | | | | | | | | |
Collapse
|