1
|
Chen Y, Gong Y, Shi M, Zhu H, Tang Y, Huang D, Wang W, Shi C, Xia X, Zhang Y, Liu J, Huang J, Liu M, Chen H, Ma Y, Wang Z, Wang L, Tu W, Zhao Y, Lin J, Jin L, Distler JH, Wu W, Wang J, Shi X. miR-3606-3p alleviates skin fibrosis by integratively suppressing the integrin/FAK, p-AKT/p-ERK, and TGF-β signaling cascades. J Adv Res 2024:S2090-1232(24)00546-0. [PMID: 39571732 DOI: 10.1016/j.jare.2024.11.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/17/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
INTRODUCTION Fibroblast abnormalities are crucial causes of skin fibrosis, including systemic sclerosis (SSc) and keloids. However, their mechanisms, including underlying microRNA regulatory mechanisms, remain elusive. OBJECTIVES This study aimed to evaluate the roles, mechanisms, and therapeutic potential of miR-3606-3p in regulating multiple fibroblast abnormalities. METHODS The miR-3606-3p levels were evaluated in skin tissues and primary fibroblasts. RNA-seq and luciferase assays were employed to identify miR-3606-3p targets. Collagen contraction, western blotting, in vivo imaging, and real-time cellular analysis were used to assess fibroblast abnormalities. The therapeutic potential of miR-3606-3p was evaluated in mice. RESULTS MiR-3606-3p decreased in skin tissues (SSc: Fold Change (FC) = - 2.95, P = 0.0101; keloid: FC = - 3.42, P < 0.0001) and primary fibroblasts (SSc: FC = - 12.74, P = 0.0278; keloid: FC = - 2.08, P = 0.0021) from skin fibrosis patients, and negatively correlated with disease severity. Mechanistically, miR-3606-3p targeted the 3'-untranslated regions (3'-UTRs) of Integrin αV (ITGAV), GRB2-associated binding protein 1 (GAB1), and transforming growth factor beta receptor 2 (TGFBR2), all of these three targets increased in skin fibrosis. Simultaneously, miR-3606-3p inhibited fibroblast's fibrogenesis, migration, inflammation, and proliferation by inhibiting ITGAV/integrin/FAK, GAB1/p-AKT/p-ERK, and TGFBR2/p-SMAD2/3 signaling. ITGAV-mediated integrin/FAK signaling unidirectionally activated the p-AKT/p-ERK and p-SMAD2/3 pathways. Knockdown of GAB1 and TGFRB2 reduced ITGAV-induced p-AKT/p-ERK and p-SMAD2/3 activities. MiR-3606-3p, si-ITGAV, si-GAB1, and si-TGFBR2 exhibited significant inhibition of fibrogenesis and migration. Inflammation was primarily inhibited by si-ITGAV and si-GAB1, while proliferation was primarily inhibited by si-TGFBR2. Moreover, miR-3606-3p significantly attenuates skin fibrosis in keloid-bearing mice. CONCLUSIONS MiR-3606-3p is downregulated in skin fibrosis. Moreover, it negatively correlates with disease severity. Functionally, miR-3606-3p inhibits fibrogenesis, migration, inflammation, and proliferation of fibroblasts. Mechanistically, miR-3606-3p inhibits ITGAV, GAB1, and TGFBR2 by targeting their 3'-UTRs. ITGAV-, GAB1-, and TGFBR2-activated integrin/AKT/ERK/SMAD2/3 signaling induced fibroblast abnormalities. In vivo, miR-3606-3p inhibits skin fibrosis in mice. Therefore, the multi-targeting, multi-phenotypic regulatory properties of miR-3606-3p suggest its potential utility in clinical treatment.
Collapse
Affiliation(s)
- Yahui Chen
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China
| | - Yiyi Gong
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China
| | - Mengkun Shi
- Department of Thoracic Surgery, Huashan Hospital & Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Haoxing Zhu
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China
| | - Yulong Tang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Delin Huang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Wei Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Chenyi Shi
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Xueyi Xia
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China
| | - Ying Zhang
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China
| | - Jianlan Liu
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China
| | - Jia Huang
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China
| | - Mengguo Liu
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China
| | - Huyan Chen
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China
| | - Yanyun Ma
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Ziyu Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Lei Wang
- Division of Rheumatology, Shanghai TCM-Integrated Hospital, Shanghai, China
| | - Wenzhen Tu
- Division of Rheumatology, Shanghai TCM-Integrated Hospital, Shanghai, China
| | - Yinhuan Zhao
- Division of Rheumatology, Shanghai TCM-Integrated Hospital, Shanghai, China
| | - Jinran Lin
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Jörg Hw Distler
- University Hospital Düsseldorf and Heinrich-Heine University, Düsseldorf, Germany
| | - Wenyu Wu
- Department of Dermatology, Huashan Hospital, Shanghai Institute of Dermatology, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, China; Department of Dermatology, Jing'an District Central Hospital, Shanghai, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, and Academy for Engineering and Technology, Fudan University, Shanghai, China.
| | - Jiucun Wang
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China; State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China; Deptartment of Allergy and Immunology, Huashan Hospital, and Research Center of Allergy and Diseases, Fudan University, Shanghai, China; Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Shanghai, China.
| | - Xiangguang Shi
- Department of Dermatology, Huashan Hospital and Human Phenome Institute, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Ghiselli F, Giovagnoni G, Felici M, Tugnoli B, Piva A, Grilli E. A mixture of organic acids and thymol protects primary chicken intestinal epithelial cells from Clostridium perfringens infection in vitro. Poult Sci 2022; 101:102101. [PMID: 36088896 PMCID: PMC9464882 DOI: 10.1016/j.psj.2022.102101] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/14/2022] [Accepted: 07/27/2022] [Indexed: 11/25/2022] Open
Abstract
Necrotic enteritis causes economic losses estimated to be up to 6 billion US dollars per year. Clinical and subclinical infections in poultry are also both correlated with decreased growth and feed efficiency. Moreover, in a context of increased antibiotic resistance, feed additives with enhanced antimicrobial properties are a useful and increasingly needed strategy. In this study, the protective effects of a blend of thymol and organic acids against the effects of Clostridium perfringens type A (CP) on chicken intestinal epithelial cells were investigated and compared to bacitracin, a widely used antibiotic in poultry production. Primary chicken intestinal epithelial cells were challenged with CP for a total time of 3 h to assess the beneficial effect of 2 doses of citric acid, dodecanoic acid, and thymol-containing blend, and compare them with bacitracin. During the challenge, different parameters were recorded, such as transepithelial electrical resistance, cell viability, mRNA expression, and reactive oxygen species production. CP induced inflammation with cytokine production and loss of epithelial barrier integrity. It was also able to induce reactive oxygen species production and increase the caspase expression leading to cellular death. The high dose of the blend acted similarly to bacitracin, preventing the disruptive effects of CP and inducing also an increase in zonula occludens-1 mRNA expression. The low dose only partially prevented the disruptive effects of CP but successfully reduced the associated inflammation. This study shows that the usage of thymol combined with 2 organic acids can protect primary chicken intestinal epithelial cells from CP-induced damages creating a valid candidate to substitute or adjuvate the antibiotic treatment against necrotic enteritis.
Collapse
|
3
|
Kumar R, Rojas IG, Edgerton M. Candida albicans Sap6 Initiates Oral Mucosal Inflammation via the Protease Activated Receptor PAR2. Front Immunol 2022; 13:912748. [PMID: 35844627 PMCID: PMC9277060 DOI: 10.3389/fimmu.2022.912748] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Candida albicans Sap6, a secreted aspartyl protease (Sap), contributes to fungal virulence in oral candidiasis. Beside its protease activity, Sap6 contains RGD (RGDRGD) motif required for its binding to host integrins. Sap6 activates immune cells to induce proinflammatory cytokines, although its ability to interact and activate human oral epithelial cells (OECs) remain unknown. Addition of purified recombinant Sap6 (rSap6) to OECs resulted in production of IL-1β and IL-8 cytokines similar to live hyphal C. albicans. OECs exposed to rSap6 showed phosphorylation of p38 and MKP1 and expression of c-Fos not found with C. albicans Δsap6, heat-inactivated Sap6, or rSap6ΔRGD . Heat inactivated rSap6 was able to induce IL-1β but not IL-8 in OECs, while rSap6ΔRGD induced IL-8 but not IL-1β suggesting parallel signaling pathways. C. albicans hyphae increased surface expression of Protease Activated Receptors PAR1, PAR2 and PAR3, while rSap6 increased PAR2 expression exclusively. Pretreatment of OECs with a PAR2 antagonist blocked rSap6-induced p38 MAPK signaling and IL-8 release, while rSap6ΔRGD had reduced MKP1 signaling and IL-1β release independent from PAR2. OECs exposed to rSap6 exhibited loss of barrier function as measured by TEER and reduction in levels of E-cadherin and occludin junctional proteins that was prevented by pretreating OECs with a PAR2 antagonist. OECs treated with PAR2 antagonist also showed reduced rSap6-mediated invasion by C. albicans cells. Thus, Sap6 may initiate OEC responses mediated both through protease activation of PAR2 and by its RGD domain. This novel role of PAR2 suggests new drug targets to block C. albicans oral infection.
Collapse
Affiliation(s)
| | | | - Mira Edgerton
- Department of Oral Biology, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
4
|
Rai V, Moellmer R, Agrawal DK. The role of CXCL8 in chronic nonhealing diabetic foot ulcers and phenotypic changes in fibroblasts: a molecular perspective. Mol Biol Rep 2022; 49:1565-1572. [PMID: 35044539 DOI: 10.1007/s11033-022-07144-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/11/2022] [Indexed: 12/23/2022]
Abstract
INTRODUCTION A persistent inflammation is perpetuated by infiltrating immune cells and cytokines secreted from these immune cells. Additionally, apoptotic keratinocytes and adipocytes in diabetes causes diabetic foot ulcer (DFU) to arrest in an inflammatory phase without progressing to the resolution phase. This leads to a nonhealing DFU and, despite advanced treatments consisting of wound debridement, off-loading the ulcer of necrotic tissue, wound dressings to keep it moist and control exudate, medication, and preventing infection, DFUs remain a clinical problem. Nonhealing DFUs pose not only an economic burden but also increased morbidity and mortality in the form of psychological stress with and increased chance of amputation, and even death. Thus, investigating the complicated underlying molecular mechanism responsible for nonhealing patterns and designing better therapeutics is warranted. This review article focuses on the role of IL-8-mediated persistent inflammation and phenotypic change of fibroblasts due to this inflammatory cascade. We have discussed various sources of interleukin (IL)-8 secretion and the possible association of IL8-fibroblast plasticity as a cause of nonhealing DFUs. MATERIAL AND METHODS A literature search on PubMed, Google Scholar, and PMC was done including the terms diabetic foot ulcer, diabetes, diabetic ulcer, chronic inflammation, interleukin 8, diabetic wound, and nonhealing diabetic foot ulcers. The articles in the English language and published in last 10 years were selected. From the pool of these, the articles describing the relationship between IL-8 and nonhealing diabetic foot ulcer and diabetic ulcer were used sorted out and used for this review article following PRISMA guidelines. CONCLUSION Increased infiltration of inflammatory immune cells, secretion of pro-inflammatory cytokines, altered keratinocyte-fibroblast function, and phenotypic changes of fibroblasts in DFUs seem to be critical to the nonhealing of DFUs. Thus, inhibiting IL-8 secretion and downstream signaling seems to be a goal of potential therapeutics.
Collapse
Affiliation(s)
- Vikrant Rai
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA.
| | - Rebecca Moellmer
- Western University College of Podiatric Medicine, Pomona, CA, 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| |
Collapse
|
5
|
Akhter N, Wilson A, Thomas R, Al-Rashed F, Kochumon S, Al-Roub A, Arefanian H, Al-Madhoun A, Al-Mulla F, Ahmad R, Sindhu S. ROS/TNF-α Crosstalk Triggers the Expression of IL-8 and MCP-1 in Human Monocytic THP-1 Cells via the NF-κB and ERK1/2 Mediated Signaling. Int J Mol Sci 2021; 22:10519. [PMID: 34638857 PMCID: PMC8508672 DOI: 10.3390/ijms221910519] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/15/2021] [Accepted: 09/22/2021] [Indexed: 01/01/2023] Open
Abstract
IL-8/MCP-1 act as neutrophil/monocyte chemoattractants, respectively. Oxidative stress emerges as a key player in the pathophysiology of obesity. However, it remains unclear whether the TNF-α/oxidative stress interplay can trigger IL-8/MCP-1 expression and, if so, by which mechanism(s). IL-8/MCP-1 adipose expression was detected in lean, overweight, and obese individuals, 15 each, using immunohistochemistry. To detect the role of reactive oxygen species (ROS)/TNF-α synergy as a chemokine driver, THP-1 cells were stimulated with TNF-α, with/without H2O2 or hypoxia. Target gene expression was measured by qRT-PCR, proteins by flow cytometry/confocal microscopy, ROS by DCFH-DA assay, and signaling pathways by immunoblotting. IL-8/MCP-1 adipose expression was significantly higher in obese/overweight. Furthermore, IL-8/MCP-1 mRNA/protein was amplified in monocytic cells following stimulation with TNF-α in the presence of H2O2 or hypoxia (p ˂ 0.0001). Synergistic chemokine upregulation was related to the ROS levels, while pre-treatments with NAC suppressed this chemokine elevation (p ≤ 0.01). The ROS/TNF-α crosstalk involved upregulation of CHOP, ERN1, HIF1A, and NF-κB/ERK-1,2 mediated signaling. In conclusion, IL-8/MCP-1 adipose expression is elevated in obesity. Mechanistically, ROS/TNF-α crosstalk may drive expression of these chemokines in monocytic cells by inducing ER stress, HIF1A stabilization, and signaling via NF-κB/ERK-1,2. NAC had inhibitory effect on oxidative stress-driven IL-8/MCP-1 expression, which may have therapeutic significance regarding meta-inflammation.
Collapse
Affiliation(s)
- Nadeem Akhter
- Department of Immunology & Microbiology, Dasman Diabetes Institute, P.O. Box 1180, Dasman 15462, Kuwait; (N.A.); (A.W.); (R.T.); (F.A.-R.); (S.K.); (A.A.-R.); (H.A.); (R.A.)
| | - Ajit Wilson
- Department of Immunology & Microbiology, Dasman Diabetes Institute, P.O. Box 1180, Dasman 15462, Kuwait; (N.A.); (A.W.); (R.T.); (F.A.-R.); (S.K.); (A.A.-R.); (H.A.); (R.A.)
| | - Reeby Thomas
- Department of Immunology & Microbiology, Dasman Diabetes Institute, P.O. Box 1180, Dasman 15462, Kuwait; (N.A.); (A.W.); (R.T.); (F.A.-R.); (S.K.); (A.A.-R.); (H.A.); (R.A.)
| | - Fatema Al-Rashed
- Department of Immunology & Microbiology, Dasman Diabetes Institute, P.O. Box 1180, Dasman 15462, Kuwait; (N.A.); (A.W.); (R.T.); (F.A.-R.); (S.K.); (A.A.-R.); (H.A.); (R.A.)
| | - Shihab Kochumon
- Department of Immunology & Microbiology, Dasman Diabetes Institute, P.O. Box 1180, Dasman 15462, Kuwait; (N.A.); (A.W.); (R.T.); (F.A.-R.); (S.K.); (A.A.-R.); (H.A.); (R.A.)
| | - Areej Al-Roub
- Department of Immunology & Microbiology, Dasman Diabetes Institute, P.O. Box 1180, Dasman 15462, Kuwait; (N.A.); (A.W.); (R.T.); (F.A.-R.); (S.K.); (A.A.-R.); (H.A.); (R.A.)
| | - Hossein Arefanian
- Department of Immunology & Microbiology, Dasman Diabetes Institute, P.O. Box 1180, Dasman 15462, Kuwait; (N.A.); (A.W.); (R.T.); (F.A.-R.); (S.K.); (A.A.-R.); (H.A.); (R.A.)
| | - Ashraf Al-Madhoun
- Department of Genetics & Bioinformatics, Dasman Diabetes Institute, P.O. Box 1180, Dasman 15462, Kuwait; (A.A.-M.); (F.A.-M.)
- Animal & Imaging Core Facility, Dasman Diabetes Institute, P.O. Box 1180, Dasman 15462, Kuwait
| | - Fahd Al-Mulla
- Department of Genetics & Bioinformatics, Dasman Diabetes Institute, P.O. Box 1180, Dasman 15462, Kuwait; (A.A.-M.); (F.A.-M.)
| | - Rasheed Ahmad
- Department of Immunology & Microbiology, Dasman Diabetes Institute, P.O. Box 1180, Dasman 15462, Kuwait; (N.A.); (A.W.); (R.T.); (F.A.-R.); (S.K.); (A.A.-R.); (H.A.); (R.A.)
| | - Sardar Sindhu
- Animal & Imaging Core Facility, Dasman Diabetes Institute, P.O. Box 1180, Dasman 15462, Kuwait
| |
Collapse
|
6
|
Frombach J, Rancan F, Kübrich K, Schumacher F, Unbehauen M, Blume-Peytavi U, Haag R, Kleuser B, Sabat R, Wolk K, Vogt A. Serine Protease-Mediated Cutaneous Inflammation: Characterization of an Ex Vivo Skin Model for the Assessment of Dexamethasone-Loaded Core Multishell-Nanocarriers. Pharmaceutics 2020; 12:pharmaceutics12090862. [PMID: 32927792 PMCID: PMC7558872 DOI: 10.3390/pharmaceutics12090862] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/02/2020] [Accepted: 09/07/2020] [Indexed: 11/29/2022] Open
Abstract
Standard experimental set-ups for the assessment of skin penetration are typically performed on skin explants with an intact skin barrier or after a partial mechanical or chemical perturbation of the stratum corneum, but they do not take into account biochemical changes. Among the various pathological alterations in inflamed skin, aberrant serine protease (SP) activity directly affects the biochemical environment in the superficial compartments, which interact with topically applied formulations. It further impacts the skin barrier structure and is a key regulator of inflammatory mediators. Herein, we used short-term cultures of ex vivo human skin treated with trypsin and plasmin as inflammatory stimuli to assess the penetration and biological effects of the anti-inflammatory drug dexamethasone (DXM), encapsulated in core multishell-nanocarriers (CMS-NC), when compared to a standard cream formulation. Despite a high interindividual variability, the combined pretreatment of the skin resulted in an average 2.5-fold increase of the transepidermal water loss and swelling of the epidermis, as assessed by optical coherence tomography, as well as in a moderate increase of a broad spectrum of proinflammatory mediators of clinical relevance. The topical application of DXM-loaded CMS-NC or DXM standard cream revealed an increased penetration into SP-treated skin when compared to untreated control skin with an intact barrier. Both formulations, however, delivered sufficient amounts of DXM to effectively suppress the production of interleukin-6 (IL-6), interleukin-8 (IL-8) and Thymic Stromal Lymphopoietin (TSLP). In conclusion, we suggest that the herein presented ex vivo inflammatory skin model is functional and could improve the selection of promising drug delivery strategies for anti-inflammatory compounds at early stages of development.
Collapse
Affiliation(s)
- Janna Frombach
- Clinical Research Center for Hair and Skin Science, Department of Dermatology, Venereology and Allergy, Charité-Universitatsmedizin Berlin, Corporate Member of Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (J.F.); (F.R.); (K.K.); (U.B.-P.)
| | - Fiorenza Rancan
- Clinical Research Center for Hair and Skin Science, Department of Dermatology, Venereology and Allergy, Charité-Universitatsmedizin Berlin, Corporate Member of Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (J.F.); (F.R.); (K.K.); (U.B.-P.)
| | - Katharina Kübrich
- Clinical Research Center for Hair and Skin Science, Department of Dermatology, Venereology and Allergy, Charité-Universitatsmedizin Berlin, Corporate Member of Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (J.F.); (F.R.); (K.K.); (U.B.-P.)
| | - Fabian Schumacher
- Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany; (F.S.); (B.K.)
| | - Michael Unbehauen
- Organic Chemistry, Institute of Chemistry and Biochemistry, Freie Universitaet Berlin, 14195 Berlin, Germany; (M.U.); (R.H.)
| | - Ulrike Blume-Peytavi
- Clinical Research Center for Hair and Skin Science, Department of Dermatology, Venereology and Allergy, Charité-Universitatsmedizin Berlin, Corporate Member of Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (J.F.); (F.R.); (K.K.); (U.B.-P.)
| | - Rainer Haag
- Organic Chemistry, Institute of Chemistry and Biochemistry, Freie Universitaet Berlin, 14195 Berlin, Germany; (M.U.); (R.H.)
| | - Burkhard Kleuser
- Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany; (F.S.); (B.K.)
| | - Robert Sabat
- Psoriasis Research and Treatment Center, Department of Dermatology, Venerology and Allergy/Institute for Medical Immunology, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (R.S.); (K.W.)
| | - Kerstin Wolk
- Psoriasis Research and Treatment Center, Department of Dermatology, Venerology and Allergy/Institute for Medical Immunology, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (R.S.); (K.W.)
| | - Annika Vogt
- Clinical Research Center for Hair and Skin Science, Department of Dermatology, Venereology and Allergy, Charité-Universitatsmedizin Berlin, Corporate Member of Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (J.F.); (F.R.); (K.K.); (U.B.-P.)
- Correspondence:
| |
Collapse
|
7
|
Zeng B, Chen C, Yi Q, Zhang X, Wu X, Zheng S, Li N, She F. N-terminal region of Helicobacter pylori CagA induces IL-8 production in gastric epithelial cells via the β1 integrin receptor. J Med Microbiol 2020; 69:457-464. [DOI: 10.1099/jmm.0.001088] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Introduction.
Helicobacter pylori
is associated with gastrointestinal disease, most notably gastric cancer. Cytotoxin-associated antigen A (CagA), an important virulence factor for
H. pylori
pathogenicity, induces host cells to release inflammatory factors, especially interleukin-8 (IL-8). The mechanism by which C-terminal CagA induces IL-8 production has been studied extensively, but little is known about the role of the N-terminus.
Aim. To investigate the effect of CagA303–456aa (a peptide in the N-terminal CagA) on IL-8 production by gastric epithelial cells.
Methodology. CagA303-456aa was produced by a prokaryotic expression system and purified by Strep-tag affinity chromatography. An integrin β1 (ITGB1)-deficient AGS cell line was constructed using the CRISPR/Cas9 technique, and NCTC 11637 cagA and/or cagL knockout mutants were constructed via homologous recombination. The levels of IL-8 production were determined by enzyme-linked immunosorbent assay (ELISA), and p38 and ERK1/2 phosphorylation were examined by Western blot.
Results. CagA303-456aa induced IL-8 expression by AGS cells. IL-8 induction by CagA303-456aawas specifically inhibited by ITGB1 deficiency. Notably, CagA303-456aa activated the phosphorylation of both p38 and ERK1/2, and blocking p38 and ERK1/2 activity significantly reduced IL-8 induction by CagA303-456aa. ITGB1 deficiency also inhibited the activation of p38 phosphorylation by CagA303-456aa. Finally, experiments in CagA and/or CagL knockout bacterial lines demonstrated that extracellular CagA might induce IL-8 production by AGS cells.
Conclusion. Residues 303–456 of the N-terminal region of CagA induce IL-8 production via a CagA303-456–ITGB1–p38–IL-8 pathway, and ERK1/2 is also involved in the release of IL-8. Extracellular CagA might induce IL-8 production before translocation into AGS cells.
Collapse
Affiliation(s)
- Bangwei Zeng
- Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian Province 350001, PR China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, 1 Xuefu North Road, University Town, Fuzhou, Fujian Province 350122, PR China
- Fujian Key Laboratory of Tumor Microbiology, School of Basic Medical Sciences, Fujian Medical University, 1 Xuefu North Road, University Town, Fuzhou, Fujian Province 350122, PR China
| | - Chu Chen
- Fujian Key Laboratory of Tumor Microbiology, School of Basic Medical Sciences, Fujian Medical University, 1 Xuefu North Road, University Town, Fuzhou, Fujian Province 350122, PR China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, 1 Xuefu North Road, University Town, Fuzhou, Fujian Province 350122, PR China
| | - Qingfeng Yi
- Fujian Key Laboratory of Tumor Microbiology, School of Basic Medical Sciences, Fujian Medical University, 1 Xuefu North Road, University Town, Fuzhou, Fujian Province 350122, PR China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, 1 Xuefu North Road, University Town, Fuzhou, Fujian Province 350122, PR China
| | - Xiaoyan Zhang
- Fujian Key Laboratory of Tumor Microbiology, School of Basic Medical Sciences, Fujian Medical University, 1 Xuefu North Road, University Town, Fuzhou, Fujian Province 350122, PR China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, 1 Xuefu North Road, University Town, Fuzhou, Fujian Province 350122, PR China
| | - Xiangyan Wu
- Fujian Key Laboratory of Tumor Microbiology, School of Basic Medical Sciences, Fujian Medical University, 1 Xuefu North Road, University Town, Fuzhou, Fujian Province 350122, PR China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, 1 Xuefu North Road, University Town, Fuzhou, Fujian Province 350122, PR China
| | - Shurong Zheng
- Fujian Key Laboratory of Tumor Microbiology, School of Basic Medical Sciences, Fujian Medical University, 1 Xuefu North Road, University Town, Fuzhou, Fujian Province 350122, PR China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, 1 Xuefu North Road, University Town, Fuzhou, Fujian Province 350122, PR China
| | - Neng Li
- Fujian Key Laboratory of Tumor Microbiology, School of Basic Medical Sciences, Fujian Medical University, 1 Xuefu North Road, University Town, Fuzhou, Fujian Province 350122, PR China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, 1 Xuefu North Road, University Town, Fuzhou, Fujian Province 350122, PR China
| | - Feifei She
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, 1 Xuefu North Road, University Town, Fuzhou, Fujian Province 350122, PR China
- Fujian Key Laboratory of Tumor Microbiology, School of Basic Medical Sciences, Fujian Medical University, 1 Xuefu North Road, University Town, Fuzhou, Fujian Province 350122, PR China
| |
Collapse
|
8
|
FPR1 gene silencing suppresses cardiomyocyte apoptosis and ventricular remodeling in rats with ischemia/reperfusion injury through the inhibition of MAPK signaling pathway. Exp Cell Res 2018; 370:506-518. [DOI: 10.1016/j.yexcr.2018.07.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/08/2018] [Accepted: 07/10/2018] [Indexed: 12/25/2022]
|
9
|
Zhu ZY, Jia CZ, Luo JM, Wang L. Polyriboinosinic-polyribocytidylic acid facilitates interleukin-6, and interleukin-8 secretion in human dermal fibroblasts via the JAK/STAT3 and p38 MAPK signal transduction pathways. Cytokine 2018; 102:1-6. [PMID: 29245047 DOI: 10.1016/j.cyto.2017.12.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/07/2017] [Accepted: 12/09/2017] [Indexed: 02/05/2023]
Abstract
Polyriboinosinic-polyribocytidylic acid (polyI:C) is a viral dsRNA analoguethat promotes wounds healing, accelerates re-epithelialization, granulation and neovascularization, and induces pro-inflammatory cytokine release. Little is known about polyI:C mediated induction of inflammatory mediators in human dermal fibroblast (HDFs), which form the primary scaffold for epithelial cells covering the wound. Here, we found that polyI:C enhances IL-6 and IL-8 mRNA expression and induces of IL-6 and IL-8 production in a concentration-dependent and time-dependent manner in HDFs. PolyI:C treatment rapidly increased phosphorylation level of both STAT3 and p38 mitogen-activated protein kinase (MAPK). Moreover, pretreatment with AG490, a Janus kinase (JAK) inhibitor, inhibited polyI:C-induced STAT3 phosphorylation and subsequent IL-6 and IL-8 release. Conversely, pretreatment with SB203580, a selective inhibitor of p38 MAPK, blocked p38 MAPK phosphorylation and IL-6 and IL-8 expression. In conclusion, polyI:C induces IL-6 and IL-8 production in HDFs via the JAK/STAT3 and p38 MAPK signaling pathways.
Collapse
Affiliation(s)
- Zhang Ying Zhu
- Department of Pathophysiology, Shantou University Medical College, 5150412, People's Republic of China
| | - Cong Zhuo Jia
- Department of Dermatology, First Affiliated Hospital, Shantou University Medical College, 515041, People's Republic of China
| | - Jian Min Luo
- Department of Pathophysiology, Shantou University Medical College, 5150412, People's Republic of China.
| | - Li Wang
- Shenzhen University General Hospital, 518055, People's Republic of China; Department of Dermatology, First Affiliated Hospital, Shantou University Medical College, 515041, People's Republic of China; Huizhou Municipal Hospital, People's Republic of China.
| |
Collapse
|
10
|
Tatour M, Shapira M, Axelman E, Ghanem S, Keren-Politansky A, Bonstein L, Brenner B, Nadir Y. Thrombin is a selective inducer of heparanase release from platelets and granulocytes via protease-activated receptor-1. Thromb Haemost 2017; 117:1391-1401. [DOI: 10.1160/th16-10-0766] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 04/09/2017] [Indexed: 12/23/2022]
Abstract
SummaryHeparanase, known to be involved in angiogenesis and metastasis, was shown to form a complex with tissue factor (TF) and to enhance the generation of factor Xa. Platelets and granulocytes contain abundant amounts of heparanase that may enhance the coagulation system upon discharge. It was the aim of this study to identify the inducer and pathway of heparanase release from these cells. Platelets and granulocytes were purified from pooled normal plasma and were incubated with ATP, ADP, epinephrine, collagen, ristocetin, arachidonic acid, serotonin, LPS and thrombin. Heparanase levels were assessed by ELISA, heparanase procoagulant activity assay and western blot analysis. The effects of selective protease-activated receptor (PAR)-1 and 2 inhibitors and PAR-1 and 4 activators were studied. An in-house synthesised inhibitory peptide to heparanase was used to evaluate platelet heparanase involvement in activation of the coagulation system. Heparanase was released from platelets only by thrombin induction while other inducers exerted no such effect. The heparanase level in a platelet was found to be 40 % higher than in a granulocyte. Heparanase released from platelets or granulocytes increased factor Xa generation by three-fold. PAR-1 activation via ERK intracellular pathway was found to induce heparanase release. In conclusion, heparanase is selectively released from platelets and granulocytes by thrombin interacting with PAR-1. Heparanase derived from platelets and granulocytes is involved in activation of the extrinsic coagulation pathway. The present study implies on a potential anticoagulant effect, in addition to anti-platelet effect, of the new clinically studied PAR-1 inhibitors.
Collapse
|
11
|
Ge S, Li T, Yao Q, Yan H, Huiyun Z, Zheng Y, Zhang B, He S. Expression of proteinase-activated receptor (PAR)-2 in monocytes from allergic patients and potential molecular mechanism. Cell Biol Toxicol 2016; 32:529-542. [PMID: 27423452 DOI: 10.1007/s10565-016-9353-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 07/05/2016] [Indexed: 02/05/2023]
Abstract
Serine proteases play an important role in inflammation via PARs. However, little is known of expression levels of PARs on monocytes of allergic patients, and influence of serine proteases and PARs on TNF-α secretion from monocytes. Using quantitative real-time PCR (qPCR) and flowcytometry techniques, we observed that the expression level of PAR-2 in monocytes of patients with allergic rhinitis and asthma was increased by 42.9 and 38.2 %. It was found that trypsin, thrombin, and tryptase induced up to 200, 320, and 310 % increase in TNF-α release from monocytes at 16 h, respectively. PAR-1 agonist peptide, SFLLR-NH2, and PAR-2 agonist peptide tc-LIGRLO-NH2 provoked up to 210 and 240 % increase in release of TNF-α. Since SCH 79797, a PAR-1 antagonist, and PD98059, an inhibitor of ERK inhibited thrombin- and SFLLR-NH2-induced TNF-α release, the action of thrombin is most likely through a PAR-1- and ERK-mediated signaling mechanism. Similarly, because FSLLRN-NH2, an inhibitor of PAR-2 diminished tryptase- and tc-LIGRLO-NH2-induced TNF-α release, the action of tryptase appears PAR-2 dependent. Moreover, in vivo study showed that both recombinant cockroach major allergens Per a 1 and Per a 7 provoked upregulation of PAR-2 and PAR-1 expression on CD14+ cells in OVA-sensitized mouse peritoneum. In conclusion, increased expression of PAR-2 in monocytes of AR and asthma implicates that PAR-2 likely play a role in allergy. PAR-2- and PAR-1-mediated TNF-α release from monocytes suggests that these unique protease receptors are involved in the pathogenesis of inflammation.
Collapse
Affiliation(s)
- Shuqing Ge
- Allergy and Clinical Immunology Research Centre, the First Affiliated Hospital of Liaoning Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning, 121001, People's Republic of China
- Department of Dentistry, the First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning, 121001, China
| | - Tao Li
- Department of Infectious Diseases, Shantou University Medical College, Shantou, 515031, China
| | - Qijian Yao
- Department of Infectious Diseases, Shantou University Medical College, Shantou, 515031, China
| | - Hongling Yan
- Clinical Research Centre, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Zhang Huiyun
- Allergy and Clinical Immunology Research Centre, the First Affiliated Hospital of Liaoning Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning, 121001, People's Republic of China
| | - Yanshan Zheng
- Department of Infectious Diseases, Shantou University Medical College, Shantou, 515031, China
| | - Bin Zhang
- Department of Dentistry, the First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning, 121001, China
| | - Shaoheng He
- Allergy and Clinical Immunology Research Centre, the First Affiliated Hospital of Liaoning Medical University, No. 2, Section 5, Renmin Street, Guta District, Jinzhou, Liaoning, 121001, People's Republic of China.
| |
Collapse
|
12
|
Nader MA, Gamiel NM, El-Kashef H, Zaghloul MS. Effect of agmatine on experimental vascular endothelial dysfunction. Hum Exp Toxicol 2015; 35:573-82. [PMID: 26424770 DOI: 10.1177/0960327115597311] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This study was designed to investigate the effect of agmatine sulfate (AG, CAS2482-00-0) in nicotine (NIC)-induced vascular endothelial dysfunction (VED) in rabbits. NIC was administered to produce VED in rabbits with or without AG for 6 weeks. Serum lipid profile, serum thiobarbituric acid reactive substances, reduced glutathione, superoxide dismutase generation, serum nitrite/nitrate, serum vascular cellular adhesion molecule-1 (VCAM-1), and aortic nuclear factor κB (NF-κB) levels were analyzed.Treatment with AG markedly improves lipid profile and prevented NIC-induced VED and oxidative stress. The mechanism of AG in improving NIC-induced VED may be due to the significant reduction in serum VCAM-1 levels and aortic NF-κB. Thus, it may be concluded that AG reduces the oxidative stress, nitric oxide production, VCAM-1 levels, and aortic NF-κB expression, thereby consequently improving the integrity of vascular endothelium.
Collapse
MESH Headings
- Agmatine/pharmacology
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Aorta, Thoracic/physiopathology
- Cholesterol, HDL/blood
- Cholesterol, LDL/blood
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Endothelium, Vascular/physiopathology
- Glutathione/blood
- Lipid Peroxidation/drug effects
- Male
- Nicotine/toxicity
- Nitric Oxide/blood
- Oxidative Stress/drug effects
- Rabbits
- Superoxide Dismutase/blood
- Triglycerides/blood
Collapse
Affiliation(s)
- M A Nader
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Dakahlia Governorate, Egypt Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, El-Madinah El-Munawarah, Kingdom of Saudi Arabia
| | - N M Gamiel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Dakahlia Governorate, Egypt
| | - H El-Kashef
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Dakahlia Governorate, Egypt
| | - M S Zaghloul
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Dakahlia Governorate, Egypt
| |
Collapse
|
13
|
Guenther F, Melzig MF. Protease-activated receptors and their biological role - focused on skin inflammation. ACTA ACUST UNITED AC 2015; 67:1623-33. [PMID: 26709036 DOI: 10.1111/jphp.12447] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 05/10/2015] [Indexed: 02/06/2023]
Abstract
OBJECTIVES For several years, protease-activated receptors (PARs) are targets of science regarding to various diseases and platelet aggregation. In the past, a number of publications related to PARs have been published, which refer to a variety of aspects. An important point of view is the inflammation of the skin, which has not been reported in detail yet. This review will provide an overview of the current knowledge on PARs, and in particular, on the involvement of PARs in terms of skin inflammation. KEY FINDINGS Wound healing is an important step after skin injury and is connected with involvement of PARs and inflammation. An important point in skin inflammation is the coagulation-dependent skin inflammation. SUMMARY PARs are a special kind of receptors, being activated by proteolytic cleavage or chemical agonists. They may play an important role in various physiological processes. It is shown that the proteases are involved in many diseases for example Parkinson's disease and Alzheimer's disease. The fact, that proteases regulate the coagulation, and are involved in interleukin and cytokine release leads to the conclusion that they are involved in inflammation processes.
Collapse
Affiliation(s)
- Florian Guenther
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | | |
Collapse
|
14
|
Schweitzer KS, Chen SX, Law S, Van Demark M, Poirier C, Justice MJ, Hubbard WC, Kim ES, Lai X, Wang M, Kranz WD, Carroll CJ, Ray BD, Bittman R, Goodpaster J, Petrache I. Endothelial disruptive proinflammatory effects of nicotine and e-cigarette vapor exposures. Am J Physiol Lung Cell Mol Physiol 2015; 309:L175-87. [PMID: 25979079 DOI: 10.1152/ajplung.00411.2014] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 05/04/2015] [Indexed: 11/22/2022] Open
Abstract
The increased use of inhaled nicotine via e-cigarettes has unknown risks to lung health. Having previously shown that cigarette smoke (CS) extract disrupts the lung microvasculature barrier function by endothelial cell activation and cytoskeletal rearrangement, we investigated the contribution of nicotine in CS or e-cigarettes (e-Cig) to lung endothelial injury. Primary lung microvascular endothelial cells were exposed to nicotine, e-Cig solution, or condensed e-Cig vapor (1-20 mM nicotine) or to nicotine-free CS extract or e-Cig solutions. Compared with nicotine-containing extract, nicotine free-CS extract (10-20%) caused significantly less endothelial permeability as measured with electric cell-substrate impedance sensing. Nicotine exposures triggered dose-dependent loss of endothelial barrier in cultured cell monolayers and rapidly increased lung inflammation and oxidative stress in mice. The endothelial barrier disruptive effects were associated with increased intracellular ceramides, p38 MAPK activation, and myosin light chain (MLC) phosphorylation, and was critically mediated by Rho-activated kinase via inhibition of MLC-phosphatase unit MYPT1. Although nicotine at sufficient concentrations to cause endothelial barrier loss did not trigger cell necrosis, it markedly inhibited cell proliferation. Augmentation of sphingosine-1-phosphate (S1P) signaling via S1P1 improved both endothelial cell proliferation and barrier function during nicotine exposures. Nicotine-independent effects of e-Cig solutions were noted, which may be attributable to acrolein, detected along with propylene glycol, glycerol, and nicotine by NMR, mass spectrometry, and gas chromatography, in both e-Cig solutions and vapor. These results suggest that soluble components of e-Cig, including nicotine, cause dose-dependent loss of lung endothelial barrier function, which is associated with oxidative stress and brisk inflammation.
Collapse
Affiliation(s)
- Kelly S Schweitzer
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Steven X Chen
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Sarah Law
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Mary Van Demark
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Christophe Poirier
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Matthew J Justice
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Walter C Hubbard
- Department of Clinical Pharmacology, The Johns Hopkins University, Baltimore, Maryland
| | - Elena S Kim
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Xianyin Lai
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Mu Wang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - William D Kranz
- Department of Chemistry and Chemical Biology; Indiana University-Purdue University, Indianapolis, Indiana
| | - Clinton J Carroll
- Department of Chemistry and Chemical Biology; Indiana University-Purdue University, Indianapolis, Indiana
| | - Bruce D Ray
- Department of Physics, Indiana University-Purdue University, Indianapolis, Indiana
| | - Robert Bittman
- Queens College, City University of New York, Flushing, New York; and
| | - John Goodpaster
- Department of Chemistry and Chemical Biology; Indiana University-Purdue University, Indianapolis, Indiana
| | - Irina Petrache
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana; Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana
| |
Collapse
|
15
|
Gong WG, Lin JL, Niu QX, Wang HM, Zhou YC, Chen SY, Liang GW. Paeoniflorin diminishes ConA-induced IL-8 production in primary human hepatic sinusoidal endothelial cells in the involvement of ERK1/2 and Akt phosphorylation. Int J Biochem Cell Biol 2015; 62:93-100. [PMID: 25748730 DOI: 10.1016/j.biocel.2015.02.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 02/17/2015] [Accepted: 02/25/2015] [Indexed: 02/05/2023]
Abstract
Liver diseases are closely associated with elevated levels of interleukin-8 (IL-8), suggesting the ability to inhibit IL-8 production could enhance the treatment of liver diseases. Paeoniflorin is a major active constituent of dried Paeoniae Radix Alba root (Baishao in Chinese) which is widely used in China to treat liver diseases. We examined the effects and underlying mechanisms of paeoniflorin on IL-8 production in primary human hepatic sinusoidal endothelial cells (HHSECs). Concanavalin A (ConA) at 20 μg/mL produced a 5.2-fold increase in IL-8 mRNA by 8h, and a 14.2-fold rise in IL-8 levels by 16 h. Inhibition of MEK (ERK kinase) and extracellular signal-regulated kinase (ERK) by PD98059 and U0126, or inhibition of phosphatidylinositol 3-kinase (PI3K) by LY294002 blocked both ConA-induced IL-8 mRNA expression and IL-8 secretion. Paeoniflorin reduced ConA-induced IL-8 mRNA expression and IL-8 release by 57.9% and 52.8%, respectively, and also decreased ConA-stimulated phosphorylation of ERK1/2 and Akt, suggesting paeoniflorin inhibits IL-8 expression and release by inhibiting the ERK1/2 and Akt pathways. Combining paeoniflorin with U0126 or LY294002 at low doses showed supra-additive inhibition of not only phospho-ERK1/2 and phospho-Akt by 46.4% and 35.0%, but also IL-8 release by 42.4% and 36.1% and IL-8 mRNA expression by 43.5% and 31.8%, respectively. In conclusion, paeoniflorin most likely contributes to the therapy for liver disease by exerting anti-inflammatory effects on HHSECs through blocking IL-8 secretion via downregulation of ERK1/2 and Akt phosphorylation.
Collapse
Affiliation(s)
- Wen-Guang Gong
- Institute of Inflammation and Immune Diseases, Department of Pathophysiology, Key Immunopharmacology Laboratory of Guangdong Province, Shantou University Medical College, Guangdong, PR China
| | - Jue-Long Lin
- Center Laboratory, Shantou University Medical College, Guangdong, PR China
| | - Qing-Xia Niu
- Institute of Inflammation and Immune Diseases, Department of Pathophysiology, Key Immunopharmacology Laboratory of Guangdong Province, Shantou University Medical College, Guangdong, PR China.
| | - Hong-Mei Wang
- Institute of Inflammation and Immune Diseases, Department of Pathophysiology, Key Immunopharmacology Laboratory of Guangdong Province, Shantou University Medical College, Guangdong, PR China
| | - Yan-Chun Zhou
- Institute of Inflammation and Immune Diseases, Department of Pathophysiology, Key Immunopharmacology Laboratory of Guangdong Province, Shantou University Medical College, Guangdong, PR China
| | - Shao-Ying Chen
- Institute of Inflammation and Immune Diseases, Department of Pathophysiology, Key Immunopharmacology Laboratory of Guangdong Province, Shantou University Medical College, Guangdong, PR China
| | - Guo-Wu Liang
- Institute of Inflammation and Immune Diseases, Department of Pathophysiology, Key Immunopharmacology Laboratory of Guangdong Province, Shantou University Medical College, Guangdong, PR China
| |
Collapse
|
16
|
Thrombin enhances NGF-mediated neurite extension via increased and sustained activation of p44/42 MAPK and p38 MAPK. PLoS One 2014; 9:e103530. [PMID: 25061982 PMCID: PMC4111596 DOI: 10.1371/journal.pone.0103530] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 06/29/2014] [Indexed: 12/13/2022] Open
Abstract
Rapid neurite remodeling is fundamental to nervous system development and plasticity. It involves neurite extension that is regulated by NGF through PI3K/AKT, p44/42 MAPK and p38 MAPK. It also involves neurite retraction that is regulated by the serine protease, thrombin. However, the intracellular signaling pathway by which thrombin causes neurite retraction is unknown. Using the PC12 neuronal cell model, we demonstrate that thrombin utilizes the PI3K/AKT pathway for neurite retraction in NGF-differentiated cells. Interestingly, however, we found that thrombin enhances NGF-induced neurite extension in differentiating cells. This is achieved through increased and sustained activation of p44/42 MAPK and p38 MAPK. Thus, thrombin elicits opposing effects in differentiated and differentiating cells through activation of distinct signaling pathways: neurite retraction in differentiated cells via PI3K/AKT, and neurite extension in differentiating cells via p44/42 MAPK and p38 MAPK. These findings, which also point to a novel cooperative role between thrombin and NGF, have significant implications in the development of the nervous system and the disease processes that afflicts it as well as in the potential of combined thrombin and NGF therapy for impaired learning and memory, and spinal cord injury which all require neurite extension and remodeling.
Collapse
|
17
|
Tao KM, Tao Y, Chen CY, Yang LQ, Lu ZJ, Sun YM, Huang SD, Yu WF. Proteinase-activated Receptor 1 Contributed to Up-regulation of Enkephalin in Keratinocytes of Patients with Obstructive Jaundice. Anesthesiology 2014; 121:127-39. [PMID: 24614324 DOI: 10.1097/aln.0000000000000210] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Abstract
Background:
Skin synthesis of endogenous opioids such as enkephalin is considered to be increased in cholestatic rodents, which may induce antinociception in cholestatic liver disease. No studies have reported yet the expression of skin enkephalin in patients with cholestasis.
Methods:
Electrical pain threshold, postoperative morphine consumption, and skin enkephalin expression were measured in patients with jaundice (n = 18) and control patients (n = 16). Male Sprague–Dawley rats (n = 52) and human keratinocyte cell line HaCaT were used in vivo and in vitro studies, respectively. Nociceptive thresholds and plasma and skin levels of methionine-enkephalin were compared in protease-activated receptors-1–antagonized and control bile duct–ligated rats. In in vitro study, the effect on thrombin-induced enkephalin expression was examined and the role of extracellular regulated protein kinases 1/2 and p38 was investigated.
Results:
The authors found that: (1) the electrical pain threshold (mean ± SD) was 1.1 ± 0.1 mA in control patients, whereas it was significantly increased in patients with jaundice (1.7 ± 0.3 mA); 48-h postoperative morphine consumption was approximately 50% higher in the control group than that in the group with jaundice; (2) Skin keratinocytes enkephalin expression was increased in the patients with jaundice; (3) Protease-activated receptors-1 antagonist 1 μg·kg−1·day−1 treatment to the bile duct–ligated rats significantly reduced plasma levels of methionine-enkephalin, nociceptive thresholds, and keratinocytes enkephalin expression; and (4) protease-activated receptors-1 activation induced enkephalin expression through phosphorylation of extracellular regulated protein kinases 1/2 and p38 in keratinocytes.
Conclusion:
Protease-activated receptors-1 activation in peripheral keratinocytes may play an important role in the local synthesis of enkephalin during cholestasis.
Collapse
Affiliation(s)
- Kun-Ming Tao
- From the Department of Anesthesiology and Intensive Care Unit, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University, Shanghai, China (K.-M.T., Y.T., C.-Y.C., L.-Q.Y., Z.-J.L., Y.-M.S., W.-F.Y.); and Department of Cardiothoracic Surgery, Institute of Cardiothoracic Surgery, Changhai Hospital, the Second Military Medical University, Shanghai, China (S.-D.H.)
| | - Yong Tao
- From the Department of Anesthesiology and Intensive Care Unit, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University, Shanghai, China (K.-M.T., Y.T., C.-Y.C., L.-Q.Y., Z.-J.L., Y.-M.S., W.-F.Y.); and Department of Cardiothoracic Surgery, Institute of Cardiothoracic Surgery, Changhai Hospital, the Second Military Medical University, Shanghai, China (S.-D.H.)
| | - Cai-Yang Chen
- From the Department of Anesthesiology and Intensive Care Unit, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University, Shanghai, China (K.-M.T., Y.T., C.-Y.C., L.-Q.Y., Z.-J.L., Y.-M.S., W.-F.Y.); and Department of Cardiothoracic Surgery, Institute of Cardiothoracic Surgery, Changhai Hospital, the Second Military Medical University, Shanghai, China (S.-D.H.)
| | - Li-Qun Yang
- From the Department of Anesthesiology and Intensive Care Unit, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University, Shanghai, China (K.-M.T., Y.T., C.-Y.C., L.-Q.Y., Z.-J.L., Y.-M.S., W.-F.Y.); and Department of Cardiothoracic Surgery, Institute of Cardiothoracic Surgery, Changhai Hospital, the Second Military Medical University, Shanghai, China (S.-D.H.)
| | - Zhi-Jie Lu
- From the Department of Anesthesiology and Intensive Care Unit, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University, Shanghai, China (K.-M.T., Y.T., C.-Y.C., L.-Q.Y., Z.-J.L., Y.-M.S., W.-F.Y.); and Department of Cardiothoracic Surgery, Institute of Cardiothoracic Surgery, Changhai Hospital, the Second Military Medical University, Shanghai, China (S.-D.H.)
| | - Yu-Ming Sun
- From the Department of Anesthesiology and Intensive Care Unit, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University, Shanghai, China (K.-M.T., Y.T., C.-Y.C., L.-Q.Y., Z.-J.L., Y.-M.S., W.-F.Y.); and Department of Cardiothoracic Surgery, Institute of Cardiothoracic Surgery, Changhai Hospital, the Second Military Medical University, Shanghai, China (S.-D.H.)
| | - Sheng-Dong Huang
- From the Department of Anesthesiology and Intensive Care Unit, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University, Shanghai, China (K.-M.T., Y.T., C.-Y.C., L.-Q.Y., Z.-J.L., Y.-M.S., W.-F.Y.); and Department of Cardiothoracic Surgery, Institute of Cardiothoracic Surgery, Changhai Hospital, the Second Military Medical University, Shanghai, China (S.-D.H.)
| | - Wei-Feng Yu
- From the Department of Anesthesiology and Intensive Care Unit, Eastern Hepatobiliary Surgery Hospital, the Second Military Medical University, Shanghai, China (K.-M.T., Y.T., C.-Y.C., L.-Q.Y., Z.-J.L., Y.-M.S., W.-F.Y.); and Department of Cardiothoracic Surgery, Institute of Cardiothoracic Surgery, Changhai Hospital, the Second Military Medical University, Shanghai, China (S.-D.H.)
| |
Collapse
|
18
|
Evaluation on potential contributions of protease activated receptors related mediators in allergic inflammation. Mediators Inflamm 2014; 2014:829068. [PMID: 24876677 PMCID: PMC4021743 DOI: 10.1155/2014/829068] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 01/20/2014] [Indexed: 01/16/2023] Open
Abstract
Protease activated receptors (PARs) have been recognized as a distinctive four-member family of seven transmembrane G protein-coupled receptors (GPCRs) that can be cleaved by certain serine proteases. In recent years, there has been considerable interest in the role of PARs in allergic inflammation, the fundamental pathologic changes of allergy, but the potential roles of PARs in allergy remain obscure. Since many of these proteases are produced and actively involved in the pathologic process of inflammation including exudation of plasma components, inflammatory cell infiltration, and tissue damage and repair, PARs appear to make important contribution to allergy. The aim of the present review is to summarize the expression of PARs in inflammatory and structural cells, the influence of agonists or antagonists of PARs on cell behavior, and the involvement of PARs in allergic disorders, which will help us to better understand the roles of serine proteases and PARs in allergy.
Collapse
|
19
|
Mercer PF, Williams AE, Scotton CJ, José RJ, Sulikowski M, Moffatt JD, Murray LA, Chambers RC. Proteinase-activated receptor-1, CCL2, and CCL7 regulate acute neutrophilic lung inflammation. Am J Respir Cell Mol Biol 2014; 50:144-57. [PMID: 23972264 DOI: 10.1165/rcmb.2013-0142oc] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
PAR1 plays a central role in mediating the interplay between coagulation and inflammation, but its role in regulating acute neutrophilic inflammation is unknown. We report that antagonism of PAR1 was highly effective at reducing acute neutrophil accumulation in a mouse model of LPS-induced lung inflammation. PAR1 antagonism also reduced alveolar-capillary barrier disruption in these mice. This protection was associated with a reduction in the expression of the chemokines, CCL2 and CCL7, but not the proinflammatory cytokines, TNF and IL-6, or the classic neutrophil chemoattractants, CXCL1 and CXCL2. Antibody neutralization of CCL2 and CCL7 significantly reduced LPS-induced total leukocyte and neutrophil accumulation, recovered from the bronchoalveolar lavage fluid of challenged mice. Immunohistochemical analysis revealed that CCL2 predominantly localized to alveolar macrophages and pulmonary epithelial cells, whereas CCL7 was restricted to the pulmonary epithelium. In keeping with these observations, the intranasal administration of recombinant CCL2 (rCCL2) and rCCL7 led to the accumulation of neutrophils within the lung airspaces of naive mice in the absence of any underlying inflammation. Flow cytometry analysis further demonstrated an increase in Ly6G(hi) neutrophils expressing the chemokine receptors, CCR1 and CCR2, isolated from mouse lungs compared with circulating neutrophils. Conversely, the expression of CXCR2 decreased on neutrophils isolated from the lung compared with circulating neutrophils. Furthermore, this switch in chemokine receptor expression was accentuated after acute LPS-induced lung inflammation. Collectively, these findings reveal a novel role for PAR1 and the chemokines, CCL2 and CCL7, during the early events of acute neutrophilic inflammation.
Collapse
Affiliation(s)
- Paul F Mercer
- 1 Centre for Inflammation and Tissue Repair, University College London, London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Expression, function and cooperating partners of protease-activated receptor type 3 in vascular endothelial cells and B lymphocytes studied with specific monoclonal antibody. Mol Immunol 2013; 54:319-26. [PMID: 23352962 DOI: 10.1016/j.molimm.2012.12.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 12/26/2012] [Indexed: 01/01/2023]
Abstract
Receptor-specific antibodies can both prevent ligand-receptor interaction and initiate receptor signaling. Previously we generated monoclonal antibody 8E8 (mAb 8E8) against protease-activated receptor type 3 (PAR3) which inhibited proliferation of B cell hybridoma. Here we used mAb 8E8 and PAR1-specific polyclonal antibody to reveal the functions and cooperating partners of PAR3 in endothelial cells and in B lymphocytes. MAb 8E8 or PAR1 agonist peptide stimulated IL-6 and IL-8 production and VCAM-1 expression in HPMEC-ST1.6R cells. PAR1 antibody stimulated only VCAM-1 expression, while ICAM-1 expression was stimulated with mAB 8E8 or PAR3 peptide. MAb 8E8 stimulated weak mitogenic response, while PAR1 antibody inhibited it in normal but not in malignant B lymphocytes. Sandwich ELISA assay demonstrated the interaction of PAR3 with PAR1 in malignant cell lines and with IgM in normal B lymphocytes. It is concluded that PAR3 cooperates with PAR1 to mediate the effect of thrombin on cytokine production and VCAM-1 expression in endothelial cells and on cell proliferation in malignant B cells. ICAM-1 expression in endothelial cells requires PAR3 without PAR1. The inhibitory effect of thrombin in normal B lymphocytes is mediated by PAR1 alone, while mitogenic and pro-survival signaling in B lymphocytes is provided through PAR3 in cooperation with BCR.
Collapse
|
21
|
Yang H, Li T, Wei J, Zhang H, He S. Induction of tumor necrosis factor (TNF) release from subtypes of T cells by agonists of proteinase activated receptors. Mediators Inflamm 2013; 2013:165453. [PMID: 24453410 PMCID: PMC3876890 DOI: 10.1155/2013/165453] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 11/22/2013] [Accepted: 11/22/2013] [Indexed: 02/05/2023] Open
Abstract
Serine proteinases have been recognized as playing an important role in inflammation via proteinase activated receptors (PARs). However, little is known about the influence of serine proteinases and PARs on TNF secretion from highly purified T cells. We challenged T cells from human peripheral blood with serine proteinases and agonist peptides of PARs and measured the levels of TNF in culture supernatants by ELISA. The results showed that thrombin and trypsin, but not tryptase, stimulated approximately up to 2.5-fold increase in TNF release from T cells following 16 h incubation. Proteinase inhibitors and PAR-1 antagonist SCH 79797 almost completely abolished thrombin- and trypsin-induced TNF release from T cells. Agonist peptides of PAR-1, but not PAR-2 induced TNF release from T cells. Moreover, trypsin- and thrombin-induced upregulated expression of TNF was observed in CD4+, IL-4+, or CD25+ T cells, but not in IFN+ or IL-17+ T cells. The signaling pathways MAPK/ERK and PI3K/Akt are involved in the thrombin- and trypsin-induced TNF release from T cells. In conclusion, thrombin and trypsin can induce TNF release from IL-4+ and CD25+ T cells through activation of PAR-1 and therefore contribute to regulation of immune response and inflammation of the body.
Collapse
Affiliation(s)
- Haiwei Yang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 300 Guangzhou Road, Jiangsu 210029, China
| | - Tao Li
- Department of Infectious Diseases, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Jifu Wei
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 300 Guangzhou Road, Jiangsu 210029, China
| | - Huiyun Zhang
- Department of Infectious Diseases, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Shaoheng He
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 300 Guangzhou Road, Jiangsu 210029, China
- Allergy and Clinical Immunology Research Centre, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning 121001, China
- *Shaoheng He:
| |
Collapse
|
22
|
Lee MMK, Chui RKS, Tam IYS, Lau AHY, Wong YH. CCR1-mediated STAT3 tyrosine phosphorylation and CXCL8 expression in THP-1 macrophage-like cells involve pertussis toxin-insensitive Gα(14/16) signaling and IL-6 release. THE JOURNAL OF IMMUNOLOGY 2012; 189:5266-76. [PMID: 23125416 DOI: 10.4049/jimmunol.1103359] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Agonists of CCR1 contribute to hypersensitivity reactions and atherosclerotic lesions, possibly via the regulation of the transcription factor STAT3. CCR1 was demonstrated to use pertussis toxin-insensitive Gα(14/16) to stimulate phospholipase Cβ and NF-κB, whereas both Gα(14) and Gα(16) are also capable of activating STAT3. The coexpression of CCR1 and Gα(14/16) in human THP-1 macrophage-like cells suggests that CCR1 may use Gα(14/16) to induce STAT3 activation. In this study, we demonstrated that a CCR1 agonist, leukotactin-1 (CCL15), could indeed stimulate STAT3 Tyr(705) and Ser(727) phosphorylation via pertussis toxin-insensitive G proteins in PMA-differentiated THP-1 cells, human erythroleukemia cells, and HEK293 cells overexpressing CCR1 and Gα(14/16). The STAT3 Tyr(705) and Ser(727) phosphorylations were independent of each other and temporally distinct. Subcellular fractionation and confocal microscopy illustrated that Tyr(705)-phosphorylated STAT3 translocated to the nucleus, whereas Ser(727)-phosphorylated STAT3 was retained in the cytosol after CCR1/Gα(14) activation. CCL15 was capable of inducing IL-6 and IL-8 (CXCL8) production in both THP-1 macrophage-like cells and HEK293 cells overexpressing CCR1 and Gα(14/16). Neutralizing Ab to IL-6 inhibited CCL15-mediated STAT3 Tyr(705) phosphorylation, whereas inhibition of STAT3 activity abolished CCL15-activated CXCL8 release. The ability of CCR1 to signal through Gα(14/16) provides a linkage for CCL15 to regulate IL-6/STAT3-signaling cascades, leading to expression of CXCL8, a cytokine that is involved in inflammation and the rupture of atherosclerotic plaque.
Collapse
Affiliation(s)
- Maggie M K Lee
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | | | | | | | | |
Collapse
|
23
|
Ortiz-Stern A, Deng X, Smoktunowicz N, Mercer PF, Chambers RC. PAR-1-dependent and PAR-independent pro-inflammatory signaling in human lung fibroblasts exposed to thrombin. J Cell Physiol 2012; 227:3575-84. [PMID: 22278285 DOI: 10.1002/jcp.24061] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Proteinase-activated receptors (PARs) are crucial in orchestrating cellular responses to coagulation proteinases, such as thrombin and FXa. Four PARs have been characterized and have been shown to be differentially expressed in mice and humans and between tissues. We have previously shown that in murine lung fibroblasts, PAR-1 is solely responsible for all cellular responses to thrombin and FXa. In contrast, we report here that in primary human lung fibroblasts (pHLFs), known PARs fail to account for all of the cellular responses to thrombin, in particular in the presence of high, but physiologically achievable concentrations of thrombin. We report that pHLFs secrete CCL2 in a PAR-1-dependent manner at low thrombin concentration (∼0.3 nM). At or above 10 nM thrombin, pharmacological antagonism (RWJ-58259) fails to block thrombin-induced CCL2 release; whereas PAR-1 cleavage-blocking monoclonal antibodies (ATAP2 and WEDE15) only partially inhibit thrombin-induced CCL2 secretion. In addition, activation of PAR-3, PAR-4, and transactivation of either PAR-2 or EGFR were ruled out as being responsible for thrombin-mediated CCL2 secretion at high yet standard concentrations of the proteinase. We further provide evidence that PAR-1-dependent and PAR-independent signaling involves the rapid phosphorylation of ERK, which in turn is absolutely required for thrombin-induced CCL2 secretion at both low and standard concentration of the proteinase. Our findings suggest the existence of a PAR-independent signaling mechanism in human lung fibroblasts and have important implications for the design of therapeutic strategies aimed at blocking pro-inflammatory signaling responses associated with excessive thrombin generation.
Collapse
|
24
|
Kunigal S, Ponnusamy MP, Momi N, Batra SK, Chellappan SP. Nicotine, IFN-γ and retinoic acid mediated induction of MUC4 in pancreatic cancer requires E2F1 and STAT-1 transcription factors and utilize different signaling cascades. Mol Cancer 2012; 11:24. [PMID: 22537161 PMCID: PMC3464875 DOI: 10.1186/1476-4598-11-24] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 04/26/2012] [Indexed: 12/12/2022] Open
Abstract
Background The membrane-bound mucins are thought to play an important biological role in cell–cell and cell–matrix interactions, in cell signaling and in modulating biological properties of cancer cell. MUC4, a transmembrane mucin is overexpressed in pancreatic tumors, while remaining undetectable in the normal pancreas, thus indicating a potential role in pancreatic cancer pathogenesis. The molecular mechanisms involved in the regulation of MUC4 gene are not yet fully understood. Smoking is strongly correlated with pancreatic cancer and in the present study; we elucidate the molecular mechanisms by which nicotine as well as agents like retinoic acid (RA) and interferon-γ (IFN-γ) induce the expression of MUC4 in pancreatic cancer cell lines CD18, CAPAN2, AsPC1 and BxPC3. Results Chromatin immunoprecipitation assays and real-time PCR showed that transcription factors E2F1 and STAT1 can positively regulate MUC4 expression at the transcriptional level. IFN-γ and RA could collaborate with nicotine in elevating the expression of MUC4, utilizing E2F1 and STAT1 transcription factors. Depletion of STAT1 or E2F1 abrogated the induction of MUC4; nicotine-mediated induction of MUC4 appeared to require α7-nicotinic acetylcholine receptor subunit. Further, Src and ERK family kinases also mediated the induction of MUC4, since inhibiting these signaling molecules prevented the induction of MUC4. MUC4 was also found to be necessary for the nicotine-mediated invasion of pancreatic cancer cells, suggesting that induction of MUC4 by nicotine and other agents might contribute to the genesis and progression of pancreatic cancer. Conclusions Our studies show that agents that can promote the growth and invasion of pancreatic cancer cells induce the MUC4 gene through multiple pathways and this induction requires the transcriptional activity of E2F1 and STAT1. Further, the Src as well as ERK signaling pathways appear to be involved in the induction of this gene. It appears that targeting these signaling pathways might inhibit the expression of MUC4 and prevent the proliferation and invasion of pancreatic cancer cells.
Collapse
Affiliation(s)
- Sateesh Kunigal
- Dept, of Tumor Biology H, Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | | | | | | | | |
Collapse
|
25
|
Cooper DM, Pechkovsky DV, Hackett TL, Knight DA, Granville DJ. Granzyme K activates protease-activated receptor-1. PLoS One 2011; 6:e21484. [PMID: 21760880 PMCID: PMC3128063 DOI: 10.1371/journal.pone.0021484] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Accepted: 06/02/2011] [Indexed: 11/19/2022] Open
Abstract
Granzyme K (GrK) is a trypsin-like serine protease that is elevated in patients with sepsis and acute lung inflammation. While GrK was originally believed to function exclusively as a pro-apoptotic protease, recent studies now suggest that GrK may possess other non-cytotoxic functions. In the context of acute lung inflammation, we hypothesized that GrK induces pro-inflammatory cytokine release through the activation of protease-activated receptors. The direct effect of extracellular GrK on PAR activation, intracellular signaling and cytokine was assessed using cultured human lung fibroblasts. Extracellular GrK induced secretion of IL-6, IL-8 and MCP-1 in a dose- and time-dependent manner in lung fibroblasts. Heat-inactivated GrK did not induce cytokine release indicating that protease activity is required. Furthermore, GrK induced activation of both the ERK1/2 and p38 MAP kinase signaling pathways, and significantly increased fibroblast proliferation. Inhibition of ERK1/2 abrogated the GrK-mediated cytokine release. Through the use of PAR-1 and PAR-2 neutralizing antibodies, it was determined that PAR-1 is essential for GrK-induced IL-6, IL-8 and MCP-1 release. In summary, extracellular GrK is capable of activating PAR-1 and inducing fibroblast cytokine secretion and proliferation.
Collapse
Affiliation(s)
- Dawn M. Cooper
- Institute for Heart and Lung Health, St. Paul's Hospital, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dmitri V. Pechkovsky
- Institute for Heart and Lung Health, St. Paul's Hospital, Vancouver, British Columbia, Canada
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tillie L. Hackett
- Institute for Heart and Lung Health, St. Paul's Hospital, Vancouver, British Columbia, Canada
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Darryl A. Knight
- Institute for Heart and Lung Health, St. Paul's Hospital, Vancouver, British Columbia, Canada
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - David J. Granville
- Institute for Heart and Lung Health, St. Paul's Hospital, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
26
|
Adams MN, Ramachandran R, Yau MK, Suen JY, Fairlie DP, Hollenberg MD, Hooper JD. Structure, function and pathophysiology of protease activated receptors. Pharmacol Ther 2011; 130:248-82. [PMID: 21277892 DOI: 10.1016/j.pharmthera.2011.01.003] [Citation(s) in RCA: 280] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 01/03/2011] [Indexed: 12/18/2022]
Abstract
Discovered in the 1990s, protease activated receptors(1) (PARs) are membrane-spanning cell surface proteins that belong to the G protein coupled receptor (GPCR) family. A defining feature of these receptors is their irreversible activation by proteases; mainly serine. Proteolytic agonists remove the PAR extracellular amino terminal pro-domain to expose a new amino terminus, or tethered ligand, that binds intramolecularly to induce intracellular signal transduction via a number of molecular pathways that regulate a variety of cellular responses. By these mechanisms PARs function as cell surface sensors of extracellular and cell surface associated proteases, contributing extensively to regulation of homeostasis, as well as to dysfunctional responses required for progression of a number of diseases. This review examines common and distinguishing structural features of PARs, mechanisms of receptor activation, trafficking and signal termination, and discusses the physiological and pathological roles of these receptors and emerging approaches for modulating PAR-mediated signaling in disease.
Collapse
Affiliation(s)
- Mark N Adams
- Mater Medical Research Institute, Aubigny Place, Raymond Terrace, South Brisbane Qld 4101, Australia
| | | | | | | | | | | | | |
Collapse
|
27
|
Bradford ST, Stamatovic SM, Dondeti RS, Keep RF, Andjelkovic AV. Nicotine aggravates the brain postischemic inflammatory response. Am J Physiol Heart Circ Physiol 2011; 300:H1518-29. [PMID: 21239632 DOI: 10.1152/ajpheart.00928.2010] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A substantial body of evidence suggests that nicotine adversely affects cerebral blood flow and the blood-brain barrier and is a risk factor for stroke. The present study investigated the effect of nicotine on cerebrovascular endothelium under basal and ischemia/reperfusion injury under in vivo condition. Nicotine (2 mg/kg sc) was administered to mice over 14 days, which resulted in plasma nicotine levels of ∼100 ng/ml, reflecting plasma concentrations in average to heavy smokers. An analysis of the phenotype of isolated brain microvessels after nicotine exposure indicated higher expression of inflammatory mediators, cytokines (IL-1β, TNF-α, and IL-18), chemokines (CCL2 and CX(3)CL1), and adhesion molecules (ICAM-1, VCAM-1, and P-selectins), and this was accompanied by enhanced leukocyte infiltration into brain during ischemia/reperfusion (P < 0.01). Nicotine had a profound effect on ischemia/reperfusion injury; i.e., increased brain infarct size (P < 0.01), worse neurological deficits, and a higher mortality rate. These experiments illuminate, for the first time, how nicotine regulates brain endothelial cell phenotype and postischemic inflammatory response at the brain-vascular interface.
Collapse
Affiliation(s)
- Shayna T Bradford
- Department of Pathology, University of Michigan, Medical School, Ann Arbor, 48109-0532, USA
| | | | | | | | | |
Collapse
|
28
|
Protease activated receptor-1 inhibits the Maspin tumor-suppressor gene to determine the melanoma metastatic phenotype. Proc Natl Acad Sci U S A 2010; 108:626-31. [PMID: 21187389 DOI: 10.1073/pnas.1006886108] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The thrombin receptor protease activated receptor-1 (PAR-1) is overexpressed in metastatic melanoma cell lines and tumor specimens. Previously, we demonstrated a significant reduction in tumor growth and experimental lung metastasis after PAR-1 silencing via systemic delivery of siRNA encapsulated into nanoliposomes. Gene expression profiling identified a 40-fold increase in expression of Maspin in PAR-1-silenced metastatic melanoma cell lines. Maspin promoter activity was significantly increased after PAR-1 silencing, suggesting that PAR1 negatively regulates Maspin at the transcriptional level. ChIP analyses revealed that PAR-1 decreases binding of Ets-1 and c-Jun transcription factors to the Maspin promoter, both known to activate Maspin transcription. PAR-1 silencing did not affect Ets-1 or c-Jun expression; rather it resulted in increased expression of the chromatin remodeling complex CBP/p300, as well as decreased activity of the CBP/p300 inhibitor p38, resulting in increased binding of Ets-1 and c-Jun to the Maspin promoter and higher Maspin expression. Functionally, Maspin expression reduced the invasive capability of melanoma cells after PAR-1 silencing, which was abrogated after rescuing with PAR-1. Furthermore, tumor growth and experimental lung metastasis was significantly decreased after expressing Maspin in a metastatic melanoma cell line. Moreover, silencing Maspin in PAR-1-silenced cells reverted the inhibition of tumor growth and experimental lung metastasis. Herein, we demonstrate a mechanism by which PAR-1 negatively regulates the expression of the Maspin tumor-suppressor gene in the acquisition of the metastatic melanoma phenotype, thus attributing an alternative function to PAR-1 other than coagulation.
Collapse
|
29
|
Rohani MG, DiJulio DH, An JY, Hacker BM, Dale BA, Chung WO. PAR1- and PAR2-induced innate immune markers are negatively regulated by PI3K/Akt signaling pathway in oral keratinocytes. BMC Immunol 2010; 11:53. [PMID: 21029417 PMCID: PMC2988058 DOI: 10.1186/1471-2172-11-53] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Accepted: 10/28/2010] [Indexed: 11/10/2022] Open
Abstract
Background Protease-Activated Receptors (PARs), members of G-protein-coupled receptors, are activated by proteolytic activity of various proteases. Activation of PAR1 and PAR2 triggers innate immune responses in human oral keratinocytes (HOKs), but the signaling pathways downstream of PAR activation in HOKs have not been clearly defined. In this study, we aimed to determine if PAR1- and PAR2-mediated signaling differs in the induction of innate immune markers CXCL3, CXCL5 and CCL20 via ERK, p38 and PI3K/Akt. Results Our data show the induction of innate immunity by PAR1 requires both p38 and ERK MAP kinases, while PAR2 prominently signals via p38. However, inhibition of PI3K enhances expression of innate immune markers predominantly via suppressing p38 phosphorylation signaled by PAR activation. Conclusion Our data indicate that proteases mediating PAR1 and PAR2 activation differentially signal via MAP kinase cascades. In addition, the production of chemokines induced by PAR1 and PAR2 is suppressed by PI3K/Akt, thus keeping the innate immune responses of HOK in balance. The results of our study provide a novel insight into signaling pathways involved in PAR activation.
Collapse
Affiliation(s)
- Maryam G Rohani
- Department of Medicine/Dermatology, University of Washington, Seattle, WA 98195-6524, USA
| | | | | | | | | | | |
Collapse
|
30
|
Hosokawa I, Hosokawa Y, Ozaki K, Yumoto H, Nakae H, Matsuo T. Proinflammatory effects of muramyldipeptide on human gingival fibroblasts. J Periodontal Res 2010; 45:193-9. [PMID: 20470259 DOI: 10.1111/j.1600-0765.2009.01217.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND AND OBJECTIVE Because human gingival fibroblasts (HGFs) are the predominant cells in periodontal tissues, we hypothesized that HGFs are contributed to receptors for components of bacteria. In this study, we focused on expression and function of nucleotide binding oligomerization domain 2 (NOD2) in HGFs, which is a mammalian cytosolic pathogen recognition molecule. MATERIAL AND METHODS Expression of NOD2 in HGFs was examined by reverse transcriptase-polymerase chain reaction (RT-PCR) and flow cytometry. Production of interleukin (IL)-6, IL-8, cc chemokine ligand2, cxc chemokine ligand10 (CXCL10) and CXCL11 from HGFs was examined by enzyme-linked immunosorbent assay (ELISA). We used RT-PCR and immunohistochemistry to detect the NOD2 expression in human gingival tissues. RESULTS We found clear NOD2 expression in HGFs. Upon stimulation with NOD2 agonist, muramyldipeptide (MDP), production of proinflammatory cytokines was enhanced. Moreover, MDP-induced production of proinflammatory cytokines was inhibited in a different manner by mitogen-activated protein kinase inhibitors and phosphatidylinositol 3-kinase inhibitor. Furthermore, MDP enhanced CXCL10 and CXCL11 productions by tumor necrosis factor-alpha (TNF-alpha)- or interferon-gamma (IFN-gamma)-stimulated HGFs, although MDP alone did not induce these chemokines. TNF-alpha and IFN-gamma increased NOD2 expression in HGFs. In addition, we detected NOD2 expression in mononuclear cells and HGFs in periodontally diseased tissues. CONCLUSION These findings indicate that MDP which induces production of cytokines and chemokines from HGFs is related to the pathogenesis of periodontal disease.
Collapse
Affiliation(s)
- I Hosokawa
- Department of Conservative Dentistry, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Tokushima, Japan.
| | | | | | | | | | | |
Collapse
|
31
|
Kaur J, Reddy K, Balakumar P. The Novel Role of Fenofibrate in Preventing Nicotine- and Sodium Arsenite-Induced Vascular Endothelial Dysfunction in the Rat. Cardiovasc Toxicol 2010; 10:227-38. [DOI: 10.1007/s12012-010-9086-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
32
|
Shan L, Redhu NS, Saleh A, Halayko AJ, Chakir J, Gounni AS. Thymic stromal lymphopoietin receptor-mediated IL-6 and CC/CXC chemokines expression in human airway smooth muscle cells: role of MAPKs (ERK1/2, p38, and JNK) and STAT3 pathways. THE JOURNAL OF IMMUNOLOGY 2010; 184:7134-43. [PMID: 20483734 DOI: 10.4049/jimmunol.0902515] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Thymic stromal lymphopoietin (TSLP) plays a pivotal role in allergic diseases such as asthma, chronic obstructive pulmonary disease, and atopic dermatitis. Enhanced TSLP expression has been detected in asthmatic airways that correlated with both the expression of Th2-attracting chemokines and with disease severity. Although cumulative evidence suggests that human airway smooth muscle (HASM) cells can initiate or perpetuate the airway inflammation by secreting a variety of inflammatory cell products such as cytokines and chemokines, the role of TSLP in this pathway is not known. In the current study, we sought to investigate whether HASM cells express the TSLP receptor (TSLPR) and whether it is functional. We first demonstrated that primary HASM cells express the transcript and protein of both TSLPR subunits (TSLPR and IL-7Ralpha). Functionally, TSLPR-mediated HASM activation induced a significant increase in CXC (IL-8/CXCL8), CC (eotaxin-1/CCL11) chemokines, and proinflammatory cytokine IL-6 expression. Furthermore, using biochemical and genetic approaches, we found that TSLP-induced proinflammatory gene expression in HASM involved the transcriptional mechanisms, MAPKs (ERK1/2, p38, and JNK), and STAT3 activation. Finally, TSLPR immunoreactivity in bronchial sections from mild allergic asthmatics suggested the potential in vivo TSLP targeting of HASM. Altogether, our data suggest that the TSLPR-mediated HASM activation induces proinflammatory cytokine and chemokines release that may facilitate inflammatory immune cells recruitment in airways. In addition, it may be inferred that TSLPR is involved in the pathogenesis of allergic asthma through the activation of HASM cells by TSLP.
Collapse
Affiliation(s)
- Lianyu Shan
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | | | | | | | |
Collapse
|
33
|
Abstract
We have previously identified αvβ3 and Fas as receptors for the streptococcal pyrogenic exotoxin B (SPE B), and G308S, a mutant of SPE B with RSD motif, which interacts with Fas only. This study aims to evaluate how SPE B interacts with cells to induce the production of IL-8. Our results showed that following exposure to SPE B or G308S, the levels of IL-8 protein and mRNA were increased and the increase was inhibited by the addition of anti-Fas antibody, suggesting that the increased production of IL-8 by SPE B is mediated through Fas receptor. In the presence of G308S, the association of FADD and procaspase 8, and activation of NF-κB were also detected. The application of siRNA of FADD and of procaspase 8 could inhibit the NF-κB activity. The proteolytic activity of caspase 8 was required for the NF-κB activity. Further studies showed that G308S could increase the phosphorylation of ERK and the translocation of NF-κB into the nucleus, and the inhibition of ERK phosphorylation decreased the IL-8 production, mRNA expression and activation of NF-κB. In addition, siRNA of procaspase 8 could inhibit the G308S-induced cleavage of MEKK1, binding of MEKK1 to caspase 8, activation of ERK and the NF-κB activity. Taken together, the production of IL-8 by SPE B in A549 cells is mediated by Fas, and followed by the activation of FADD, caspase 8, MEKK1, ERK and NF-κB.
Collapse
|
34
|
Rao SP, Sikora L, Hosseinkhani MR, Pinkerton KE, Sriramarao P. Exposure to environmental tobacco smoke induces angiogenesis and leukocyte trafficking in lung microvessels. Exp Lung Res 2009; 35:119-35. [PMID: 19263281 DOI: 10.1080/01902140802449729] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Exposure to environmental tobacco smoke (ETS) is known to contribute to and exacerbate inflammatory diseases of the lung such as chronic obstructive pulmonary disease (COPD) and asthma. The effect of ETS on angiogenesis and leukocyte recruitment, both of which promote lung inflammation, was investigated using lung tissue from mice exposed to aged and diluted sidestream cigarette smoke or fresh air for 12 weeks and transplanted into dorsal skin-fold chambers in nude mice. Lung tissue from mice exposed to cigarette smoke for 12 weeks exhibited significantly increased vascular density (angiogenesis) associated with selectin-mediated increased intravascular leukocyte rolling and adhesion compared to fresh air-exposed lung tissue by intravital microscopy. Further, neutrophils from nicotine-exposed mice displayed significantly increased rolling and adhesion compared to control neutrophils in microvessels of nicotine-exposed lungs versus control lung microvessels, suggesting that nicotine in cigarette smoke can augment leukocyte-endothelial interactions. ETS-induced angiogenesis and leukocyte trafficking may play a key role in airway recruitment of inflammatory cells in ETS-associated disorders such as COPD bronchitis or asthma.
Collapse
Affiliation(s)
- Savita P Rao
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota 55108, USA
| | | | | | | | | |
Collapse
|
35
|
Suri SS, Rakotondradany F, Myles AJ, Fenniri H, Singh B. The role of RGD-tagged helical rosette nanotubes in the induction of inflammation and apoptosis in human lung adenocarcinoma cells through the P38 MAPK pathway. Biomaterials 2009; 30:3084-90. [DOI: 10.1016/j.biomaterials.2009.02.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Accepted: 02/03/2009] [Indexed: 12/14/2022]
|
36
|
PKC isoenzymes differentially modulate the effect of thrombin on MAPK-dependent RPE proliferation. Biosci Rep 2009; 28:307-17. [PMID: 18636965 DOI: 10.1042/bsr20080083] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Thrombin signalling through PAR (protease-activated receptor)-1 is involved in cellular processes, such as proliferation, differentiation and cell survival. Following traumatic injury to the eye, thrombin signalling may participate in disorders, such as PVR (proliferative vitreoretinopathy), a human eye disease characterized by the uncontrolled proliferation, transdifferentiation and migration of otherwise quiescent RPE (retinal pigment epithelium) cells. PARs activate the Ras/Raf/MEK/ERK MAPK pathway (where ERK is extracellular-signal-regulated kinase, MAPK is mitogen-activated protein kinase and MEK is MAPK/ERK kinase) through the activation of G(alpha) and G(betagamma) heterotrimeric G-proteins, and the downstream stimulation of the PLC (phospholipase C)-beta/PKC (protein kinase C) and PI3K (phosphoinositide 3-kinase) signalling axis. In the present study, we examined the molecular signalling involved in thrombin-induced RPE cell proliferation, using rat RPE cells in culture as a model system for PVR pathogenesis. Our results showed that thrombin activation of PAR-1 induces RPE cell proliferation through Ras-independent activation of the Raf/MEK/ERK1/2 MAPK signalling cascade. Pharmacological analysis revealed that the activation of 'conventional' PKC isoforms is essential for proliferation, although thrombin-induced phosphorylation of ERK1/2 requires the activation of atypical PKCzeta by PI3K. Consistently, thrombin-induced ERK1/2 activation and RPE cell proliferation were prevented completely by PI3K or PKCzeta inhibition. These results suggest that thrombin induces RPE cell proliferation by joint activation of PLC-dependent and atypical PKC isoforms and the Ras-independent downstream stimulation of the Raf/MEK/ERK1/2 MAPK cascade. The present study is the first report demonstrating directly thrombin-induced ERK phosphorylation in the RPE, and the involvement of atypical PKCzeta in this process.
Collapse
|
37
|
Arayatrakoollikit U, Pavasant P, Yongchaitrakul T. Thrombin induces osteoprotegerin synthesis via phosphatidylinositol 3'-kinase/mammalian target of rapamycin pathway in human periodontal ligament cells. J Periodontal Res 2008; 43:537-43. [PMID: 18565131 DOI: 10.1111/j.1600-0765.2007.01071.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND OBJECTIVE Thrombin influences the biological behavior of periodontal ligament cells and plays multiple roles in the early stages of bone healing. Osteoprotegerin (OPG) is one of the key molecules that regulate bone homeostasis and prevent osteoclastogenesis. The purpose of this study was to evaluate the biological effects of thrombin on OPG synthesis in human periodontal ligament (HPDL) cells in vitro. MATERIAL AND METHODS Cells were treated with various concentrations (0.001, 0.01 and 0.1 U/mL) of thrombin. The mRNA expression and protein synthesis of OPG, as well as of receptor activator of nuclear factor kappaB ligand (RANKL), were determined by reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blot analysis, respectively. The influence of thrombin on OPG synthesis and its signaling pathway were investigated using inhibitors. RESULTS Thrombin profoundly induces protein synthesis of OPG at 0.1 U/mL. The inductive effect was inhibited by cycloheximide, but not by indomethacin. The phosphatidylinositol 3'-kinase (PI3K) inhibitor, LY294002, and the mammalian target of rapamycin (mTOR) inhibitor, rapamycin, exerted an inhibitory effect on the thrombin-induced OPG synthesis. In addition, the effect was inhibited by protease-activated receptor (PAR)-1 antagonist. Activation of phospho-Akt (p-Akt) was observed and the effect was abolished by LY294002. CONCLUSION Thrombin induces OPG synthesis in HPDL cells post-transcriptionally, possibly through PAR-1. The regulation was through the PI3K/Akt and mTOR pathway. This finding suggests that thrombin may play a significant role in alveolar bone repair and homeostasis of periodontal tissue, partly through the OPG/RANKL system.
Collapse
Affiliation(s)
- U Arayatrakoollikit
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | | | | |
Collapse
|
38
|
Ichiki T, Jougasaki M, Setoguchi M, Imamura J, Nakashima H, Matsuoka T, Sonoda M, Nakamura K, Minagoe S, Tei C. Cardiotrophin-1 stimulates intercellular adhesion molecule-1 and monocyte chemoattractant protein-1 in human aortic endothelial cells. Am J Physiol Heart Circ Physiol 2008; 294:H750-63. [DOI: 10.1152/ajpheart.00161.2007] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Intercellular adhesion molecule-1 (ICAM-1) and monocyte chemoattractant protein-1 (MCP-1) play critical roles in mediating monocyte adhesion to the vascular endothelium and monocyte migration into the subendothelial regions of the vessels. Inasmuch as cardiotrophin-1 (CT-1), an IL-6-type cytokine, was expressed in human atherosclerotic plaque, we examined whether CT-1 induces monocyte adhesion and migration by stimulating gene and protein expressions of ICAM-1 and MCP-1 in human aortic endothelial cells (HAECs). Immunocytochemistry revealed that CT-1 increased intensity of ICAM-1 and MCP-1 immunoreactivity in HAECs. Adhesion assay and chemotaxis assay revealed that CT-1 increased human monocytic THP-1 cell adhesion to HAECs and promoted chemotaxis in THP-1 cells, which were attenuated by anti-ICAM-1 and anti-MCP-1 antibody, respectively. Western blot analysis showed that CT-1 increased phosphorylation of ERK1/2 MAP kinase, p38 MAP kinase, and Akt and that their inhibitors, PD-98059, SB-203580, and LY-294002, respectively, inhibited phosphorylation. RNase protection assay and ELISA demonstrated that CT-1 increased gene and protein expressions of ICAM-1 and MCP-1. EMSA revealed that CT-1 enhanced NF-κB DNA-binding activity. CT-1-mediated upregulation of ICAM-1 and MCP-1 was suppressed by PD-98059, SB-203580, LY-294002, and parthenolide. The present study demonstrates that CT-1 promotes monocyte adhesion and migration by stimulating ICAM-1 and MCP-1 through mechanisms that involve ERK1/2 MAP kinase, p38 MAP kinase, phosphatidylinositol 3-kinase, and NF-κB pathways and suggests that CT-1 plays an important role in the pathophysiology of vascular inflammation and atherosclerosis.
Collapse
|
39
|
Ostrowska E, Reiser G. Protease-activated receptor (PAR)-induced interleukin-8 production in airway epithelial cells requires activation of MAP kinases p44/42 and JNK. Biochem Biophys Res Commun 2008; 366:1030-5. [DOI: 10.1016/j.bbrc.2007.12.064] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2007] [Accepted: 12/10/2007] [Indexed: 11/17/2022]
|
40
|
Niu QX, Chen HQ, Chen ZY, Fu YL, Lin JL, He SH. Induction of inflammatory cytokine release from human umbilical vein endothelial cells by agonists of proteinase-activated receptor-2. Clin Exp Pharmacol Physiol 2008; 35:89-96. [PMID: 18047634 DOI: 10.1111/j.1440-1681.2007.04755.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
1. Human endothelial cells express proteinase-activated receptor-2 (PAR-2), inflammatory cytokines and trypsin (EC 3.4.21.4). However, little is known about the mechanism through which trypsin induces cytokine release from endothelial cells. 2. In the present study, we investigated the effect of trypsin on cytokine release from primary cultures of human umbilical vein endothelial cells (HUVEC) using an antibody based protein microarray and ELISA. 3. The results showed that 1 microg/mL trypsin induced release of 32 different inflammatory factors, whereas 100 micromol/L Ser-Leu-Ile-Gly-Lys-Val-NH2 (SLIGKV-NH2) only stimulated secretion of 16 inflammatory factors from HUVEC, as assessed by an antibody based protein microarray. Because the release of interleukin (IL)-1a, IL-8, IL-10 and IL-12 was markedly increased following PAR-2 activation, their release was investigated further using ELISA. Increases in release of up to approximately 4.8-, 4.3-, 4.1- and 1.8-fold were observed for IL-1a, IL-10, IL-12 and IL-8, respectively, when HUVEC were challenged with trypsin for 16 h. Agonist peptides of PAR-2, namely SLIGKV-NH2 and trans-cinnamoyl-Leu-Ile-Gly-Arg-Leu-Orn-NH2 (tc-LIGRLO-NH2), also provoked significant release of IL-8. Trypsin-induced cytokine release was inhibited by its inhibitors soybean trypsin inhibitor, alpha1-antitrypsin and the inhibitor peptide of PAR-2 Phe-Ser-Leu-Leu-Arg-Tyr-NH2 (FSLLRY-NH2). 4. These data indicate the action of trypsin on HUVEC is most likely through activation of PAR-2, suggesting that PAR-2-related mechanisms are involved in the inflammatory process in humans.
Collapse
Affiliation(s)
- Qing-Xia Niu
- Allergy and Inflammation Research Institute, Key Immunopharmacology Laboratory of Guangdong Province, Shantou University Medical College, Shantou, Guangdong, China
| | | | | | | | | | | |
Collapse
|
41
|
Cirillo P, Pacileo M, De Rosa S, Calabrò P, Gargiulo A, Angri V, Prevete N, Fiorentino I, Ucci G, Sasso L, Petrillo G, Musto D'Amore S, Chiariello M. HMG-CoA reductase inhibitors reduce nicotine-induced expression of cellular adhesion molecules in cultured human coronary endothelial cells. J Vasc Res 2007; 44:460-470. [PMID: 17657162 DOI: 10.1159/000106464] [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] [Received: 11/06/2006] [Accepted: 05/16/2007] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Smoking predisposes to the development of atherosclerosis and of its complications. The mechanisms responsible for these effects are not completely understood. We have investigated whether nicotine might promote a proatherosclerotic state in human coronary endothelial cells (HCAECs), studying the role of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors in preventing these phenomena. METHODS AND RESULTS Real-time PCR showed that nicotine induced a dose-dependent increase in mRNA levels for vascular cellular adhesion molecule-1 (VCAM-1)/intercellular adhesion molecule-1 (ICAM-1). Fluorescent-activated cell sorting analysis showed that nicotine induced expression of functionally active VCAM-1/ICAM-1, since they increased leukocyte adherence to HCAECs. Oxygen free radicals, Rho A and nuclear factor kappaB (NF-kappaB) play a pivotal role in modulating these effects. Indeed, nicotine caused oxygen free radical production as well as activation of Rho A and NF-kappaB pathways, evaluated by malondialdehyde levels, pulldown assay and by electrophoretic mobility shift assay, respectively. Superoxide dimutase, Rho A (Y-27639) and NF-kappaB inhibitors (pyrrolidine dithiocarbamate ammonium, Bay 11-7082) suppressed nicotine effects on CAM expression. HMG-CoA reductase inhibitors prevented these nicotine-mediated effects by inhibiting free radical generation and by modulating activation of Rho A and NF-kappaB pathways. CONCLUSIONS Nicotine promotes CAM expression on HCAECs, shifting them toward a proatherosclerotic state. These effects might explain, at least in part, the deleterious cardiovascular consequences of cigarette smoking. HMG-CoA reductase inhibitors play an important role in preventing these phenomena.
Collapse
Affiliation(s)
- Plinio Cirillo
- Division of Cardiology, University of Naples Federico II, Naples, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Induction of MMP-9 release from human dermal fibroblasts by thrombin: involvement of JAK/STAT3 signaling pathway in MMP-9 release. BMC Cell Biol 2007; 8:14. [PMID: 17480240 PMCID: PMC1876221 DOI: 10.1186/1471-2121-8-14] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2006] [Accepted: 05/07/2007] [Indexed: 02/05/2023] Open
Abstract
Background It has been recognized that dermal fibroblasts and matrix metalloproteases (MMP) play crucial roles in wound healing process in skin. Thrombin was found to stimulate IL-8 release from human dermal fibroblasts (HDFs). However, little is known of the effect of thrombin on secretion of MMPs from dermal fibroblasts. In the present study, the influence of thrombin on proMMP-2 and proMMP-9 activity release from primary cultured HDFs, and its potential signaling pathways were investigated. Results The results showed that thrombin induced proMMP-9, but not proMMP-2 release from HDFs in a dose dependent manner at 6 h following incubation. Thrombin also upregulated expression of proMMP-9 mRNA in HDFs. Hirudin completely abolished the action of thrombin on HDFs. An agonist peptide of protease-activated receptor-1, SFLLR-NH2 stimulated an enhanced release of proMMP-9 from HDFs. AG490, an inhibitor of STAT3 inhibited basal and thrombin-provoked proMMP-9 release and phosphorylation of STAT3. PD98059, an inhibitor of MAPK and LY294002, an inhibitor PI3K failed to significantly inhibit thrombin induced proMMP-9 release. Conclusion Thrombin is a potent stimulus of proMMP-9 release from HDFs. Thrombin induced proMMP-9 release is most likely through activation of PAR-1. JAK/STAT3 signaling pathway is involved in proMMP-9 release from HDFs.
Collapse
|