1
|
Gramstad OR, Schjalm C, Mollnes TE, Nielsen EW. Increased thromboinflammatory load in hereditary angioedema. Clin Exp Immunol 2023; 214:170-181. [PMID: 37561062 PMCID: PMC10714191 DOI: 10.1093/cei/uxad091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/05/2023] [Accepted: 08/09/2023] [Indexed: 08/11/2023] Open
Abstract
C1 inhibitor (C1Inh) is a serine protease inhibitor involved in the kallikrein-kinin system, the complement system, the coagulation system, and the fibrinolytic system. In addition to the plasma leakage observed in hereditary angioedema (HAE), C1Inh deficiency may also affect these systems, which are important for thrombosis and inflammation. The aim of this study was to investigate the thromboinflammatory load in C1Inh deficiency. We measured 27 cytokines including interleukins, chemokines, interferons, growth factors, and regulators using multiplex technology. Complement activation (C4d, C3bc, and sC5b-C9/TCC), haemostatic markers (β-thromboglobulin (β-TG), thrombin-antithrombin complexes (TAT), prothrombin fragment 1 + 2 (F1 + 2), active plasminogen activator inhibitor-1 (PAI-1), and the neutrophil activation marker myeloperoxidase (MPO) were measured by enzyme immunoassays. Plasma and serum samples were collected from 20 patients with HAE type 1 or 2 in clinical remission and compared with 20 healthy age- and sex-matched controls. Compared to healthy controls, HAE patients had significantly higher levels of tumour necrosis factor (TNF), interleukin (IL)-1β, IL-2, IL-4, IL-6, IL-7, IL-9, IL-12, and IL-17A, chemokine ligand (CXCL) 8, chemokine ligand (CCL) 3, CCL4, IL-1 receptor antagonist (IL-1RA), granulocyte-macrophage colony-stimulating factor (GM-CSF), fibroblast growth factor (FGF) 2 and platelet-derived growth factor (PDGF)-BB. HAE patients also had higher levels of TAT and F1 + 2. Although granulocyte colony-stimulating factor (G-CSF), β-TG and PAI-1 were higher in HAE patients, the differences did not reach statistical significance after correction for multiple testing. In conclusion, C1Inh deficiency is associated with an increased baseline thromboinflammatory load. These findings may reflect that HAE patients are in a subclinical attack state outside of clinically apparent oedema attacks.
Collapse
Affiliation(s)
- Olav Rogde Gramstad
- Department of Dermatology and Venerology, Oslo University Hospital, Oslo, Norway
| | - Camilla Schjalm
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Tom Eirik Mollnes
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, University of Oslo, Oslo, Norway
- Research Laboratory, Nordland Hospital, Bodø, Norway
| | - Erik Waage Nielsen
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Laboratory, Nordland Hospital, Bodø, Norway
- Department of Anesthesia and Intensive Care Medicine, Nordland Hospital, Bodø, Norway
- Institute of Clinical Medicine, University of Tromsø, Tromsø, Norway
- Faculty of Nursing and Health Sciences, Nord University, Bodø, Norway
| |
Collapse
|
2
|
Zhang Y, Sun L, Wang X, Zhou Q. Integrative analysis of HASMCs gene expression profile revealed the role of thrombin in the pathogenesis of atherosclerosis. BMC Cardiovasc Disord 2023; 23:191. [PMID: 37046189 PMCID: PMC10091598 DOI: 10.1186/s12872-023-03211-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
We explored the effect of thrombin on human aortic smooth muscle cells (HASMCs) and further analyzed its role in the pathogenesis of atherosclerosis (AS). Thrombin-induced differentially expressed genes (DEGs) in HASMCs were identified by analyzing expression profiles from the GEO. Subsequently, enrichment analysis, GSEA, PPI network, and gene-microRNAs networks were interrogated to identify hub genes and associated pathways. Enrichment analysis results indicated that thrombin causes HASMCs to secrete various pro-inflammatory cytokines and chemokines, exacerbating local inflammatory response in AS. Moreover, we identified 9 HUB genes in the PPI network, which are closely related to the inflammatory response and the promotion of the cell cycle. Additionally, we found that thrombin inhibits lipid metabolism and autophagy of HASMCs, potentially contributing to smooth muscle-derived foam cell formation. Our study deepens a mechanistic understanding of the effect of thrombin on HASMCs and provides new insight into treating AS.
Collapse
Affiliation(s)
- Yichen Zhang
- The Second Hospital of Shandong University, Jinan, Shandong Province, China
- Jinan Central Hospital, Shandong University, Jinan, Shandong Province, China
| | - Lin Sun
- Jinan Central Hospital, Shandong University, Jinan, Shandong Province, China
| | - Xingsheng Wang
- Jinan Central Hospital, Shandong University, Jinan, Shandong Province, China
| | - Qingbo Zhou
- The Second Hospital of Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
3
|
Systemic Delivery of Clopidogrel Inhibits Neointimal Formation in a Mouse Vein Graft Model. J Cardiovasc Pharmacol 2022; 80:832-841. [PMID: 36027583 DOI: 10.1097/fjc.0000000000001361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 07/30/2022] [Indexed: 12/13/2022]
Abstract
ABSTRACT Clopidogrel inhibits platelet aggregation and has beneficial effects on patients undergoing coronary artery bypass grafting surgery, but it is unknown whether clopidogrel inhibits the neointima formation of grafted veins. In this study, we used a murine vein graft model to study the effect of clopidogrel on intima hyperplasia of the vein graft. Vein grafting was performed among C57BL/6J mice, immediately after surgery; 1 mg/kg clopidogrel and vehicle control were used to inject mice peritoneally daily for 2 weeks. As compared with the vehicle, clopidogrel significantly inhibited the neointima formation of vein grafts at 4 weeks after surgeries. The immunohistochemistry study showed that as compared with the vehicle, clopidogrel significantly decreased the rate of proliferating cell nuclear antigen-positive cells in the wall of vein grafts and significantly increased the expression of vascular smooth muscle cell (VSMC) contractile protein markers (α-smooth muscle actin, calponin, and SM22) within the neointima area of vein grafts. Clopidogrel significantly decreased the plasma interleukin 6 (IL-6) level at 1 week after surgery as compared with the vehicle. We isolated VSMCs from mouse aortic arteries. As compared with the vehicle, clopidogrel significantly inhibited thrombin-induced VSMC proliferation and migration, significantly decreased IL-6 mRNA expression and protein secretion, and increased intracellular cyclic adenosine monophosphate generation in a dose-dependent manner. In conclusion, systemic delivery of clopidogrel inhibits neointima formation of the mouse vein graft, the mechanisms of which are associated with its inhibitory effects on VSMC proliferation, migration, and the tendency to synthetic phenotype after vein graft surgery, reducing the expression of IL-6 and increasing the intracellular cyclic adenosine monophosphate level.
Collapse
|
4
|
Dukhin OA, Kalinsaya AI, Shpektor AV, Vasilieva EY. [The role of thrombin in the pathogenesis of atherosclerosis and its complications]. KARDIOLOGIIA 2022; 62:73-81. [PMID: 35414364 DOI: 10.18087/cardio.2022.3.n1968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/26/2022] [Indexed: 06/14/2023]
Abstract
Thrombin is a key regulator of the homeostasis system. Also, it actively participates in progression of various systemic diseases, including atherosclerosis. There is a large amount of experimental and clinical data on the involvement of thrombin in the pathogenesis of ischemic heart disease (IHD). Thus, studying thrombin activity regulation is promising. Also, the question whether it is possible to use biomarkers of thrombin activity as predictors of cardiovascular complications in IHD patients is relevant. The present review focuses on major mechanisms of thrombin functioning, its role in development and progression of atherosclerosis, and available tests for evaluation of thrombin functional activity. Major clinical studies are discussed that evaluated the efficacy of thrombin inhibitors and protease-activated receptor antagonists.
Collapse
Affiliation(s)
- O A Dukhin
- A.I. Yevdokimov Moscow State University of Medicine and Dentistry; Moscow Clinical City Hospital named after I.V. Davydovsky
| | - A I Kalinsaya
- A.I. Yevdokimov Moscow State University of Medicine and Dentistry; Moscow Clinical City Hospital named after I.V. Davydovsky
| | - A V Shpektor
- A.I. Yevdokimov Moscow State University of Medicine and Dentistry
| | - E Yu Vasilieva
- A.I. Yevdokimov Moscow State University of Medicine and Dentistry; Moscow Clinical City Hospital named after I.V. Davydovsky
| |
Collapse
|
5
|
Alexander ET, Gilmour SK. Immunomodulatory role of thrombin in cancer progression. Mol Carcinog 2022; 61:527-536. [PMID: 35338515 DOI: 10.1002/mc.23398] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 11/06/2022]
Abstract
Coagulation proteases and the generation of thrombin are increased in tumors. In addition, chemotherapeutic agents commonly used to treat malignant cancers can exacerbate cancer-associated thromboses. Thrombin can modify tumor cell behavior directly through the activation of protease-activated receptors (PAR) or indirectly by generating fibrin matrices. In addition to its role in generating fibrin to promote hemostasis, thrombin acts directly on multiple effector cells of the immune system impacting both acute and chronic inflammatory processes. Thrombin-mediated release of interleukin-6, tumor necrosis factor-α, and monocyte chemoattractant protein-1 leads to the accumulation of multiple tumor-infiltrating immunosuppressive cell populations including myeloid derived suppresser cells, M2-like macrophages, and T regulatory cells. Ablation of PAR-1 from the tumor microenvironment, but not the tumor, has been shown to dramatically reduce tumor growth and metastasis in multiple tumor models. Thrombin-activated platelets release immunosuppressive cytokines including transforming growth factor-β that can inhibit natural killer cell activity, helping tumor cells to evade host immunosurveillance. Taken together, there is strong evidence that thrombin influences cancer progression via multiple mechanisms, including the tumor immune response, with thrombin emerging as a target for novel therapeutic strategies for cancer.
Collapse
Affiliation(s)
- Eric T Alexander
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania, USA
| | - Susan K Gilmour
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania, USA
| |
Collapse
|
6
|
An outcome analysis of predictive factors for portal or splenic vein thrombosis after distal pancreatectomy. Surg Today 2020; 50:1282-1289. [PMID: 32346760 DOI: 10.1007/s00595-020-02004-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/05/2020] [Indexed: 02/07/2023]
Abstract
PURPOSES The aim of this study was to explore predictive factors for portal or splenic vein thrombosis (VT) that might cause serious problems after distal pancreatectomy (DP). METHODS A total of 230 patients who underwent DP between 2008 and 2017 were retrospectively reviewed to identify predictive factors for portal or splenic VT. RESULTS Ultimately, 164 patients were analyzed. Portal or splenic VT was significantly correlated with age < 65 years old, benign tumor, laparoscopic surgery, preservation of the inferior mesenteric vein (IMV) and left gastric vein (LGV), preservation of the IMV only, no drainage vein, length of the residual splenic vein (RSV) ≥ 26 mm, vessel dissection with a linear stapler, and intra-abdominal abscess (all P < 0.05). Furthermore, a multivariate analysis indicated that the length of the RSV (odds ratio [OR]: 9.15, P = 0.03) was an independent predictive factor for portal VT and that the length of the RSV (OR: 37.9, P < 0.01), vessel dissection with a linear stapler (OR: 6.49, P = 0.03), and intra-abdominal abscess (OR: 23.0, P = 0.02) were independent predictive factors for splenic VT. CONCLUSION As the length of the RSV was significantly associated with portal or splenic VT, a follow-up imaging diagnosis might be recommended for such cases.
Collapse
|
7
|
Lordan R, Tsoupras A, Zabetakis I. Platelet activation and prothrombotic mediators at the nexus of inflammation and atherosclerosis: Potential role of antiplatelet agents. Blood Rev 2020; 45:100694. [PMID: 32340775 DOI: 10.1016/j.blre.2020.100694] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 03/22/2020] [Accepted: 04/07/2020] [Indexed: 12/20/2022]
Abstract
Platelets are central to inflammation-related manifestations of cardiovascular diseases (CVD) such as atherosclerosis. Platelet-activating factor (PAF), thrombin, thromboxane A2 (TxA2), and adenosine diphosphate (ADP) are some of the key agonists of platelet activation that are at the intersection between a plethora of inflammatory pathways that modulate pro-inflammatory and coagulation processes. The aim of this article is to review the role of platelets and the relationship between their structure, function, and the interactions of their constituents in systemic inflammation and atherosclerosis. Antiplatelet therapies are discussed with a view to primary prevention of CVD by the clinical reduction of platelet reactivity and inflammation. Current antiplatelet therapies are effective in reducing cardiovascular risk but increase bleeding risk. Novel therapeutic antiplatelet approaches beyond current pharmacological modalities that do not increase the risk of bleeding require further investigation. There is potential for specifically designed nutraceuticals that may become safer alternatives to pharmacological antiplatelet agents for the primary prevention of CVD but there is serious concern over their efficacy and regulation, which requires considerably more research.
Collapse
Affiliation(s)
- Ronan Lordan
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute (HRI), University of Limerick, Limerick, Ireland; Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-5158, USA.
| | - Alexandros Tsoupras
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute (HRI), University of Limerick, Limerick, Ireland
| | - Ioannis Zabetakis
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute (HRI), University of Limerick, Limerick, Ireland
| |
Collapse
|
8
|
Libby P, Hansson GK. From Focal Lipid Storage to Systemic Inflammation: JACC Review Topic of the Week. J Am Coll Cardiol 2019; 74:1594-1607. [PMID: 31537270 PMCID: PMC6910128 DOI: 10.1016/j.jacc.2019.07.061] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 06/12/2019] [Accepted: 07/07/2019] [Indexed: 12/24/2022]
Abstract
Concepts of atherogenesis have evolved considerably with time. Early animal experiments showed that a cholesterol-rich diet could induce fatty lesion formation in arteries. The elucidation of lipoprotein metabolism ultimately led to demonstrating the clinical benefits of lipid lowering. The view of atheromata as bland accumulations of smooth muscle cells that elaborated an extracellular matrix that could entrap lipids then expanded to embrace inflammation as providing pathways that could link risk factors to atherogenesis. The characterization of leukocyte adhesion molecules and their control by proinflammatory cytokines, the ability of chemokines to recruit leukocytes, and the identification of inflammatory cell subtypes in lesions spurred the unraveling of innate and adaptive immune pathways that contribute to atherosclerosis and its thrombotic complications. Such pathophysiologic insights have led to the identification of biomarkers that can define categories of risk and direct therapies and to the development of new treatments.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Göran K Hansson
- Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
9
|
Fabjan A, Bajrović FF. Novel Direct Anticoagulants and Atherosclerosis. Curr Vasc Pharmacol 2018; 17:29-34. [PMID: 29412112 DOI: 10.2174/1570161116666180206111217] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 09/13/2017] [Accepted: 11/07/2017] [Indexed: 12/31/2022]
Abstract
Coagulation factors can affect cellular processes that include inflammatory signaling by acting on endothelial protease activated receptors, vascular smooth muscle and inflammatory cells beyond the coagulation cascade. This is important in the pathogenesis of atherosclerosis. Accordingly, experimental data points to beneficial effects of coagulation protease inhibitors on the attenuation of atherosclerosis progression in animal models. However, available clinical data do not support the use of anticoagulants as an add-on treatment of atherosclerosis. New clinical studies are needed with a better selection of patients to clarify the role of novel direct anticoagulants in the management of atherosclerosis.
Collapse
Affiliation(s)
- Andrej Fabjan
- Department of Vascular Neurology and Intensive Care, Neurological Clinic, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Institute of Physiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Fajko F Bajrović
- Department of Vascular Neurology and Intensive Care, Neurological Clinic, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
10
|
Poterucha TJ, Libby P, Goldhaber SZ. More than an anticoagulant: Do heparins have direct anti-inflammatory effects? Thromb Haemost 2017; 117:437-444. [DOI: 10.1160/th16-08-0620] [Citation(s) in RCA: 178] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/20/2016] [Indexed: 11/05/2022]
Abstract
SummaryThe heparins, well-known for their anticoagulant properties, may also have anti-inflammatory effects that could contribute to their effectiveness in the treatment of venous thromboembolism and other vascular diseases. This review focuses on the inflammatory pathophysiology that underlies the development of thrombosis and the putative effects of heparin on these pathways. We present evidence supporting the use of heparin for other indications, including autoimmune disease, malignancy, and disseminated intravascular coagulation. These considerations highlight the need for further research to elucidate the mechanisms of the possible pleiotropic effects of the heparins, with a view to advancing treatments based upon heparin derivatives.
Collapse
|
11
|
Proteolytic signatures define unique thrombin-derived peptides present in human wound fluid in vivo. Sci Rep 2017; 7:13136. [PMID: 29030565 PMCID: PMC5640616 DOI: 10.1038/s41598-017-13197-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/19/2017] [Indexed: 01/13/2023] Open
Abstract
The disease burden of failing skin repair and non-healing ulcers is extensive. There is an unmet need for new diagnostic approaches to better predict healing activity and wound infection. Uncontrolled and excessive protease activity, of endogenous or bacterial origin, has been described as a major contributor to wound healing impairments. Proteolytic peptide patterns could therefore correlate and “report” healing activity and infection. This work describes a proof of principle delineating a strategy by which peptides from a selected protein, human thrombin, are detected and attributed to proteolytic actions. With a particular focus on thrombin-derived C-terminal peptides (TCP), we show that distinct peptide patterns are generated in vitro by the human S1 peptidases human neutrophil elastase and cathepsin G, and the bacterial M4 peptidases Pseudomonas aeruginosa elastase and Staphylococcus aureus aureolysin, respectively. Corresponding peptide sequences were identified in wound fluids from acute and non-healing ulcers, and notably, one peptide, FYT21 (FYTHVFRLKKWIQKVIDQFGE), was only present in wound fluid from non-healing ulcers colonized by P. aeruginosa and S. aureus. Our result is a proof of principle pointing at the possibility of defining peptide biomarkers reporting distinct proteolytic activities, of potential implication for improved diagnosis of wound healing and infection.
Collapse
|
12
|
Ebrahimi S, Rahmani F, Behnam-Rassouli R, Hoseinkhani F, Parizadeh MR, Keramati MR, Khazaie M, Avan A, Hassanian SM. Proinflammatory signaling functions of thrombin in cancer. J Cell Physiol 2017; 232:2323-2329. [PMID: 28004386 DOI: 10.1002/jcp.25753] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 12/20/2016] [Indexed: 12/19/2022]
Abstract
Thrombin-induced activation of protease-activated receptors (PARs) represents a link between inflammation and cancer. Proinflammatory signaling functions of thrombin are associated with several inflammatory diseases including neurodegenerative, cardiovascular, and of special interest in this review cancer. Thrombin-induced inflammatory responses up-regulates expression of cytokines, adhesion molecules, angiogenic factors, and matrix-degrading proteases that facilitate tumor cells proliferation, angiogenesis, invasion, and metastasis. This review summarizes the current knowledge about the mechanisms of thrombin-mediated proinflammatory responses in cancer pathology for a better understanding and hence a better management of this disease.
Collapse
Affiliation(s)
- Safieh Ebrahimi
- Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzad Rahmani
- Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Fatemeh Hoseinkhani
- Department of Medical Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Parizadeh
- Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Keramati
- Cancer Molecular Pathology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaie
- Department of Medical Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Molecular Medicine Group, Department of Modern Sciences and Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Microanatomy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
13
|
The Effect of Post-Resistance Exercise Amino Acids on Plasma MCP-1 and CCR2 Expression. Nutrients 2016; 8:nu8070409. [PMID: 27384580 PMCID: PMC4963885 DOI: 10.3390/nu8070409] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 06/22/2016] [Accepted: 06/29/2016] [Indexed: 12/22/2022] Open
Abstract
The recruitment and infiltration of classical monocytes into damaged muscle is critical for optimal tissue remodeling. This study examined the effects of an amino acid supplement on classical monocyte recruitment following an acute bout of lower body resistance exercise. Ten resistance-trained men (24.7 ± 3.4 years; 90.1 ± 11.3 kg; 176.0 ± 4.9 cm) ingested supplement (SUPP) or placebo (PL) immediately post-exercise in a randomized, cross-over design. Blood samples were obtained at baseline (BL), immediately (IP), 30-min (30P), 1-h (1H), 2-h (2H), and 5-h (5H) post-exercise to assess plasma concentrations of monocyte chemoattractant protein 1 (MCP-1), myoglobin, cortisol and insulin concentrations; and expressions of C-C chemokine receptor-2 (CCR2), and macrophage-1 antigen (CD11b) on classical monocytes. Magnitude-based inferences were used to provide inferences on the true effects of SUPP compared to PL. Changes in myoglobin, cortisol, and insulin concentrations were similar between treatments. Compared to PL, plasma MCP-1 was “very likely greater” (98.1% likelihood effect) in SUPP at 2H. CCR2 expression was “likely greater” at IP (84.9% likelihood effect), “likely greater” at 1H (87.7% likelihood effect), “very likely greater” at 2H (97.0% likelihood effect), and “likely greater” at 5H (90.1% likelihood effect) in SUPP, compared to PL. Ingestion of SUPP did not influence CD11b expression. Ingestion of an amino acid supplement immediately post-exercise appears to help maintain plasma MCP-1 concentrations and augment CCR2 expression in resistance trained men.
Collapse
|
14
|
Gupta GK, Agrawal T, Rai V, Del Core MG, Hunter WJ, Agrawal DK. Vitamin D Supplementation Reduces Intimal Hyperplasia and Restenosis following Coronary Intervention in Atherosclerotic Swine. PLoS One 2016; 11:e0156857. [PMID: 27271180 PMCID: PMC4894559 DOI: 10.1371/journal.pone.0156857] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/21/2016] [Indexed: 11/18/2022] Open
Abstract
Vitamin D is a fat-soluble steroid hormone that activates vitamin D receptor to regulate multiple downstream signaling pathways and transcription of various target genes. There is an association between vitamin D deficiency and increased risk for cardiovascular disease. However, most of the studies are observational and associative in nature with limited data on clinical application. Thus, there is a need for more prospective randomized controlled studies to determine whether or not vitamin D supplementation provides cardiovascular protection. In this study, we examined the effects of the deficiency and supplementation of vitamin D on coronary restenosis following coronary intervention in atherosclerotic Yucatan microswine. Twelve Yucatan microswine were fed vitamin D-deficient (n = 4) or -sufficient (n = 8) high cholesterol diet for 6-months followed by coronary intervention. Post-intervention, swine in the vitamin D-sufficient high cholesterol diet group received daily oral supplementation of either 1,000 IU (n = 4) or 3,000 IU (n = 4) vitamin D3. Six months later, optical coherence tomography (OCT) was performed to monitor the development of intimal hyperplasia and restenosis. Animals were euthanized to isolate arteries for histomorphometric and immunohistochemical studies. Animals had graded levels of serum 25(OH)D; vitamin D-deficient (15.33 ± 1.45 ng/ml), vitamin D-sufficient + 1,000 IU oral vitamin D post-intervention (32.27 ± 1.20 ng/ml), and vitamin D-sufficient + 3,000 IU oral vitamin D post-intervention (51.00 ± 3.47 ng/ml). Findings from the OCT and histomorphometric studies showed a decrease in intimal hyperplasia and restenosis in vitamin D-supplemented compared to vitamin D-deficient swine. Vitamin D supplementation significantly decreased serum levels of TNF-α and IFN-γ, upregulated serum levels of IL-10, and had no effect on serum IL-6 levels. These findings suggest that vitamin D supplementation limits neointimal formation following coronary intervention in atherosclerotic swine and provide the support for vitamin D supplementation to protect against the development of coronary restenosis.
Collapse
Affiliation(s)
- Gaurav K. Gupta
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States of America
| | - Tanupriya Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States of America
| | - Vikrant Rai
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States of America
| | - Michael G. Del Core
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States of America
| | - William J. Hunter
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States of America
| | - Devendra K. Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE, United States of America
- * E-mail:
| |
Collapse
|
15
|
Nakano M, Yahagi K, Otsuka F, Sakakura K, Finn AV, Kutys R, Ladich E, Fowler DR, Joner M, Virmani R. Causes of Early Stent Thrombosis in Patients Presenting With Acute Coronary Syndrome. J Am Coll Cardiol 2014; 63:2510-2520. [DOI: 10.1016/j.jacc.2014.02.607] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 02/18/2014] [Accepted: 02/24/2014] [Indexed: 12/01/2022]
|
16
|
El Amrani M, El Kharras A, Asserraji M. Partially thrombosed aneurysm of the abdominal aorta: Unusual cause of chronic inflammation and resistance to recombinant human erythropoietin. Indian J Nephrol 2014; 24:38-40. [PMID: 24574630 PMCID: PMC3927190 DOI: 10.4103/0971-4065.125069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Diagnosis of chronic inflammatory syndrome is often a challenge. In dialysis patients, this condition leads to resistance to recombinant human erythropoietin (rHuEPO). We here report a case of a 72-year-old male undergoing chronic hemodialysis and developed rHuEPO resistance without any obvious etiology. Investigations showed a partially thromosed aneurysm of the infrarenal aorta. Antiplatelet therapy was started with a satisfactory outcome.
Collapse
Affiliation(s)
- M El Amrani
- Department of Nephrology and Dialysis, First Medical and Surgical Center, Agadir, Morocco
| | - A El Kharras
- Department of Radiology, First Medical and Surgical Center, Agadir, Morocco
| | - M Asserraji
- Department of Nephrology and Dialysis, First Medical and Surgical Center, Agadir, Morocco
| |
Collapse
|
17
|
Nieuwenhuizen L, Falkenburg WJJ, Schutgens REG, Roosendaal G, van Veghel K, Biesma DH, Lafeber FPJG. Stimulation of naïve monocytes and PBMCs with coagulation proteases results in thrombin-mediated and PAR-1-dependent cytokine release and cell proliferation in PBMCs only. Scand J Immunol 2013; 77:339-49. [PMID: 23421536 DOI: 10.1111/sji.12033] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 01/29/2013] [Indexed: 11/26/2022]
Abstract
Protease-activated receptors (PARs) are stimulated by proteolytic cleavage of their extracellular domain. Coagulation proteases, such as FVIIa, the binary TF-FVIIa complex, free FXa, the ternary TF-FVIIa-FXa complex and thrombin, are able to stimulate PARs. Whereas the role of PARs on platelets is well known, their function in naïve monocytes and peripheral blood mononuclear cells (PBMCs) is largely unknown. This is of interest because PAR-mediated interactions of coagulation proteases with monocytes and PBMCs in diseases with an increased activation of coagulation may promote inflammation. To evaluate PAR-mediated inflammatory reactions in naïve monocytes and PBMCs stimulated with coagulation proteases. For this, PAR expression at protein and RNA level on naïve monocytes and PBMCs was evaluated with flow cytometry and RT-PCR. In addition, cytokine release (IL-1β, IL-6, IL-8, IL-10, TNF-α) in stimulated naïve and PBMC cell cultures was determined. In this study, it is demonstrated that naïve monocytes express all four PARs at the mRNA level, and PAR-1, -3 and -4 at the protein level. Stimulation of naïve monocytes with coagulation proteases did not result in alterations in PAR expression or in the induction of inflammation involved cytokines like interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-8, interleukin-10 or tumour necrosis factor-α. In contrast, stimulation of PBMCs with coagulation proteases resulted in thrombin-mediated induction of IL-1β and IL-6 cytokine production and PBMC cell proliferation in a PAR-1-dependent manner. These data demonstrate that naïve monocytes are not triggered by coagulation proteases, whereas thrombin is able to elicit pro-inflammatory events in a PAR-1-dependent manner in PBMCs.
Collapse
Affiliation(s)
- L Nieuwenhuizen
- Rheumatology & Clinical Immunology, University Medical Center, Utrecht, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
18
|
Fougerat A, Smirnova NF, Gayral S, Malet N, Hirsch E, Wymann MP, Perret B, Martinez LO, Douillon M, Laffargue M. Key role of PI3Kγ in monocyte chemotactic protein-1-mediated amplification of PDGF-induced aortic smooth muscle cell migration. Br J Pharmacol 2012; 166:1643-53. [PMID: 22251152 DOI: 10.1111/j.1476-5381.2012.01866.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Vascular smooth muscle cell (SMC) migration within the arterial wall is a crucial event in atherogenesis and restenosis. Monocyte chemotactic protein-1/CC-chemokine receptor 2 (MCP-1/CCR2) signalling is involved in SMC migration processes but the molecular mechanisms have not been well characterized. We investigated the role of PI3Kγ in SMC migration induced by MCP-1. EXPERIMENTAL APPROACHES A pharmacological PI3Kγ inhibitor, adenovirus encoding inactive forms of PI3Kγ and genetic deletion of PI3Kγ were used to investigate PI3Kγ functions in the MCP-1 and platelet-derived growth factor (PDGF) signalling pathway and migration process in primary aortic SMC. KEY RESULTS The γ isoform of PI3K was shown to be the major signalling molecule mediating PKB phosphorylation in MCP-1-stimulated SMC. Using a PI3Kγ inhibitor and an adenovirus encoding a dominant negative form of PI3Kγ, we demonstrated that PI3Kγ is essential for SMC migration triggered by MCP-1. PDGF receptor stimulation induced MCP-1 mRNA and protein accumulation in SMCs. Blockade of the MCP-1/CCR2 pathway or pharmacological inhibition of PI3Kγ reduced PDGF-stimulated aortic SMC migration by 50%. Thus PDGF promotes an autocrine loop involving MCP-1/CCR2 signalling which is required for PDGF-mediated SMC migration. Furthermore, SMCs isolated from PI3Kγ-deficient mice (PI3Kγ(-/-)), or mice expressing an inactive PI3Kγ (PI3Kγ(KD/KD)), migrated less than control cells in response to MCP-1 and PDGF. CONCLUSIONS AND IMPLICATIONS PI3Kγ is essential for MCP-1-stimulated aortic SMC migration and amplifies cell migration induced by PDGF by an autocrine/paracrine loop involving MCP-1 secretion and CCR2 activation. PI3Kγ is a promising target for the treatment of aortic fibroproliferative pathologies.
Collapse
|
19
|
Libby P, Roberts WC. Peter Libby, MD: a conversation with the editor. Am J Cardiol 2012; 110:741-60. [PMID: 22673635 DOI: 10.1016/j.amjcard.2012.04.058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 04/15/2012] [Indexed: 01/10/2023]
|
20
|
Bruce AC, Peirce SM. Exogenous thrombin delivery promotes collateral capillary arterialization and tissue reperfusion in the murine spinotrapezius muscle ischemia model. Microcirculation 2012; 19:143-54. [PMID: 21954923 DOI: 10.1111/j.1549-8719.2011.00138.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE We examined the effects of exogenously delivered thrombin on cell recruitment in skeletal muscle and the formation of new collateral arterioles in the microvasculature in response to ligation-induced ischemia. METHODS Thrombin or vehicle was locally applied to both ligated and nonoperated Balb/c spinotrapezius muscles, which were harvested after three or seven days, imaged using confocal microscopy, and analyzed. RESULTS Thrombin treatment resulted in accelerated arterialization of collateral capillaries and accelerated tissue reperfusion in ischemic muscles. Uninjured muscle treated with thrombin displayed increased vascular cell adhesion molecule 1 expression on arteriole and venule endothelium, increased expression of smooth muscle α-actin on capillary-sized vessels, increased infiltration by CD11b(+) leukocytes, and mast cell infiltration and degranulation. CONCLUSIONS Exogenous delivery of thrombin enhances microvascular collateral development in response to ischemic insult, and accelerates tissue reperfusion. Elicited responses from multiple cell types probably contribute to these effects.
Collapse
Affiliation(s)
- Anthony C Bruce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | | |
Collapse
|
21
|
Lee H, Hamilton JR. Physiology, pharmacology, and therapeutic potential of protease-activated receptors in vascular disease. Pharmacol Ther 2012; 134:246-59. [DOI: 10.1016/j.pharmthera.2012.01.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Accepted: 01/17/2012] [Indexed: 01/09/2023]
|
22
|
Maeda A, Ohta K, Ohta K, Nakayama Y, Hashida Y, Toma T, Saito T, Maruhashi K, Yachie A. Effects of antithrombin III treatment in vascular injury model of mice. Pediatr Int 2011; 53:747-753. [PMID: 21410592 DOI: 10.1111/j.1442-200x.2011.03350.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Balloon angioplasty has recently been adopted as an acceptable form of treatment for stenotic vessel lesions of congenital heart diseases. However, precise mechanisms of restenosis and thrombosis, which are the most common complications after these procedures, are unknown. METHODS We examined the effects of antithrombin III (ATIII) on inflammation, thrombus formation, and remodeling of vascular wall after guidewire-induced injury in the femoral artery of mice. ATIII or saline was administered as a bolus intravenous infusion before injury. RESULTS Seventy-two hours after injury, approximately half of the saline-treated vessels showed macroscopic thrombus formation. In contrast, no thrombi were seen in the arteries pretreated with ATIII. Significantly higher levels of inflammation were induced in the injured vessels than in the sham-operated controls, as determined by CD11b-positive cell density in the adventitial area. ATIII treatment resulted in marked reduction of inflammatory cell infiltration. Twenty-eight days after injury, similar levels of neointimal proliferation were found in the injured arteries in both groups. CONCLUSIONS Our results suggested that a high dose of ATIII may influence the sequelae of arterial injury by reducing mural thrombus formation and limiting the inflammatory reaction of the vessel wall without altering the process of vascular remodeling.
Collapse
Affiliation(s)
- Akiko Maeda
- Department of Pediatrics, Angiogenesis and Vascular Development, Graduate School of Medical Science, Kanazawa UniversityDepartment of Pediatrics, National Hospital Organization Kanazawa Medical Center, Kanazawa, Japan
| | - Kunio Ohta
- Department of Pediatrics, Angiogenesis and Vascular Development, Graduate School of Medical Science, Kanazawa UniversityDepartment of Pediatrics, National Hospital Organization Kanazawa Medical Center, Kanazawa, Japan
| | - Kazuhide Ohta
- Department of Pediatrics, Angiogenesis and Vascular Development, Graduate School of Medical Science, Kanazawa UniversityDepartment of Pediatrics, National Hospital Organization Kanazawa Medical Center, Kanazawa, Japan
| | - Yuko Nakayama
- Department of Pediatrics, Angiogenesis and Vascular Development, Graduate School of Medical Science, Kanazawa UniversityDepartment of Pediatrics, National Hospital Organization Kanazawa Medical Center, Kanazawa, Japan
| | - Yoko Hashida
- Department of Pediatrics, Angiogenesis and Vascular Development, Graduate School of Medical Science, Kanazawa UniversityDepartment of Pediatrics, National Hospital Organization Kanazawa Medical Center, Kanazawa, Japan
| | - Tomoko Toma
- Department of Pediatrics, Angiogenesis and Vascular Development, Graduate School of Medical Science, Kanazawa UniversityDepartment of Pediatrics, National Hospital Organization Kanazawa Medical Center, Kanazawa, Japan
| | - Takekatsu Saito
- Department of Pediatrics, Angiogenesis and Vascular Development, Graduate School of Medical Science, Kanazawa UniversityDepartment of Pediatrics, National Hospital Organization Kanazawa Medical Center, Kanazawa, Japan
| | - Keiko Maruhashi
- Department of Pediatrics, Angiogenesis and Vascular Development, Graduate School of Medical Science, Kanazawa UniversityDepartment of Pediatrics, National Hospital Organization Kanazawa Medical Center, Kanazawa, Japan
| | - Akihiro Yachie
- Department of Pediatrics, Angiogenesis and Vascular Development, Graduate School of Medical Science, Kanazawa UniversityDepartment of Pediatrics, National Hospital Organization Kanazawa Medical Center, Kanazawa, Japan
| |
Collapse
|
23
|
Anand SX, Viles-Gonzalez JF, Mahboobi SK, Heerdt PM. Bivalirudin utilization in cardiac surgery: shifting anticoagulation from indirect to direct thrombin inhibition. Can J Anaesth 2010; 58:296-311. [DOI: 10.1007/s12630-010-9423-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Accepted: 11/03/2010] [Indexed: 01/19/2023] Open
|
24
|
Gigante B, Bennet AM, Leander K, Vikström M, de Faire U. The interaction between coagulation factor 2 receptor and interleukin 6 haplotypes increases the risk of myocardial infarction in men. PLoS One 2010; 5:e11300. [PMID: 20585578 PMCID: PMC2891999 DOI: 10.1371/journal.pone.0011300] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2010] [Accepted: 06/03/2010] [Indexed: 01/26/2023] Open
Abstract
The aim of the study was to investigate if the interaction between the coagulation factor 2 receptor (F2R) and the interleukin 6 (IL6) haplotypes modulates the risk of myocardial infarction (MI) in the Stockholm Heart Epidemiology Program (SHEEP). Seven SNPs at the F2R locus and three SNPs at the IL6 locus were genotyped. Haplotypes and haplotype pairs (IL6*F2R) were generated. A logistic regression analysis was performed to analyze the association of the haplotypes and haplotype pairs with the MI risk. Presence of an interaction between the two haplotypes in each haplotype pair was calculated using two different methods: the statistical, on a multiplicative scale, which includes the cross product of the two factors into the logistic regression model; the biological, on an additive scale, which evaluates the relative risk associated with the joint presence of both factors. The ratio between the observed and the predicted effect of the joint exposure, the synergy index (S), indicates the presence of a synergy (S>1) or of an antagonism (S<1). None of the haplotypes within the two loci was associated with the risk of MI. Out of 22 different haplotype pairs, the haplotype pair 17 GGG*ADGTCCT was associated with an increased risk of MI with an OR (95%CI) of 1.58 (1.05-2.41) (p = 0.02) in the crude and an OR of 1.72 (1.11-2.67) (p = 0.01) in the adjusted analysis. We observed the presence of an interaction on a multiplicative scale with an OR (95%CI) of 2.24 (1.27-3.95) (p = 0.005) and a slight interactive effect between the two haplotypes on an additive scale with an OR (95%CI) of 1.56 (1.02-2.37) (p = 0.03) and S of 1.66 (0.89-31). In conclusion, our results support the hypothesis that the interaction between these two functionally related genes may influence the risk of MI and suggest new mechanisms involved in the genetic susceptibility to MI.
Collapse
Affiliation(s)
- Bruna Gigante
- Division of Cardiovascular Epidemiology, Institute of Environmental Medicine, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
25
|
Jiang R, Wang NP, Tanaka KA, Levy JH, Guyton RA, Zhao ZQ, Vinten-Johansen J. Factor Xa induces tissue factor expression in endothelial cells by P44/42 MAPK and NF-κB-dependent pathways. J Surg Res 2010; 169:319-27. [PMID: 20451919 DOI: 10.1016/j.jss.2010.01.041] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Revised: 01/17/2010] [Accepted: 01/25/2010] [Indexed: 11/25/2022]
Abstract
BACKGROUND Tissue factor (TF) is an initiator of coagulation. The serine protease factor Xa (FXa) is the convergence point of the extrinsic and intrinsic components of the coagulation cascade. In addition to its hemostatic function, FXa elicits inflammatory responses in endothelial cells that may be important in surgical procedures in which inflammation is triggered. This study tested the hypothesis that FXa can up-regulate TF on vascular endothelial cells by a mitogen-activated protein kinase (MAPK)- and NF-κB-dependent pathway. METHODS AND RESULTS Incubation of cultured human umbilical vein endothelial cells (HUVECs) with FXa increased TF protein expression and activity in a dose-dependent manner. Pre-incubation of HUVECs with the serine protease inhibitor antithrombin, which targets not only thrombin but also FXa and FIXa, inhibited FXa-induced TF expression, but the selective thrombin inhibitor hirudin did not inhibit FXa-induced TF expression, ruling out a thrombin-mediated pathway. After 10 min incubation with HUVECs, FXa rapidly induced P44/42 MAPK activation (immunoblotting of phosphorylated P44/42 MAPK) with a peak at 30 min. The MEK 1/2 inhibitor PD98059 partially reduced FXa-induced TF expression and activity (3.82 ± 0.11 vs 6.54 ± 0.08 fmol/min/cm(2), P < 0.05). NF-κB was activated by FXa, confirmed by cytoplasmic IkBα degradation and increased NF-κB P65 nuclear translocation. Interruption of the NF-κB pathway by the IkBα phosphorylation inhibitor Bay 11-7802 abrogated FXa-induced TF protein expression and activity (1.93 ± 0.02 versus 6.54 ± 0.08 fmol/min/cm(2), P < 0.05). However, inhibition of PI3 kinase by LY 294002 did not attenuate FXa-induced TF protein expression and activity. CONCLUSIONS (1) FXa up-regulates TF protein expression and activity in HUVECs, (2) FXa-induced up-regulation of TF is independent of the thrombin-PAR1 pathway, and (3) the MAPK and NF-κB pathways, but not PI3 kinase pathway, are involved in FXa-induced TF expression on human umbilical endothelial cells. FXa may be a feed-forward alternative mechanism of activating TF expression and activity, thereby increasing a procoagulant state or inflammation. This mechanism may be important in the pro-inflammatory state initiated by cardiac surgical procedures.
Collapse
Affiliation(s)
- Rong Jiang
- Cardiothoracic Research Laboratory, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University Hospital Midtown, Atlanta, Georgia, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Hao F, Tan M, Wu DD, Xu X, Cui MZ. LPA induces IL-6 secretion from aortic smooth muscle cells via an LPA1-regulated, PKC-dependent, and p38alpha-mediated pathway. Am J Physiol Heart Circ Physiol 2009; 298:H974-83. [PMID: 20044439 DOI: 10.1152/ajpheart.00895.2009] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Lysophosphatidic acid (LPA) is a potent bioactive lysophospholipid. Accumulated evidence supports a role for LPA in inflammation. To profile LPA-induced cytokine production in vascular smooth muscle cells (SMCs), we used a cytokine antibody array system and found that LPA prominently induces the secretion of IL-6 and monocyte chemoattractant protein (MCP)-1 from human aortic SMCs (HASMCs). The mechanism by which LPA induces MCP-1 expression in SMCs has been previously reported. However, LPA induction of IL-6 secretion from vascular SMCs and its regulatory mechanism are unknown. The present study reveals that LPA induces the expression of IL-6 mRNA and protein in HASMCs as well as the secretion of IL-6 protein in a time-dependent manner. Our results demonstrate that LPA-specific receptor 1 (LPA(1)) mediates LPA-induced IL-6 secretion and that LPA induction of IL-6 is independent of the EGF receptor pathway. Our data further show that PKC-mediated p38 MAPK is responsible for the IL-6 secretion. Finally, small interfering RNA depletion experiments revealed that p38alpha is specifically responsible for the LPA-induced IL-6 secretion. The present study profiles the regulatory relationship between LPA and multiple cytokines in vascular SMCs for the first time, provides the first evidence that LPA upregulates IL-6 in vascular SMCs, and reveals the regulatory mechanism of LPA-induced IL-6 production in HASMCs. In light of the emerging roles of LPA and IL-6 in vascular inflammation, the understanding of the regulatory mechanism may contribute to the treatment and prevention of cardiovascular disorders.
Collapse
Affiliation(s)
- Feng Hao
- Department of Pathobiology, The University of Tennessee College of Veterinary Medicine, Knoxville, 37996, USA
| | | | | | | | | |
Collapse
|
27
|
Sprague AH, Khalil RA. Inflammatory cytokines in vascular dysfunction and vascular disease. Biochem Pharmacol 2009; 78:539-52. [PMID: 19413999 PMCID: PMC2730638 DOI: 10.1016/j.bcp.2009.04.029] [Citation(s) in RCA: 924] [Impact Index Per Article: 61.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2009] [Revised: 04/23/2009] [Accepted: 04/27/2009] [Indexed: 12/12/2022]
Abstract
The vascular inflammatory response involves complex interaction between inflammatory cells (neutrophils, lymphocytes, monocytes, macrophages), endothelial cells (ECs), vascular smooth muscle cells (VSMCs), and extracellular matrix (ECM). Vascular injury is associated with increased expression of adhesion molecules by ECs and recruitment of inflammatory cells, growth factors, and cytokines, with consequent effects on ECs, VSMCs and ECM. Cytokines include tumor necrosis factors, interleukins, lymphokines, monokines, interferons, colony stimulating factors, and transforming growth factors. Cytokines are produced by macrophages, T-cells and monocytes, as well as platelets, ECs and VSMCs. Circulating cytokines interact with specific receptors on various cell types and activate JAK-STAT, NF-kappaB, and Smad signaling pathways leading to an inflammatory response involving cell adhesion, permeability and apoptosis. Cytokines also interact with mitochondria to increase the production of reactive oxygen species. Cytokine-induced activation of these pathways in ECs modifies the production/activity of vasodilatory mediators such as nitric oxide, prostacyclin, endothelium-derived hyperpolarizing factor, and bradykinin, as well as vasoconstrictive mediators such as endothelin and angiotensin II. Cytokines interact with VSMCs to activate Ca(2+), protein kinase C, Rho-kinase, and MAPK pathways, which promote cell growth and migration, and VSM reactivity. Cytokines also interact with integrins and matrix metalloproteinases (MMPs) and modify ECM composition. Persistent increases in cytokines are associated with vascular dysfunction and vascular disease such as atherosclerosis, abdominal aortic aneurysm, varicose veins and hypertension. Genetic and pharmacological tools to decrease the production of cytokines or to diminish their effects using cytokine antagonists could provide new approaches in the management of inflammatory vascular disease.
Collapse
Affiliation(s)
- Alexander H Sprague
- Division of Vascular Surgery and Endovascular Therapy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
28
|
Parmar JH, Aslam M, Standfield NJ. Percutaneous transluminal angioplasty of lower limb arteries causes a systemic inflammatory response. Ann Vasc Surg 2009; 23:569-76. [PMID: 19467836 DOI: 10.1016/j.avsg.2009.02.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2008] [Revised: 01/28/2009] [Accepted: 02/17/2009] [Indexed: 10/20/2022]
Abstract
BACKGROUND Percutaneous transluminal angioplasty (PTA) of a lower limb arterial segment is a well-established treatment for suitable lesions for critical or noncritical lower limb ischemia. Our aim was to define the inflammatory response after PTA by measuring inflammatory markers. METHODS Twenty-five patients having PTA were compared with 20 patients having angiography alone. Interleukin-6 (IL-6), IL-8, IL-10, and tumor necrosis factor-alpha (TNF-alpha) were measured sequentially. The difference between postprocedure and preprocedure baseline levels were compared statistically between angiography alone and PTA. Patients were followed up to 1 year after the procedure, and the failure rate of PTA was noted. RESULTS IL-6 and TNF-alpha were significantly higher in PTA patients at 1 hr after PTA (p < 0.05), and the IL-6 level only was significantly higher at 24 hr post-PTA (p < 0.05) compared to angiography alone (Mann-Whitney test). IL-8 and IL-10 levels did not differ significantly in the PTA group. At 1 year after the procedure, 45% of PTAs had failed. There was no statistically significant correlation between failed PTA and inflammatory response. CONCLUSION PTA appears to cause a significant inflammatory response compared to angiography alone. This demonstrates a systemic manifestation of localized ischemia/reperfusion injury. Further investigation of the inflammatory response due to ischemia/reperfusion injury and its correlation with restenosis is recommended.
Collapse
Affiliation(s)
- Jitesh H Parmar
- Vascular Surgery Department, Hammersmith Hospital, London, UK.
| | | | | |
Collapse
|
29
|
Osipov RM, Bianchi C, Clements RT, Feng J, Liu Y, Xu SH, Robich MP, Wagstaff J, Sellke FW. Thrombin fragment (TP508) decreases myocardial infarction and apoptosis after ischemia reperfusion injury. Ann Thorac Surg 2009; 87:786-93. [PMID: 19231390 DOI: 10.1016/j.athoracsur.2008.12.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Revised: 12/02/2008] [Accepted: 12/05/2008] [Indexed: 12/16/2022]
Abstract
BACKGROUND Myocardial ischemia-reperfusion injury may lead to cardiac dysfunction or death. This study investigates the potential efficacy of a novel thrombin fragment (TP508) on myocardial ischemia-reperfusion injury. METHODS Fourteen male Yucatan pigs underwent 60 minutes of mid-left anterior descending coronary artery occlusion followed by 120 minutes of reperfusion. Pigs received either saline vehicle (control, n = 7) or thrombin fragment TP508 (n = 7) as a bolus (0.5 mg/kg) 50 minutes into the ischemic period, followed by continuous intravenous infusion (1.25 mg x kg(-1) x h(-1)) during reperfusion. Myocardial function was monitored throughout the experiments. Monastryl blue/triphenyl tetrazolium chloride staining was utilized to measure the area at risk and infarcted tissue. Apoptosis was assessed by Western blotting and dUTP nick-end labeling (TUNEL) staining. Coronary microvascular reactivity to endothelium-dependent factors (adenosine diphosphate, substance P, A23187) and endothelium-independent factor (sodium nitroprusside) was examined. RESULTS Global and regional left ventricular function was not significantly different between groups. Endothelium-dependent coronary microvascular relaxation was greater in the TP508 group and associated with higher endothelial nitric oxide synthase phosphorylation. Both infarct size and TUNEL staining was significantly decreased in the TP508 group compared with the control group (p < 0.05). Expression of the cell survival proteins B-cell lymphoma 2 (2.2-fold, p < 0.05) and heat shock protein-73 (1.6-fold, p < 0.05) was higher in the TP508 group. Expression of the cell-death-signaling proteins poly adenosine diphosphate-ribose polymerase (1.6-fold, p < 0.05), cleaved poly adenosine diphosphate-ribose polymerase (6.4-fold, p < 0.05), and B-cell lymphoma 2/adenovirus E1B 19 kDa-interacting protein 3 (3.8-fold, p < 0.05) was significantly higher in the TP508 group in the ischemic territory. CONCLUSIONS This study demonstrates that TP508 decreases infarct size, improves endothelial microvascular function, and induces cell-survival signaling in the setting of ischemia-reperfusion injury. Thus, TP508 may be a useful agent to attenuate myocardial reperfusion injury.
Collapse
Affiliation(s)
- Robert M Osipov
- Department of General Surgery, Cardiothoracic Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Borissoff JI, Spronk HMH, Heeneman S, ten Cate H. Is thrombin a key player in the 'coagulation-atherogenesis' maze? Cardiovasc Res 2009; 82:392-403. [PMID: 19228706 DOI: 10.1093/cvr/cvp066] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
In addition to its established roles in the haemostatic system, thrombin is an intriguing coagulation protease demonstrating an array of effects on endothelial cells, vascular smooth muscle cells (VSMC), monocytes, and platelets, all of which are involved in the pathophysiology of atherosclerosis. There is mounting evidence that thrombin acts as a powerful modulator of many processes like regulation of vascular tone, permeability, migration and proliferation of VSMC, recruitment of monocytes into the atherosclerotic lesions, induction of diverse pro-inflammatory markers, and all of these are related to the progression of cardiovascular disease. Recent studies in transgenic mice models indicate that the deletion of the natural thrombin inhibitor heparin cofactor II promotes an accelerated atherogenic state. Moreover, the reduction of thrombin activity levels in apolipoprotein E-deficient mice, because of the administration of the direct thrombin inhibitor melagatran, attenuates plaque progression and promotes stability in advanced atherosclerotic lesions. The combined evidence points to thrombin as a pivotal contributor to vascular pathophysiology. Considering the clinical development of selective anticoagulants including direct thrombin inhibitors, it is a relevant moment to review the different thrombin-induced mechanisms that contribute to the initiation, formation, progression, and destabilization of atherosclerotic plaques.
Collapse
Affiliation(s)
- Julian Ilcheff Borissoff
- Laboratory for Clinical Thrombosis and Hemostasis, Department of Internal Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center+ (MUMC+), Maastricht, The Netherlands
| | | | | | | |
Collapse
|
31
|
Wang Z, Kong L, Kang J, Morgan JH, Shillcutt SD, Robinson JS, Nakayama DK. Thrombin stimulates mitogenesis in pig cerebrovascular smooth muscle cells involving activation of pro-matrix metalloproteinase-2. Neurosci Lett 2009; 451:199-203. [DOI: 10.1016/j.neulet.2009.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Revised: 12/23/2008] [Accepted: 01/02/2009] [Indexed: 11/28/2022]
|
32
|
Loppnow H, Werdan K, Buerke M. Vascular cells contribute to atherosclerosis by cytokine- and innate-immunity-related inflammatory mechanisms. Innate Immun 2008; 14:63-87. [PMID: 18713724 DOI: 10.1177/1753425908091246] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases are the human diseases with the highest death rate and atherosclerosis is one of the major underlying causes of cardiovascular diseases. Inflammatory and innate immune mechanisms, employing monocytes, innate receptors, innate cytokines, or chemokines are suggested to be involved in atherogenesis. Among the inflammatory pathways the cytokines are central players. Plasma levels of cytokines and related proteins, such as CRP, have been investigated in cardiovascular patients, tissue mRNA expression was analyzed and correlations to vascular diseases established. Consistent with these findings the generation of cytokine-deficient animals has provided direct evidence for a role of cytokines in atherosclerosis. In vitro cell culture experiments further support the suggestion that cytokines and other innate mechanisms contribute to atherogenesis. Among the initiation pathways of atherogenesis are innate mechanisms, such as toll-like-receptors (TLRs), including the endotoxin receptor TLR4. On the other hand, innate cytokines, such as IL-1 or TNF, or even autoimmune triggers may activate the cells. Cytokines potently activate multiple functions relevant to maintain or spoil homeostasis within the vessel wall. Vascular cells, not least smooth muscle cells, can actively contribute to the inflammatory cytokine-dependent network in the blood vessel wall by: (i) production of cytokines; (ii) response to these potent cell activators; and (iii) cytokine-mediated interaction with invading cells, such as monocytes, T-cells, or mast cells. Activation of these pathways results in accumulation of cells and increased LDL- and ECM-deposition which may serve as an 'immunovascular memory' resulting in an ever-growing response to subsequent invasions. Thus, vascular cells may potently contribute to the inflammatory pathways involved in development and acceleration of atherosclerosis.
Collapse
Affiliation(s)
- Harald Loppnow
- Martin-Luther-Universität Halle-Wittenberg, Universitätsklinik und Poliklinik für Innere Medizin , Halle (Saale), Germany.
| | | | | |
Collapse
|
33
|
Procoagulant signalling mechanisms in lung inflammation and fibrosis: novel opportunities for pharmacological intervention? Br J Pharmacol 2008; 153 Suppl 1:S367-78. [PMID: 18223674 DOI: 10.1038/sj.bjp.0707603] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
There is compelling evidence that uncontrolled activation of the coagulation cascade following lung injury contributes to the development of lung inflammation and fibrosis in acute lung injury/acute respiratory distress syndrome (ALI/ARDS) and fibrotic lung disease. This article reviews our current understanding of the mechanisms leading to the activation of the coagulation cascade in response to lung injury and the evidence that excessive procoagulant activity is of pathophysiological significance in these disease settings. Current evidence suggests that the tissue factor-dependent extrinsic pathway is the predominant mechanism by which the coagulation cascade is locally activated in the lungs of patients with ALI/ARDS and pulmonary fibrosis. Whilst, fibrin deposition might contribute to the pathophysiology of ALI/ARDS following systemic insult; current evidence suggests that the cellular effects mediated via activation of proteinase-activated receptors (PARs) may be of particular importance in influencing inflammatory and fibroproliferative responses in experimental models involving direct injury to the lung. In this regard, studies in PAR(1) knockout mice have shown that this receptor plays a major role in orchestrating the interplay between coagulation, inflammation and lung fibrosis. This review will focus on our current understanding of excessive procoagulant signalling in acute and chronic lung injury and will highlight the novel opportunities that this may present for therapeutic intervention.
Collapse
|
34
|
Hayakawa Y, Kurimoto M, Nagai S, Kurosaki K, Tsuboi Y, Hamada H, Hayashi N, Endo S. Thrombin-induced cell proliferation and platelet-derived growth factor-AB release from A172 human glioblastoma cells. J Thromb Haemost 2007; 5:2219-26. [PMID: 17958740 DOI: 10.1111/j.1538-7836.2007.02739.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND In a previous study, we found that thrombin induced proliferation of TM-1 and T98G human glioma cells and that the mitogenic effect was abolished by hirudin. OBJECTIVES We investigated thrombin's effects on the proliferation of A172 human glioblastoma cells and the induction of growth factors. Furthermore, we examined whether or not the expression of heparin cofactor II (HCII) in A172 cells using adenovirus vector could suppress thrombin's effects. METHODS The effect of thrombin on cell proliferation was assessed using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide assay. The amount of growth factors in the conditioned medium was measured by enzyme-linked immunosorbent assay. The level of platelet-derived growth factor (PDGF)-B mRNA was assessed by reverse transcriptase-polymerase chain reaction analysis. RESULTS Thrombin-induced proliferation of A172 cells primarily depended on the enhanced secretion of PDGF-AB by thrombin. The action of thrombin depended on its proteolytic activity. However, thrombin-induced PDGF-AB secretion was not abolished by anti-protease-activated receptor (PAR) antibody. The PAR-1 agonist peptide had no effect on cell growth and PDGF-AB levels. Thrombin did not increase PDGF-B gene expression. Expression of HCII effectively suppressed thrombin-induced PDGF-AB release. CONCLUSIONS These results indicate that thrombin may play an important role in the proliferation of A172 cells by inducing PDGF-AB secretion and that thrombin's action is mediated by its proteolytic activity. Inhibition of thrombin's proteolytic activity may be a new therapeutic method for gliomas.
Collapse
Affiliation(s)
- Y Hayakawa
- Department of Neurosurgery, Faculty of Medicine, University of Toyama, Toyama, Japan
| | | | | | | | | | | | | | | |
Collapse
|
35
|
O'Brien M, Morrison JJ, Smith TJ. Expression of prothrombin and protease activated receptors in human myometrium during pregnancy and labor. Biol Reprod 2007; 78:20-6. [PMID: 17901076 DOI: 10.1095/biolreprod.107.062182] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
As thrombin is proposed to be involved in stimulating myometrial contractility during labor and preterm labor, we aimed to investigate the expression of prothrombin (F7), the precursor of thrombin, its receptors, the protease-activated receptor (PAR) family (F2R, F2RL1, F2RL2, and F2RL3), and prothrombinase FGL2 in human myometrium during pregnancy and labor. Messenger RNA and protein were isolated from human pregnant laboring and nonlaboring myometrial tissue and from human primary myometrial smooth muscle cells. Semiquantitative RT-PCR, real-time fluorescence RT-PCR, Western blotting, and fluorescence microscopy were performed to determine the expression levels of F7, FGL2, F2R, F2RL1, F2RL2, and F2RL3 in the myometrial tissues and cells. The expression of mRNA and protein for these molecules is reported for the first time in human myometrium at term pregnancy, at labor, and in the nonpregnant state. Importantly, an increase in F2R and a significant increase in F2RL3 mRNA expression at labor were demonstrated. Statistically significant increases in F2R and F2RL3 protein expression was also detected in human myometrium at labor. Furthermore, FGL2 mRNA expression at labor, and FGL2 protein expression at term pregnancy and at labor was observed in this tissue for the first time. The expression of F7, FGL2, F2R, F2RL1, F2RL2, and F2RL3 in human myometrium reveals that all the machinery necessary for thrombin activation and cellular activity is present in the myometrium during pregnancy and labor. These data, in conjunction with the demonstrated increase in F2R and F2RL3 expression at labor, suggest a principal role for these molecules in the regulation of myometrial function at labor, including preterm labor.
Collapse
Affiliation(s)
- Margaret O'Brien
- National Center for Biomedical and Engineering Science, National University of Ireland Galway, Galway, Ireland.
| | | | | |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW The aim of this article is to highlight the importance of thrombotic processes in the development and complications of atherosclerotic vascular disease. RECENT FINDINGS Thrombin generated at sites of vascular inflammation activates major atheroma-associated cells including endothelial cells, platelets, smooth muscle cells, monocytes, and macrophages. Thrombin-activated cells produce a plethora of inflammatory mediators, such as regulated upon activation normal T cell expressed presumed secreted, macrophage migration inhibitory factor, and CD40 ligand, that promote atherosclerotic lesion formation and atherothrombotic complications of vascular disease. Additionally, thrombin-induced inflammatory mediators stimulate tissue factor procoagulant activity within atheroma to initiate a positive feedback loop where thrombin activation launches inflammatory signals that lead to further thrombin activation. Platelets, the main cellular effectors of the thrombotic system, also play a central role in the biology of atherosclerosis by producing inflammatory mediators and directing leukocyte incorporation into plaques through platelet-mediated leukocyte adhesion. SUMMARY New research has identified signaling pathways that intertwine thrombotic and inflammatory pathways with the development and progression of atherosclerosis. These signaling pathways contain positive feedback loops that propagate atherogenesis. Targeting molecular regulators at the interface of thrombosis and inflammation simultaneously may reduce thrombosis and inflammation, thus breaking pathological cycles that promote atherosclerosis and associated thrombotic complications.
Collapse
Affiliation(s)
- Kevin Croce
- Donald W. Reynolds Cardiovascular Clinical Research Center, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
37
|
Artuc M, Hermes B, Algermissen B, Henz BM. Expression of prothrombin, thrombin and its receptors in human scars. Exp Dermatol 2006; 15:523-9. [PMID: 16761961 DOI: 10.1111/j.1600-0625.2006.00444.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The coagulation system is thought to play a pivotal role during the initial phase of wound healing, but mechanisms and cells involved are only partly understood. We have therefore examined human scars for the expression of thrombin, its precursor prothrombin and the thrombin receptors, thrombomodulin (TM) and protease-activated receptor-1 (PAR-1), compared with normal skin. Biopsies of scars were obtained from primary excision sites of melanoma patients (n = 20) and were compared with normal skin distant from the scar (n = 10), using immunohistochemistry. In addition, polymerase chain reaction analyses were performed on scar versus normal tissue and on cultured keratinocytes, fibroblasts and endothelial cells before and after stimulation with selected cytokines known to be active in wound healing. Normal epidermis was stained for prothrombin, thrombin, TM and PAR-1, and dermal tissue was stained only for TM and PAR-1. In scar tissue, thrombin and TM were upregulated in the epidermis and all four molecules in the dermis, independent of the age of the scars. In tissue extracts, mRNA expression of PAR-1 and prothrombin expression were, however, unchanged and TM even slightly decreased in scars, compared with normal skin. On analysis of cultured cells, keratinocytes expressed mRNA for PAR-1, TM and prothrombin, endothelial cells for PAR-1 and TM, and fibroblasts for PAR-1. An upregulation of PAR-1 mRNA was induced in fibroblasts on exposure to tumor necrosis factor-alpha (TNF-alpha), while it remained unchanged in endothelial cells in response to TNF-alpha. A downregulation of TM was induced in endothelial cells on exposure to TNF-alpha. These findings, showing a marked modulation of thrombin, PAR-1 and TM even in older human scar tissue, suggest that the coagulation system is not only involved during clotting, but also during the inflammatory and tissue remodelling phases of wound healing.
Collapse
MESH Headings
- Cells, Cultured
- Child
- Cicatrix/metabolism
- Cicatrix/pathology
- Dermis/cytology
- Dermis/drug effects
- Dermis/metabolism
- Endothelial Cells/cytology
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Epidermal Cells
- Epidermis/drug effects
- Epidermis/metabolism
- Fibroblasts/cytology
- Fibroblasts/drug effects
- Fibroblasts/metabolism
- Gene Expression/drug effects
- Gene Expression/genetics
- Humans
- Immunohistochemistry
- Keratinocytes/cytology
- Keratinocytes/drug effects
- Keratinocytes/metabolism
- Male
- Prothrombin/genetics
- Prothrombin/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, PAR-1/genetics
- Receptor, PAR-1/metabolism
- Receptors, Thrombin/genetics
- Receptors, Thrombin/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Skin/drug effects
- Skin/metabolism
- Skin/pathology
- Thrombin/genetics
- Thrombin/metabolism
- Thrombomodulin/genetics
- Thrombomodulin/metabolism
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- Metin Artuc
- Department of Dermatology and Allergy, Universitätsmedizin Berlin Charité, Berlin, Germany.
| | | | | | | |
Collapse
|
38
|
Li T, Wang H, He S. Induction of interleukin-6 release from monocytes by serine proteinases and its potential mechanisms. Scand J Immunol 2006; 64:10-6. [PMID: 16784486 DOI: 10.1111/j.1365-3083.2006.01772.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Serine proteinases have been recognized playing an important role in inflammation via proteinase-activated receptors (PAR). However, little is known of the influence of serine proteinases and PAR on interleukin-6 (IL-6) secretion from highly purified monocytes. We challenged monocytes from human peripheral blood with serine proteinases and agonist peptides of PAR and measured the levels of IL-6, IL-1beta and IL-12 in culture supernatants by enzyme-linked immunosorbent assay. The results showed that thrombin, trypsin, tryptase and elastase stimulated approximately up to 2.9-, 2.0-, 1.8- and 2.1-fold increase in IL-6 release from monocytes following 16 h of incubation, respectively. Proteinase inhibitors inhibited the actions of proteinases on monocytes. Agonist peptides of PAR-1 (SFLLR-NH(3)) and PAR-4 (GYPGQV-NH(2)), but not PAR-3 (TFRGAP-NH(2)), also induced IL-6 release from monocytes. The proteinases and agonists of PAR failed to stimulate IL-1beta and IL-12 secretion. In conclusion, the induction of IL-6 secretion by serine proteinases may be through the activation of PAR.
Collapse
Affiliation(s)
- T Li
- Allergy and Inflammation Research Institute, The Key Immunopharmacology Laboratory of Guangdong Province, Shantou University Medical College, Shantou 515031, Guangdong Province, China
| | | | | |
Collapse
|
39
|
Wahlgren CM, Sten-Linder M, Egberg N, Kalin B, Blohmé L, Swedenborg J. The Role of Coagulation and Inflammation After Angioplasty in Patients with Peripheral Arterial Disease. Cardiovasc Intervent Radiol 2006; 29:530-5. [PMID: 16729229 DOI: 10.1007/s00270-005-0159-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
PURPOSE Restenosis remains a frequent complication after angioplasty in peripheral arterial disease. Inflammation plays a critical role in the vascular response to injury. Effective medical treatment to improve patency after angioplasty is still elusive. The aims of this prospective clinical study were to investigate changes in blood coagulation and inflammatory markers after angioplasty and their significance for restenosis. METHODS Thirty-four patients with peripheral arterial disease underwent angioplasty of the iliac and superficial femoral arteries. Ten patients undergoing diagnostic angiography were included in the study as controls. Plasma levels of tissue factor, prothrombin fragment 1 + 2, D-dimer, P-selectin, C-reactive protein (CRP), and fibrinogen were analyzed before and after angioplasty. Patients were followed up with angiography after 6 months to assess restenosis. RESULTS CRP was elevated the day after angioplasty (6.6 mg/l, p = 0.0001) and tended to peak after 1 week (11 mg/l, p = 0.09). There was a significant increase of D-dimer and P-selectin 1-4 hr after angioplasty (0.4 mg/l, p = 0.001 and 68 ng/ml, p = 0.05, respectively). None of the biochemical markers was a statistically significant predictor of restenosis. CONCLUSION We have observed a much more prolonged inflammatory response than previously noted, but only minor changes in coagulation activity after angioplasty. The biochemical markers, before and after angioplasty, were not related to restenosis. Further studies are needed to delineate the molecular mechanisms behind these observations and their involvement in thrombosis and restenosis. If these pathways are further defined, improved treatment strategies, including antithrombotic treatments and statins, could be tailored to modulate postprocedural inflammation.
Collapse
Affiliation(s)
- C M Wahlgren
- Department of Vascular Surgery, Karolinska University Hospital, Stockholm, 171 76, Sweden.
| | | | | | | | | | | |
Collapse
|
40
|
Day JRS, Taylor KM, Lidington EA, Mason JC, Haskard DO, Randi AM, Landis RC. Aprotinin inhibits proinflammatory activation of endothelial cells by thrombin through the protease-activated receptor 1. J Thorac Cardiovasc Surg 2006; 131:21-7. [PMID: 16399290 DOI: 10.1016/j.jtcvs.2005.08.050] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2005] [Revised: 07/26/2005] [Accepted: 08/31/2005] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Thrombin is generated in significant quantities during cardiopulmonary bypass and mediates adverse events, such as platelet aggregation and proinflammatory responses, through activation of the high-affinity thrombin receptor protease-activated receptor 1, which is expressed on platelets and endothelium. Thus antagonism of protease-activated receptor 1 might have broad therapeutic significance. Aprotinin, used clinically to reduce transfusion requirements and the inflammatory response to bypass, has been shown to inhibit protease-activated receptor 1 on platelets in vitro and in vivo. Here we have examined whether aprotinin inhibits endothelial protease-activated receptor 1 activation and resulting proinflammatory responses induced by thrombin. METHODS Protease-activated receptor 1 expression and function were examined in cultured human umbilical vein endothelial cells after treatment with alpha-thrombin at 0.02 to 0.15 U/mL in the presence or absence of aprotinin (200-1600 kallikrein inhibitory units/mL). Protease-activated receptor 1 activation was assessed by using an antibody, SPAN-12, which detects only the unactivated receptor, and thrombin-mediated calcium fluxes. Other thrombin-dependent inflammatory pathways investigated were phosphorylation of the p42/44 mitogen-activated protein kinase, upregulation of the early growth response 1 transcription factor, and production of the proinflammatory cytokine interleukin 6. RESULTS Pretreatment of cultured endothelial cells with aprotinin significantly spared protease-activated receptor 1 receptor cleavage (P < .0001) and abrogated calcium fluxes caused by thrombin. Aprotinin inhibited intracellular signaling through p42/44 mitogen-activated protein kinase (P < .05) and early growth response 1 transcription factor (P < .05), as well as interleukin 6 secretion caused by thrombin (P < .005). CONCLUSIONS This study demonstrates that endothelial cell activation by thrombin and downstream inflammatory responses can be inhibited by aprotinin in vitro through blockade of protease-activated receptor 1. Our results provide a new molecular basis to help explain the anti-inflammatory properties of aprotinin reported clinically.
Collapse
Affiliation(s)
- Jonathan R S Day
- Eric Bywaters Centre, Imperial College London, Faculty of Medicine, Hammersmith Hospital, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
41
|
Ye HJ, Zhao SP. Anti-atherogenic properties of fibrates may be largely due to their anti-inflammatory effects. Med Hypotheses 2006; 66:495-500. [PMID: 16289368 DOI: 10.1016/j.mehy.2005.09.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2005] [Accepted: 09/27/2005] [Indexed: 11/18/2022]
Abstract
Current understanding of the pathophysiology of atherosclerosis has undergone a remarkable evolution. Compelling evidence has evolved at both the basic science and clinical level for the importance of inflammation in the pathogenesis of atherosclerosis and its complications. Recent research has shown that both systemic and local inflammation plays a central role in all phases of the atherosclerotic process. Inflammatory cells dominate early atherosclerotic lesions, inflammatory cytokines accelerate progression of the lesions, and activation of inflammation can elicit acute coronary syndromes. Robust clinical studies have affirmed that fibrates are anti-atherogenic and can improve the cardiovascular risk profile. Fibrates not only modulate the serum concentrations of triglyceride and cholesterol, but also inhibit systemic inflammatory statue and inflammatory response in vascular cells. Fibrates act anti-inflammatory effects in monocyte/macrophage, T lymphocyte, endothelial cells, vascular smooth muscle cells and adipocytes. Since atherosclerosis is now regarded as an inflammatory disease and those inflammatory cells play critical important roles in the initiation and development of atherosclerosis, we hypothesize that anti-atherogenic properties of fibrates may be largely due to their anti-inflammatory effects.
Collapse
Affiliation(s)
- Hui-Jun Ye
- Department of Cardiology, The Second XiangYa Hospital, Central South University, Middle Renmin Road No. 139, ChangSha, HuNan 410011, People's Republic of China.
| | | |
Collapse
|
42
|
Abstract
The coagulation cascade and protease-activated receptors (PARs) together provide an elegant mechanism that links mechanical information in the form of tissue injury to cellular responses. These receptors appear to largely account for the cellular effects of thrombin and can mediate signaling to other trypsin-like proteases. An important role for PARs in hemostasis and thrombosis is established in animal models, and studies in knockout mice and nonhuman primates raise the question of whether PAR inhibition might offer an appealing new approach to the prevention and treatment of thrombosis. PARs may also trigger inflammatory responses to tissue injury. For example, PAR activation on endothelial cells and perhaps sensory afferents can trigger local accumulation of leukocytes and platelets and transudation of plasma. However, panoply of signaling systems and cell types orchestrates inflammatory responses, and efforts to define the relative importance and roles of PARs in various inflammatory processes are just beginning. Lastly, roles for PARs in blood vessel formation and other processes during embryonic development are emerging, and whether these reflect new roles for the coagulation cascade and/or PAR signaling to other proteases remains to be explored.
Collapse
Affiliation(s)
- S R Coughlin
- Cardiovascular Research Institute, Department of Medicine, University of California, San Francisco, CA 94143-0130, USA.
| |
Collapse
|
43
|
Abstract
Inflammation is now recognized as being pivotal in the pathogenesis of atherosclerosis. This review highlights key concepts in our current understanding of the role of inflammation in the initiation, progression, and complication of atherosclerosis. The role of various triggers and amplifiers and the innate and adaptive immunity in the cascade of inflammatory events are also presented. Progress in our understanding of the inflammatory pathways in atherosclerosis has provided further mechanistic insight into the clinical benefits of current medical therapy and may alter our future treatment and preventive strategies.
Collapse
Affiliation(s)
- Alice Y Tiong
- Department of Cardiology, Concord Repatriation General Hospital, Sydney, Australia.
| | | |
Collapse
|
44
|
Steinhoff M, Buddenkotte J, Shpacovitch V, Rattenholl A, Moormann C, Vergnolle N, Luger TA, Hollenberg MD. Proteinase-activated receptors: transducers of proteinase-mediated signaling in inflammation and immune response. Endocr Rev 2005; 26:1-43. [PMID: 15689571 DOI: 10.1210/er.2003-0025] [Citation(s) in RCA: 364] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Serine proteinases such as thrombin, mast cell tryptase, trypsin, or cathepsin G, for example, are highly active mediators with diverse biological activities. So far, proteinases have been considered to act primarily as degradative enzymes in the extracellular space. However, their biological actions in tissues and cells suggest important roles as a part of the body's hormonal communication system during inflammation and immune response. These effects can be attributed to the activation of a new subfamily of G protein-coupled receptors, termed proteinase-activated receptors (PARs). Four members of the PAR family have been cloned so far. Thus, certain proteinases act as signaling molecules that specifically regulate cells by activating PARs. After stimulation, PARs couple to various G proteins and activate signal transduction pathways resulting in the rapid transcription of genes that are involved in inflammation. For example, PARs are widely expressed by cells involved in immune responses and inflammation, regulate endothelial-leukocyte interactions, and modulate the secretion of inflammatory mediators or neuropeptides. Together, the PAR family necessitates a paradigm shift in thinking about hormone action, to include proteinases as key modulators of biological function. Novel compounds that can modulate PAR function may be potent candidates for the treatment of inflammatory or immune diseases.
Collapse
Affiliation(s)
- Martin Steinhoff
- Department of Dermatology and Boltzmann Institute for Immunobiology of the Skin, University of Münster, von-Esmarch-Strasse 58, 48149 Münster, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Hayakawa Y, Hirashima Y, Yamamoto H, Hayashi N, Kurimoto M, Kuwayama N, Endo S. Adenovirus-mediated expression of heparin cofactor II inhibits thrombin-induced cellular responses in fibroblasts and vascular smooth muscle cells. Thromb Res 2005; 116:357-63. [PMID: 16038721 DOI: 10.1016/j.thromres.2005.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2004] [Revised: 01/03/2005] [Accepted: 01/04/2005] [Indexed: 11/21/2022]
Abstract
Heparin cofactor II functions as a physiological inhibitor of thrombin activity. The rate of inactivation of thrombin by heparin cofactor II is increased in the presence of dermatan sulfate, which is produced by fibroblasts or smooth muscle cells. To elucidate the role of heparin cofactor II in the extravascular cells, we induced expression of heparin cofactor II in cultured human fibroblasts or vascular smooth muscle cells using adenovirus-mediated gene transfer. After infection of adenovirus vector, these cells secreted heparin cofactor II protein into culture medium. The expressed heparin cofactor II formed the complex with exogenous thrombin and inhibited the proteolytic activity of thrombin. Expression of heparin cofactor II by infection of adenovirus vector inhibited thrombin-induced tissue-type plasminogen activator and interleukin-6 releases from fibroblasts and thrombin-induced interleukin-6 release from vascular smooth muscle cells. These findings show that fibroblasts and vascular smooth muscle cells expressing heparin cofactor II are resistant to thrombin-induced cellular responses.
Collapse
Affiliation(s)
- Yumiko Hayakawa
- Department of Neurosurgery, Faculty of Medicine, Toyama Medical and Pharmaceutical University, 2630 Sugitani, Toyama 930-0194, Japan.
| | | | | | | | | | | | | |
Collapse
|
46
|
Naldini A, Carraro F, Baldari CT, Paccani SR, Bernini C, Keherly MJ, Carney DH. The thrombin peptide, TP508, enhances cytokine release and activates signaling events. Peptides 2004; 25:1917-26. [PMID: 15501523 DOI: 10.1016/j.peptides.2004.05.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2004] [Revised: 05/26/2004] [Accepted: 05/27/2004] [Indexed: 10/26/2022]
Abstract
The thrombin peptide, TP508, accelerates tissue repair and initiates a cascade of cellular events. We have previously shown that alpha-thrombin induces cytokine expression in human mononuclear cells. We, therefore, investigated the possibility that TP508 might activate cytokine production and intracellular signaling pathways associated with cytokine activation. Our results show that TP508 induces cytokine expression in human mononuclear cells. TP508 treatment enhances extracellular signal-regulated kinase (Erk1/2) activities in U937 cells, as well as Erk1/2 and p38 activation in Jurkat T cells. These data support the hypothesis that TP508 may accelerate tissue repair through the activation of the inflammatory response.
Collapse
Affiliation(s)
- Antonella Naldini
- Department of Physiology, University of Siena, Via Aldo Moro, 53100 Siena, Italy.
| | | | | | | | | | | | | |
Collapse
|
47
|
Légaré JF, Ross DB. Suggestion for functional model to test effects of decellularization of rat aortic valve allografts on leaflet destruction and extracellular matrix remodeling. J Thorac Cardiovasc Surg 2004; 128:155-6; author reply 156. [PMID: 15224037 DOI: 10.1016/j.jtcvs.2004.03.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
48
|
Yang MS, Lee J, Ji KA, Min KJ, Lee MA, Jou I, Joe E. Thrombin induces suppressor of cytokine signaling 3 expression in brain microglia via protein kinase Cdelta activation. Biochem Biophys Res Commun 2004; 317:811-6. [PMID: 15081412 DOI: 10.1016/j.bbrc.2004.03.118] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2004] [Indexed: 11/30/2022]
Abstract
Microglia (brain macrophages) are activated upon brain damage. In this study, we demonstrated that thrombin, a pro-inflammatory stimulator of microglia, induced expression of suppressors of cytokine signaling (SOCS) in microglia. RT-PCR analysis and Northern blot analysis showed that thrombin induced SOCS3 mRNA expression. Further experiments indicated SOCS3 expression was not affected by cycloheximide, indicating thrombin directly stimulated SOCS3 transcript expression without de novo protein synthesis. We investigated whether PKCdelta played a role in thrombin-stimulated SOCS3 expression. We found that thrombin activated PKCdelta, and the specific inhibitor of PKCdelta, rottlerin, significantly suppressed thrombin-stimulated SOCS3 expression. In thrombin-pretreated cells, microglial activation-induced by another inflammatory stimulator, lipopolysaccharide, was attenuated compared to that in non-pretreated cells. These results suggest thrombin induce not only proinflammatory mediators but also negative feedback regulators of inflammation, SOCS, which prevent prolonged inflammatory reactions in microglia.
Collapse
Affiliation(s)
- Myung-Soon Yang
- Neuroscience graduate program, Ajou University School of Medicine, Suwon 442-721, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
49
|
Damiano BP, Derian CK, Maryanoff BE, Zhang HC, Gordon PA. RWJ-58259: a selective antagonist of protease activated receptor-1. ACTA ACUST UNITED AC 2004; 21:313-26. [PMID: 14647534 DOI: 10.1111/j.1527-3466.2003.tb00124.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Protease activated receptor-1 (PAR-1) is a key mediator of the cellular actions of alpha-thrombin. Thus, antagonism of this unique G-protein coupled receptor with a small molecule represents a means of selectively inhibiting thrombin's cellular actions without inhibiting its proteolytic activity. RWJ-58259 (alphaS)-N-[(1S)-3-amino-1-[[(phenylmethyl)- amino]carbonyl]propyl]-alpha-[[[[[1-(2,6-dichlorophenyl)methyl]-3-(1-pyrrolidinylmethyl)-1H-indazol-6-yl]amino]carbonyl]amino]-3,4-difluorobenzenepropanamide) is a potent and selective inhibitor of PAR-1 identified as part of a synthetic chemistry program based upon a de novo design approach. RWJ-58259 inhibited thrombin-induced platelet aggregation in human platelets with an IC50 of 0.37 microM without inhibiting thrombin's proteolytic activity or aggregation induced by other agonists. RWJ-58259 was not effective in guinea pig models of thrombosis. This reflected the presence of a second thrombin-sensitive receptor system in guinea pigs (PAR-3/4) and the selectivity of RWJ-58259 for PAR-1. However, RWJ-58259 was effective in a non-human primate model of thrombosis. Because human platelets have a PAR expression profile similar to the non-human primate, PAR-1 antagonism has the potential to be antithrombotic in humans. RWJ-58259 also inhibited thrombin-induced intracellular calcium signaling and proliferation in rat vascular smooth muscle cells. Perivascular application of RWJ-58259 in vivo significantly inhibited arterial injury-induced stenosis in a rat model of balloon angioplasty. These preclinical results suggest a potential clinical utility of RWJ-58259 for treatment of thrombotic disorders and vascular injury associated with acute coronary interventions and atherosclerosis. Given the potential role of PAR-1 in thrombin's actions in other cell types and disease states, RWJ-58259 provides a means for assessing additional clinical utilities of PAR-1 antagonism in disease conditions such as inflammation, cancer and neurodegeneration.
Collapse
Affiliation(s)
- Bruce P Damiano
- Johnson and Johnson Pharmaceutical Research and Development, Spring House, PA 19477-0776, USA.
| | | | | | | | | |
Collapse
|
50
|
Colognato R, Slupsky JR, Jendrach M, Burysek L, Syrovets T, Simmet T. Differential expression and regulation of protease-activated receptors in human peripheral monocytes and monocyte-derived antigen-presenting cells. Blood 2003; 102:2645-52. [PMID: 12805069 DOI: 10.1182/blood-2002-08-2497] [Citation(s) in RCA: 168] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Protease-activated receptors (PARs) are stimulated by proteolytic cleavage of their extracellular domain, unmasking a new N-terminus acting as tethered ligand. Whereas the role of PARs in platelets is well known, their presence and function in human monocytes and other antigen-presenting cells has not been characterized. Here it is demonstrated that human peripheral monocytes and monocyte-derived macrophages and dendritic cells differentially express PARs. Human monocytes express mainly PAR1 and less PAR3. Differentiation of monocytes into macrophages by either macrophage colony-stimulating factor (M-CSF) or granulocyte-macrophage colony-stimulating factor (GM-CSF) elicits enhanced expression of PAR1, PAR2, and PAR3. In contrast, dendritic cells differentiated from monocytes by GM-CSF and interleukin-4 (IL-4) strongly down-regulated PAR1, PAR2, and PAR3, both at the mRNA and the protein level. Down-regulation of the PAR expression was apparently due to IL-4, because treatment of macrophages with IL-4 caused down-regulation of PAR1, PAR2, and PAR3. PAR4 mRNA expression remained undetectable in any of the cell types investigated. Stimulation of PAR1, PAR2, and PAR3 with thrombin, trypsin, or established receptor-activating peptides (PAR-APs) triggered cytosolic Ca2+ responses, indicating functionally active PARs. Further, stimulation of monocytes or macrophages with thrombin or PAR1-AP, but not with PAR2-or PAR4-AP, triggers expression of monocyte chemoattractant protein-1 (MCP-1) both at the mRNA and the protein level. These data demonstrate that differentiation of human monocytes is associated with differential expression of functionally active PARs that mediate distinct regulatory functions in inflammation and atherogenesis.
Collapse
Affiliation(s)
- Renato Colognato
- Department of Pharmacology of Natural Products and Clinical Pharmacology, University of Ulm, Germany
| | | | | | | | | | | |
Collapse
|