1
|
Aleksandrowicz K, Hempel D, Polityńska B, Wojtukiewicz AM, Honn KV, Tang DG, Wojtukiewicz MZ. The Complex Role of Thrombin in Cancer and Metastasis: Focus on Interactions with the Immune System. Semin Thromb Hemost 2024; 50:462-473. [PMID: 37984359 DOI: 10.1055/s-0043-1776875] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Thrombin, a pleiotropic enzyme involved in coagulation, plays a crucial role in both procoagulant and anticoagulant pathways. Thrombin converts fibrinogen into fibrin, initiates platelet activation, and promotes clot formation. Thrombin also activates anticoagulant pathways, indirectly inhibiting factors involved in coagulation. Tissue factor triggers thrombin generation, and the overexpression of thrombin in various cancers suggests that it is involved in tumor growth, angiogenesis, and metastasis. Increased thrombin generation has been observed in cancer patients, especially those with metastases. Thrombin exerts its effects through protease-activated receptors (PARs), particularly PAR-1 and PAR-2, which are involved in cancer progression, angiogenesis, and immunological responses. Thrombin-mediated signaling promotes angiogenesis by activating endothelial cells and platelets, thereby releasing proangiogenic factors. These functions of thrombin are well recognized and have been widely described. However, in recent years, intriguing new findings concerning the association between thrombin activity and cancer development have come to light, which justifies a review of this research. In particular, there is evidence that thrombin-mediated events interact with the immune system, and may regulate its response to tumor growth. It is also worth reevaluating the impact of thrombin on thrombocytes in conjunction with its multifaceted influence on tumor progression. Understanding the role of thrombin/PAR-mediated signaling in cancer and immunological responses is crucial, particularly in the context of developing immunotherapies. In this systematic review, we focus on the impact of the thrombin-related immune system response on cancer progression.
Collapse
Affiliation(s)
- Karolina Aleksandrowicz
- Department of Clinical Oncology, Medical University, Białystok, Poland
- Comprehensive Cancer Center, Bialystok, Poland
| | - Dominika Hempel
- Department of Clinical Oncology, Medical University, Białystok, Poland
- Comprehensive Cancer Center, Bialystok, Poland
| | - Barbara Polityńska
- Department of Psychology and Philosophy, Medical University of Białystok, Białystok, Poland
| | - Anna M Wojtukiewicz
- Department of Psychology and Philosophy, Medical University of Białystok, Białystok, Poland
| | - Kenneth V Honn
- Department of Pathology-School of Medicine, Bioactive Lipids Research Program, Detroit, Michigan
- Department of Chemistry, Wayne State University, Detroit, Michigan
- Department of Oncology, Wayne State University, Detroit, Michigan
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Marek Z Wojtukiewicz
- Department of Clinical Oncology, Medical University, Białystok, Poland
- Comprehensive Cancer Center, Bialystok, Poland
| |
Collapse
|
2
|
Russo V, Falco L, Tessitore V, Mauriello A, Catapano D, Napolitano N, Tariq M, Caturano A, Ciccarelli G, D’Andrea A, Giordano A. Anti-Inflammatory and Anticancer Effects of Anticoagulant Therapy in Patients with Malignancy. Life (Basel) 2023; 13:1888. [PMID: 37763292 PMCID: PMC10532829 DOI: 10.3390/life13091888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Optimizing the anticoagulation therapy is of pivotal importance in patients with a malignant tumor, as venous thromboembolism (VTE) has become the second-leading cause of death in this population. Cancer can highly increase the risk of thrombosis and bleeding. Consequently, the management of cancer-associated VTE is complex. In recent years, translational research has intensified, and several studies have highlighted the role of inflammatory cytokines in cancer growth and progression. Simultaneously, the pleiotropic effects of anticoagulants currently recommended for VTE have emerged. In this review, we describe the anti-inflammatory and anticancer effects of both direct oral anticoagulants (DOACs) and low-molecular-weight heparins (LWMHs).
Collapse
Affiliation(s)
- Vincenzo Russo
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, NA, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Luigi Falco
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, NA, Italy
| | - Viviana Tessitore
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, NA, Italy
| | - Alfredo Mauriello
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, NA, Italy
| | - Dario Catapano
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, NA, Italy
| | - Nicola Napolitano
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, NA, Italy
| | - Moiz Tariq
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, NA, Italy
| | - Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, NA, Italy (A.D.)
| | - Giovanni Ciccarelli
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, NA, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Antonello D’Andrea
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, NA, Italy (A.D.)
- Cardiology Unit, Umberto I Hospital, 84014 Nocera Inferiore, SA, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
3
|
Schiffer S, Schwers S, Heitmeier S. The effect of rivaroxaban on biomarkers in blood and plasma: a review of preclinical and clinical evidence. J Thromb Thrombolysis 2023; 55:449-463. [PMID: 36746885 PMCID: PMC10110699 DOI: 10.1007/s11239-023-02776-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/15/2023] [Indexed: 02/08/2023]
Abstract
Rivaroxaban is a direct, oral factor Xa inhibitor that is used for the prevention and treatment of various thromboembolic disorders. Several preclinical and clinical studies have utilized specific molecules as biomarkers to investigate the potential role of rivaroxaban beyond its anticoagulant activity and across a range of biological processes. The aim of this review is to summarize the existing evidence regarding the use of blood-based biomarkers to characterize the effects of rivaroxaban on coagulation and other pathways, including platelet activation, inflammation and endothelial effects. After a literature search using PubMed, almost 100 preclinical and clinical studies were identified that investigated the effects of rivaroxaban using molecular biomarkers. In agreement with the preclinical data, clinical studies reported a trend for reduction in the blood concentrations of D-dimers, thrombin-antithrombin complex and prothrombin fragment 1 + 2 following treatment with rivaroxaban in both healthy individuals and those with various chronic conditions. Preclinical and also some clinical studies have also reported a potential impact of rivaroxaban on the concentrations of platelet activation biomarkers (von Willebrand factor, P-selectin and thrombomodulin), endothelial activation biomarkers (matrix metalloproteinase-9, intercellular adhesion molecule-1 and vascular cell adhesion molecule-1) and inflammation biomarkers (interleukin-6, tumor necrosis factor-α and monocyte chemoattractant protein-1). Based on the results of biomarker studies, molecular biomarkers can be used in addition to traditional coagulation assays to increase the understanding of the anticoagulation effects of rivaroxaban. Moreover, there is preliminary evidence to suggest that rivaroxaban may have an impact on the biological pathways of platelet activation, endothelial activation and inflammation; however, owing to paucity of clinical data to investigate the trends reported in preclinical studies, further investigation is required to clarify these observations.
Collapse
Affiliation(s)
- Sonja Schiffer
- Bayer AG, Pharmaceuticals, R&D, 42113 Wuppertal, Germany
| | | | | |
Collapse
|
4
|
Peng Q, Nowocin A, Ratnasothy K, Smith RA, Smyth LA, Lechler RI, Dorling A, Lombardi G. Inhibition of thrombin on endothelium enhances recruitment of regulatory T cells during IRI and when combined with adoptive Treg transfer, significantly protects against acute tissue injury and prolongs allograft survival. Front Immunol 2023; 13:980462. [PMID: 36793549 PMCID: PMC9924086 DOI: 10.3389/fimmu.2022.980462] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 12/28/2022] [Indexed: 01/31/2023] Open
Abstract
Ischemia-reperfusion injury (IRI) amplifies T cell alloimmune responses after transplantation with thrombin playing a key pro-inflammatory role. To explore the influence of thrombin on regulatory T cell recruitment and efficacy we used a well-established model of IRI in the native murine kidney. Administration of the cytotopic thrombin inhibitor PTL060 inhibited IRI, and by skewing expression of chemokines (reducing CCL2 and CCL3 but increasing CCL17 and CCL22) increased the infiltration of M2 macrophages and Tregs. When PTL060 was combined with infusion of additional Tregs, these effects were further amplified. To test the benefits of thrombin inhibition in a transplant model, BALB/c hearts were transplanted into B6 mice with or without perfusion with PTL060 in combination with Tregs. Thrombin inhibition or Treg infusion alone led to small increments in allograft survival. However, the combined therapy led to modest graft prolongation by the same mechanisms as in renal IRI; graft survival was accompanied by increased numbers of Tregs and anti-inflammatory macrophages, and reduced expression of pro-inflammatory cytokines. While the grafts succumbed to rejection associated with the emergence of alloantibody, these data suggest that thrombin inhibition within the transplant vasculature enhances the efficacy of Treg infusion, a therapy that is currently entering the clinic to promote transplant tolerance.
Collapse
Affiliation(s)
- Qi Peng
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Mucosal Biology, King’s College London, London, United Kingdom
| | - Anna Nowocin
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Mucosal Biology, King’s College London, London, United Kingdom
| | - Kulachelvy Ratnasothy
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Mucosal Biology, King’s College London, London, United Kingdom
| | - Richard A. Smith
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Mucosal Biology, King’s College London, London, United Kingdom
| | - Lesley A. Smyth
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Mucosal Biology, King’s College London, London, United Kingdom,School of Health, Sport and Bioscience, University of East London, London, United Kingdom
| | - Robert I. Lechler
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Mucosal Biology, King’s College London, London, United Kingdom
| | - Anthony Dorling
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Mucosal Biology, King’s College London, London, United Kingdom
| | - Giovanna Lombardi
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Mucosal Biology, King’s College London, London, United Kingdom,*Correspondence: Giovanna Lombardi,
| |
Collapse
|
5
|
Wilkinson H, Leonard H, Robson MG, Smith R, Tam E, McVey JH, Kirckhofer D, Chen D, Dorling A. Manipulation of tissue factor-mediated basal PAR-2 signalling on macrophages determines sensitivity for IFNγ responsiveness and significantly modifies the phenotype of murine DTH. Front Immunol 2022; 13:999871. [PMID: 36172348 PMCID: PMC9510775 DOI: 10.3389/fimmu.2022.999871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundTissue factor (TF) generates proteases that can signal through PAR-1 and PAR-2. We have previously demonstrated PAR-1 signalling primes innate myeloid cells to be exquisitely sensitive to interferon-gamma (IFNγ). In this work we explored how TF mediated PAR-2 signalling modulated responsiveness to IFNγ and investigated the interplay between PAR-1/-2 signalling on macrophages.MethodologyWe characterised how TF through PAR-2 influenced IFNγ sensitivity in vitro using PCR and flow cytometry. and how it influenced oxazolone-induced delayed type hypersensitivity (DTH) responses in vivo. We investigated how basal signalling through PAR-2 influenced PAR-1 signalling using a combination of TF-inhibitors and PAR-1 &-2 agonists and antagonists. Finally, we investigated whether this system could be targeted therapeutically using 3-mercaptopropionyl-F-Cha-Cha-RKPNDK (3-MP), which has actions on both PAR-1 and -2.ResultsTF delivered a basal signal through PAR-2 that upregulated SOCS3 expression and blunted M1 polarisation after IFNγ stimulation, opposing the priming achieved by signalling through PAR-1. PAR-1 and -2 agonists or antagonists could be used in combination to modify this basal signal in vitro and in vivo. 3-MP, by virtue of its PAR-2 agonist properties was superior to agents with only PAR-1 antagonist properties at reducing M1 polarisation induced by IFNγ and suppressing DTH. Tethering a myristoyl electrostatic switch almost completely abolished the DTH response.ConclusionsTF-mediated signalling through PARs-1 and -2 act in a homeostatic way to determine how myeloid cells respond to IFNγ. 3-MP, an agent that simultaneously inhibits PAR-1 whilst delivering a PAR-2 signal, can almost completely abolish immune responses dependent on M1 polarisation, particularly if potency is enhanced by targeting to cell membranes; this has potential therapeutic potential in multiple diseases.
Collapse
Affiliation(s)
- Hannah Wilkinson
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
- *Correspondence: Anthony Dorling, ; Hannah Wilkinson,
| | - Hugh Leonard
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Michael G. Robson
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Richard Smith
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
| | - ElLi Tam
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
| | - John H. McVey
- School of Bioscience & Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Daniel Kirckhofer
- Department of Early Discovery Biochemistry, Genentech Inc., South San Francisco, CA, United States
| | - Daxin Chen
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Anthony Dorling
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
- *Correspondence: Anthony Dorling, ; Hannah Wilkinson,
| |
Collapse
|
6
|
Shi K, Liu Y, Zhang Q, Ran CP, Hou J, Zhang Y, Wang XB. Severe Type of COVID-19: Pathogenesis, Warning Indicators and Treatment. Chin J Integr Med 2021; 28:3-11. [PMID: 34962616 PMCID: PMC8713541 DOI: 10.1007/s11655-021-3313-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2021] [Indexed: 12/15/2022]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2, is a major public health issue. The epidemic is unlikely to be contained until the global launch of safe and effective vaccines that could prevent serious illnesses and provide herd immunity. Although most patients have mild flu-like symptoms, some develop severe illnesses accompanied by multiple organ dysfunction. The identification of pathophysiology and early warning biomarkers of a severe type of COVID-19 contribute to the treatment and prevention of serious complications. Here, we review the pathophysiology, early warning indicators, and effective treatment of Chinese and Western Medicine for patients with a severe type of COVID-19.
Collapse
Affiliation(s)
- Ke Shi
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Department of Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100007, China
| | - Yao Liu
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Qun Zhang
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Chong-Ping Ran
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Jie Hou
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Department of Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100007, China
| | - Yi Zhang
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Xian-Bo Wang
- Department of Gastroenterology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100007, China.
| |
Collapse
|
7
|
Chu SJ, Tang SE, Pao HP, Wu SY, Liao WI. Protease-Activated Receptor-1 Antagonist Protects Against Lung Ischemia/Reperfusion Injury. Front Pharmacol 2021; 12:752507. [PMID: 34658893 PMCID: PMC8514687 DOI: 10.3389/fphar.2021.752507] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/20/2021] [Indexed: 01/14/2023] Open
Abstract
Protease-activated receptor (PAR)-1 is a thrombin-activated receptor that plays an essential role in ischemia/reperfusion (IR)-induced acute inflammation. PAR-1 antagonists have been shown to alleviate injuries in various IR models. However, the effect of PAR-1 antagonists on IR-induced acute lung injury (ALI) has not yet been elucidated. This study aimed to investigate whether PAR-1 inhibition could attenuate lung IR injury. Lung IR was induced in an isolated perfused rat lung model. Male rats were treated with the specific PAR-1 antagonist SCH530348 (vorapaxar) or vehicle, followed by ischemia for 40 min and reperfusion for 60 min. To examine the role of PAR-1 and the mechanism of SCH530348 in lung IR injury, western blotting and immunohistochemical analysis of lung tissue were performed. In vitro, mouse lung epithelial cells (MLE-12) were treated with SCH530348 or vehicle and subjected to hypoxia-reoxygenation (HR). We found that SCH530348 decreased lung edema and neutrophil infiltration, attenuated thrombin production, reduced inflammatory factors, including cytokine-induced neutrophil chemoattractant-1, interleukin-6 and tumor necrosis factor-α, mitigated lung cell apoptosis, and downregulated the phosphoinositide 3-kinase (PI3K), nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) pathways in IR-injured lungs. In addition, SCH530348 prevented HR-induced NF-κB activation and inflammatory chemokine production in MLE12 cells. Our results demonstrate that SCH530348 exerts protective effects by blocking PAR-1 expression and modulating the downstream PI3K, NF-κB and MAPK pathways. These findings indicate that the PAR-1 antagonist protects against IR-induced ALI and is a potential therapeutic candidate for lung protection following IR injury.
Collapse
Affiliation(s)
- Shi-Jye Chu
- Department of Internal Medicine, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
| | - Shih-En Tang
- Division of Pulmonary and Critical Care, Department of Internal Medicine, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan.,Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Hsin-Ping Pao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Yu Wu
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Wen-I Liao
- Department of Emergency Medicine, National Defense Medical Center, Tri-Service General Hospital, Taipei, Taiwan
| |
Collapse
|
8
|
D’Amélio F, Vigerelli H, de Brandão Prieto da Silva ÁR, Kerkis I. Bothrops moojeni venom and its components - an overview. JOURNAL OF VENOM RESEARCH 2021; 11:26-33. [PMID: 34123362 PMCID: PMC8169028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 05/28/2021] [Accepted: 05/28/2021] [Indexed: 11/23/2022]
Abstract
Belonging to the Viperidae family, Bothrops moojeni are widely distributed in South America, tropical savanna ecoregion (Cerrado) of Argentina, Bolivia, Brazil, and Paraguay with medical importance in Brazil. Accidents caused by this species have a rapid local action with the development of tissue inflammation, causing erythema, pain, and increased clotting time, which can culminate in gangrene or tissue necrosis. Bothrops moojeni venom has a rich composition that remains underexplored, which is of utmost importance, both for elucidating the envenoming process and the vast library of new bioactive molecules kind of venom can offer. This review aims to analyze which components of the venom have already been characterized towards its structure and biological effect and highlight the pharmacological and biotechnological potential of this venom. Although snake venoms have been studied for their toxic effects for generations, innovative studies address their components as tools for discovering new therapeutic targets and new molecules with pharmacological and biotechnological potential.
Collapse
Affiliation(s)
- Fernanda D’Amélio
- 1Laboratory of Genetics, Butantan Institute, São Paulo, SP, Brazil,2The Postgraduate Program in Toxinology, Butantan Institute, São Paulo, SP, Brazil,*Correspondence to: Irina Kerkis, and Fernanda D’Amélio,
| | - Hugo Vigerelli
- 1Laboratory of Genetics, Butantan Institute, São Paulo, SP, Brazil,2The Postgraduate Program in Toxinology, Butantan Institute, São Paulo, SP, Brazil,3Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo, SP, Brazil
| | | | - Irina Kerkis
- 1Laboratory of Genetics, Butantan Institute, São Paulo, SP, Brazil,3Centre of Excellence in New Target Discovery (CENTD), Butantan Institute, São Paulo, SP, Brazil,*Correspondence to: Irina Kerkis, and Fernanda D’Amélio,
| |
Collapse
|
9
|
Wilkinson H, Leonard H, Chen D, Lawrence T, Robson M, Goossens P, McVey JH, Dorling A. PAR-1 signaling on macrophages is required for effective in vivo delayed-type hypersensitivity responses. iScience 2021; 24:101981. [PMID: 33458623 PMCID: PMC7797913 DOI: 10.1016/j.isci.2020.101981] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 11/13/2020] [Accepted: 12/17/2020] [Indexed: 12/15/2022] Open
Abstract
Delayed-type hypersensitivity (DTH) responses underpin chronic inflammation. Using a model of oxazolone-induced dermatitis and a combination of transgenic mice, adoptive cell transfer, and selective agonists/antagonists against protease activated receptors, we show that that PAR-1 signaling on macrophages by thrombin is required for effective granuloma formation. Using BM-derived macrophages (BMMs) in vitro, we show that thrombin signaling induced (a) downregulation of cell membrane reverse cholesterol transporter ABCA1 and (b) increased expression of IFNγ receptor and enhanced co-localization within increased areas of cholesterol-rich membrane microdomains. These two key phenotypic changes combined to make thrombin-primed BMMs sensitive to M1 polarization by 1000-fold less IFNγ, compared to resting BMMs. We confirm that changes in ABCA1 expression were directly responsible for the exquisite sensitivity to IFNγ in vitro and for the impact on granuloma formation in vivo. These data indicate that PAR-1 signaling plays a hitherto unrecognized and critical role in DTH responses.
Collapse
Affiliation(s)
- Hannah Wilkinson
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Hugh Leonard
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Daxin Chen
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Toby Lawrence
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology & Microbial Sciences, King's College London, London SE1 9RT, UK
| | - Michael Robson
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Pieter Goossens
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229HX Maastricht, the Netherlands
| | - John H McVey
- School of Bioscience & Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Anthony Dorling
- Department of Inflammation Biology, School of Immunology & Microbial Sciences, King's College London, Guy's Hospital, London SE1 9RT, UK
| |
Collapse
|
10
|
Schweickert PG, Yang Y, White EE, Cresswell GM, Elzey BD, Ratliff TL, Arumugam P, Antoniak S, Mackman N, Flick MJ, Konieczny SF. Thrombin-PAR1 signaling in pancreatic cancer promotes an immunosuppressive microenvironment. J Thromb Haemost 2021; 19:161-172. [PMID: 33064371 PMCID: PMC7790967 DOI: 10.1111/jth.15115] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 09/11/2020] [Accepted: 09/23/2020] [Indexed: 12/18/2022]
Abstract
Essentials Elimination of PDAC tumor cell PAR1 increased cytotoxic T cells and reduced tumor macrophages. PAR1KO PDAC cells are preferentially eliminated from growing tumors. Thrombin-PAR1 signaling in PDAC tumor cells drives an immunosuppressive gene signature. Csf2 and Ptgs2 are thrombin-PAR1 downstream immune suppressor genes in PDAC tumor cells. ABSTRACT: Background Pancreatic ductal adenocarcinoma (PDAC) is characterized by a prothrombotic state and a lack of host antitumor immune responsiveness. Linking these two key features, we previously demonstrated that tumor-derived coagulation activity promotes immune evasion. Specifically, thrombin-protease-activated receptor-1 (PAR1) signaling in mouse PDAC cells drives tumor growth by evading cytotoxic CD8a+ cells. Methods Syngeneic mixed cell tumor growth, transcriptional analyses, and functional tests of immunosuppressive response genes were used to identify cellular and molecular immune evasion mechanisms mediated by thrombin-PAR-1 signaling in mouse PDAC tumor cells. Results Elimination of tumor cell PAR1 in syngeneic graft studies increased cytotoxic T lymphocyte (CTL) infiltration and decreased tumor-associated macrophages in the tumor microenvironment. Co-injection of PAR1-expressing and PAR1-knockout (PAR-1KO ) tumor cells into immunocompetent mice resulted in preferential elimination of PAR-1KO cells from developing tumors, suggesting that PAR1-dependent immune evasion is not reliant on CTL exclusion. Transcriptomics analyses revealed no PAR1-dependent changes in the expression of immune checkpoint proteins and no difference in major histocompatibility complex-I cell surface expression. Importantly, thrombin-PAR1 signaling in PDAC cells upregulated genes linked to immunosuppression, including Csf2 and Ptgs2. Functional analyses confirmed that both Csf2 and Ptgs2 are critical for PDAC syngeneic graft tumor growth and overexpression of each factor partially restored tumor growth of PAR1KO cells in immunocompetent mice. Conclusions Our results provide novel insight into the mechanisms of a previously unrecognized pathway coupling coagulation to PDAC immune evasion by identifying PAR1-dependent changes in the tumor microenvironment, a PAR1-driven immunosuppressive gene signature, and Csf2 and Ptgs2 as critical PAR1 downstream targets.
Collapse
Affiliation(s)
- Patrick G. Schweickert
- Purdue University, Department of Biological Sciences and
the Purdue Center for Cancer Research, West Lafayette, Indiana, USA
| | - Yi Yang
- University of North Carolina, Department of Pathology and
Laboratory Medicine, the Lineberger Comprehensive Cancer Center, and the UNC Blood
Research Center, Chapel Hill, North Carolina, USA
| | - Emily E. White
- Purdue University, Department of Biological Sciences and
the Purdue Center for Cancer Research, West Lafayette, Indiana, USA
| | - Gregory M. Cresswell
- Purdue University, Department of Comparative Pathobiology
and the Purdue Center for Cancer Research, West Lafayette, Indiana, USA
| | - Bennett D. Elzey
- Purdue University, Department of Comparative Pathobiology
and the Purdue Center for Cancer Research, West Lafayette, Indiana, USA
| | - Timothy L. Ratliff
- Purdue University, Department of Comparative Pathobiology
and the Purdue Center for Cancer Research, West Lafayette, Indiana, USA
| | - Paritha Arumugam
- Cincinnati Children’s Hospital Medical Center,
Division of Pulmonary Biology, Cincinnati, Ohio, USA
| | - Silvio Antoniak
- University of North Carolina, Department of Pathology and
Laboratory Medicine, the Lineberger Comprehensive Cancer Center, and the UNC Blood
Research Center, Chapel Hill, North Carolina, USA
| | - Nigel Mackman
- University of North Carolina, Department of Medicine and
the UNC Blood Research Center, Chapel Hill, North Carolina, USA
| | - Matthew J. Flick
- University of North Carolina, Department of Pathology and
Laboratory Medicine, the Lineberger Comprehensive Cancer Center, and the UNC Blood
Research Center, Chapel Hill, North Carolina, USA
| | - Stephen F. Konieczny
- Purdue University, Department of Biological Sciences and
the Purdue Center for Cancer Research, West Lafayette, Indiana, USA
| |
Collapse
|
11
|
Abstract
The recent emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the ensuing global pandemic has presented a health emergency of unprecedented magnitude. Recent clinical data has highlighted that coronavirus disease 2019 (COVID-19) is associated with a significant risk of thrombotic complications ranging from microvascular thrombosis, venous thromboembolic disease, and stroke. Importantly, thrombotic complications are markers of severe COVID-19 and are associated with multiorgan failure and increased mortality. The evidence to date supports the concept that the thrombotic manifestations of severe COVID-19 are due to the ability of SARS-CoV-2 to invade endothelial cells via ACE-2 (angiotensin-converting enzyme 2), which is expressed on the endothelial cell surface. However, in patients with COVID-19 the subsequent endothelial inflammation, complement activation, thrombin generation, platelet, and leukocyte recruitment, and the initiation of innate and adaptive immune responses culminate in immunothrombosis, ultimately causing (micro)thrombotic complications, such as deep vein thrombosis, pulmonary embolism, and stroke. Accordingly, the activation of coagulation (eg, as measured with plasma D-dimer) and thrombocytopenia have emerged as prognostic markers in COVID-19. Given thrombotic complications are central determinants of the high mortality rate in COVID-19, strategies to prevent thrombosis are of critical importance. Several antithrombotic drugs have been proposed as potential therapies to prevent COVID-19-associated thrombosis, including heparin, FXII inhibitors, fibrinolytic drugs, nafamostat, and dipyridamole, many of which also possess pleiotropic anti-inflammatory or antiviral effects. The growing awareness and mechanistic understanding of the prothrombotic state of COVID-19 patients are driving efforts to more stringent diagnostic screening for thrombotic complications and to the early institution of antithrombotic drugs, for both the prevention and therapy of thrombotic complications. The shifting paradigm of diagnostic and treatment strategies holds significant promise to reduce the burden of thrombotic complications and ultimately improve the prognosis for patients with COVID-19.
Collapse
Affiliation(s)
- James D. McFadyen
- From the Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.D.M., H.S., K.P.)
- Clinical Hematology Department (J.D.M., H.S.), Alfred Hospital, Melbourne, Victoria, Australia
- Department of Medicine, Monash University, Melbourne, Victoria, Australia (J.D.M., H.S., K.P.)
| | - Hannah Stevens
- From the Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (J.D.M., H.S., K.P.)
- Clinical Hematology Department (J.D.M., H.S.), Alfred Hospital, Melbourne, Victoria, Australia
- Department of Medicine, Monash University, Melbourne, Victoria, Australia (J.D.M., H.S., K.P.)
| | - Karlheinz Peter
- Department of Cardiology (K.P.), Alfred Hospital, Melbourne, Victoria, Australia
- Department of Medicine, Monash University, Melbourne, Victoria, Australia (J.D.M., H.S., K.P.)
| |
Collapse
|
12
|
Chen D, Li K, Festenstein S, Karegli J, Wilkinson H, Leonard H, Wei L, Ma N, Xia M, Tam H, Wang J, Xu Q, McVey JH, Smith RAG, Dorling A. Regression of Atherosclerosis in ApoE-/- Mice Via Modulation of Monocyte Recruitment and Phenotype, Induced by Weekly Dosing of a Novel "Cytotopic" Anti-Thrombin Without Prolonged Anticoagulation. J Am Heart Assoc 2020; 9:e014811. [PMID: 32611229 PMCID: PMC7670518 DOI: 10.1161/jaha.119.014811] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 06/22/2020] [Indexed: 01/08/2023]
Abstract
Background Anticoagulants induce atherosclerosis regression in animal models but exploiting this clinically is limited by bleeding events. Here we test a novel thrombin inhibitor, PTL060, comprising hirulog covalently linked to a synthetic myristoyl electrostatic switch to tether to cell membranes. Methods and Results ApoE-/- mice were fed chow or high-fat diets, before transplantation of congenic aortic segments or injection of PTL060, parental hirulog, control saline, or labeled CD11b positive cells. Aortic transplants from transgenic mice expressing anticoagulants on endothelium did not develop atherosclerosis. A single intravenous injection of PTL060, but not hirulog inhibited atheroma development by >50% compared with controls when assessed 4 weeks later. Mice had prolonged bleeding times for only one seventh of the time that PTL060 was biologically active. Repeated weekly injections of PTL060 but not hirulog caused regression of atheroma. We dissected 2 contributory mechanisms. First, the majority of CCR2+ (C-C chemokine receptor type 2+) monocytes recruited into plaques expressed CCR7 (C-C chemokine receptor type 7), ABCA1 (ATP-binding cassette transporter - 1), and interleukin-10 in PTL060 mice, a phenotype seen in <20% of CCR2+ recruits in controls. Second, after several doses, there was a significant reduction in monocyte recruits, the majority of which were CCR2-negative with a similar regression-associated phenotype. Regression equivalent to that induced by intravenous PTL060 was induced by adoptive transfer of CD11b+ cells pre-coated with PTL060. Conclusions Covalent linkage of a myristoyl electrostatic switch onto hirulog in PTL060 uncouples the pharmacodynamic effects on hemostasis and atherosclerosis, such that plaque regression, mediated predominantly via effects on monocytes, is accompanied by only transient anticoagulation.
Collapse
Affiliation(s)
- Daxin Chen
- Department of Inflammation BiologySchool of Immunology and Microbial SciencesKing’s College London, Guy’s HospitalLondonUnited Kingdom
| | - Ke Li
- Core Research Laboratorythe Second Affiliated Hospital, School of MedicineJiaotong UniversityXi’anChina
| | - Sam Festenstein
- Department of Inflammation BiologySchool of Immunology and Microbial SciencesKing’s College London, Guy’s HospitalLondonUnited Kingdom
| | - Julieta Karegli
- Department of Inflammation BiologySchool of Immunology and Microbial SciencesKing’s College London, Guy’s HospitalLondonUnited Kingdom
| | - Hannah Wilkinson
- Department of Inflammation BiologySchool of Immunology and Microbial SciencesKing’s College London, Guy’s HospitalLondonUnited Kingdom
| | - Hugh Leonard
- Department of Inflammation BiologySchool of Immunology and Microbial SciencesKing’s College London, Guy’s HospitalLondonUnited Kingdom
| | - Lin‐Lin Wei
- Core Research Laboratorythe Second Affiliated Hospital, School of MedicineJiaotong UniversityXi’anChina
| | - Ning Ma
- Core Research Laboratorythe Second Affiliated Hospital, School of MedicineJiaotong UniversityXi’anChina
| | - Min Xia
- Thrombosis Research InstituteLondonUnited Kingdom
| | - Henry Tam
- Department of ImagingImperial College Healthcare NHS TrustCharing Cross HospitalLondonUnited Kingdom
| | - Jian‐an Wang
- Department of CardiologySecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
| | - Qingbo Xu
- Cardiovascular DivisionKing’s College LondonJames Black CentreLondonUnited Kingdom
| | - John H. McVey
- School of Bioscience & MedicineFaculty of Health and Medical SciencesUniversity of SurreyGuildfordUnited Kingdom
| | - Richard A. G. Smith
- Department of Inflammation BiologySchool of Immunology and Microbial SciencesKing’s College London, Guy’s HospitalLondonUnited Kingdom
| | - Anthony Dorling
- Department of Inflammation BiologySchool of Immunology and Microbial SciencesKing’s College London, Guy’s HospitalLondonUnited Kingdom
| |
Collapse
|
13
|
Cantrell R, Palumbo JS. The thrombin–inflammation axis in cancer progression. Thromb Res 2020; 191 Suppl 1:S117-S122. [DOI: 10.1016/s0049-3848(20)30408-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/18/2019] [Accepted: 11/21/2019] [Indexed: 02/07/2023]
|
14
|
Xin JZ, Wu JM, Hu GM, Gu HJ, Feng YN, Wang SX, Cong WW, Li MZ, Xu WL, Song Y, Xiao H, Zhang YY, Wang L. α 1-AR overactivation induces cardiac inflammation through NLRP3 inflammasome activation. Acta Pharmacol Sin 2020; 41:311-318. [PMID: 31530901 PMCID: PMC7468364 DOI: 10.1038/s41401-019-0305-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 08/29/2019] [Indexed: 01/08/2023] Open
Abstract
Acute sympathetic stress causes excessive secretion of catecholamines and induces cardiac injuries, which are mainly mediated by β-adrenergic receptors (β-ARs). However, α1-adrenergic receptors (α1-ARs) are also expressed in the heart and are activated upon acute sympathetic stress. In the present study, we investigated whether α1-AR activation induced cardiac inflammation and the underlying mechanisms. Male C57BL/6 mice were injected with a single dose of α1-AR agonist phenylephrine (PE, 5 or 10 mg/kg, s.c.) with or without pretreatment with α-AR antagonist prazosin (5 mg/kg, s.c.). PE injection caused cardiac dysfunction and cardiac inflammation, evidenced by the increased expression of inflammatory cytokine IL-6 and chemokines MCP-1 and MCP-5, as well as macrophage infiltration in myocardium. These effects were blocked by prazosin pretreatment. Furthermore, PE injection significantly increased the expression of NOD-like receptor protein 3 (NLRP3) and the cleavage of caspase-1 (p20) and interleukin-18 in the heart; similar results were observed in both Langendorff-perfused hearts and cultured cardiomyocytes following the treatment with PE (10 μM). Moreover, PE-induced NLRP3 inflammasome activation and cardiac inflammation was blocked in Nlrp3-/- mice compared with wild-type mice. In conclusion, α1-AR overactivation induces cardiac inflammation by activating NLRP3 inflammasomes.
Collapse
|
15
|
Peng Q, Ratnasothy K, Boardman DA, Jacob J, Tung SL, McCluskey D, Smyth LA, Lechler RI, Dorling A, Lombardi G. Protease Activated Receptor 4 as a Novel Modulator of Regulatory T Cell Function. Front Immunol 2019; 10:1311. [PMID: 31275306 PMCID: PMC6591367 DOI: 10.3389/fimmu.2019.01311] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 05/23/2019] [Indexed: 01/19/2023] Open
Abstract
Regulatory T cells (Tregs) are a subpopulation of T cells that maintain immunological tolerance. In inflammatory responses the function of Tregs is tightly controlled by several factors including signaling through innate receptors such as Toll like receptors and anaphylatoxin receptors allowing an effective immune response to be generated. Protease-activated receptors (PARs) are another family of innate receptors expressed on multiple cell types and involved in the pathogenesis of autoimmune disorders. Whether proteases are able to directly modulate Treg function is unknown. Here, we show using two complimentary approaches that signaling through PAR-4 influences the expression of CD25, CD62L, and CD73, the suppressive capacity, and the stability of Tregs, via phosphorylation of FoxO1 and negative regulation of PTEN and FoxP3. Taken together, our results demonstrate an important role of PAR4 in tuning the function of Tregs and open the possibility of targeting PAR4 to modulate immune responses.
Collapse
Affiliation(s)
- Qi Peng
- MRC Centre for Transplantation, Guy's Hospital, King's College London, London, United Kingdom.,NIHR Biomedical Research Centre, Guy's Hospital, Guy's & St Thomas' NHS Foundation Trust, King's College London, London, United Kingdom
| | - Kulachelvy Ratnasothy
- MRC Centre for Transplantation, Guy's Hospital, King's College London, London, United Kingdom.,NIHR Biomedical Research Centre, Guy's Hospital, Guy's & St Thomas' NHS Foundation Trust, King's College London, London, United Kingdom
| | - Dominic A Boardman
- MRC Centre for Transplantation, Guy's Hospital, King's College London, London, United Kingdom.,NIHR Biomedical Research Centre, Guy's Hospital, Guy's & St Thomas' NHS Foundation Trust, King's College London, London, United Kingdom
| | - Jacinta Jacob
- MRC Centre for Transplantation, Guy's Hospital, King's College London, London, United Kingdom.,NIHR Biomedical Research Centre, Guy's Hospital, Guy's & St Thomas' NHS Foundation Trust, King's College London, London, United Kingdom
| | - Sim Lai Tung
- MRC Centre for Transplantation, Guy's Hospital, King's College London, London, United Kingdom.,NIHR Biomedical Research Centre, Guy's Hospital, Guy's & St Thomas' NHS Foundation Trust, King's College London, London, United Kingdom
| | - Daniel McCluskey
- MRC Centre for Transplantation, Guy's Hospital, King's College London, London, United Kingdom
| | - Lesley A Smyth
- MRC Centre for Transplantation, Guy's Hospital, King's College London, London, United Kingdom.,School of Health, Sport and Bioscience, University of East London, London, United Kingdom
| | - Robert I Lechler
- MRC Centre for Transplantation, Guy's Hospital, King's College London, London, United Kingdom.,NIHR Biomedical Research Centre, Guy's Hospital, Guy's & St Thomas' NHS Foundation Trust, King's College London, London, United Kingdom
| | - Anthony Dorling
- MRC Centre for Transplantation, Guy's Hospital, King's College London, London, United Kingdom.,NIHR Biomedical Research Centre, Guy's Hospital, Guy's & St Thomas' NHS Foundation Trust, King's College London, London, United Kingdom
| | - Giovanna Lombardi
- MRC Centre for Transplantation, Guy's Hospital, King's College London, London, United Kingdom.,NIHR Biomedical Research Centre, Guy's Hospital, Guy's & St Thomas' NHS Foundation Trust, King's College London, London, United Kingdom
| |
Collapse
|
16
|
Lou J, Hu Y, Wu MD, Che LQ, Wu YF, Zhao Y, Tian BP, Bao ZQ, Zhu C, Wu YP, He LL, Bai CX, Zhou J, Ying SM, Li W, Chen ZH, Chen DX, Dorling A, Shen HH. Endothelial cell-specific anticoagulation reduces inflammation in a mouse model of acute lung injury. Acta Pharmacol Sin 2019; 40:769-780. [PMID: 30446733 DOI: 10.1038/s41401-018-0175-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 09/24/2018] [Indexed: 01/11/2023] Open
Abstract
Tissue factor (TF)-dependent coagulation contributes to lung inflammation and the pathogenesis of acute lung injury (ALI). In this study, we explored the roles of targeted endothelial anticoagulation in ALI using two strains of transgenic mice expressing either a membrane-tethered human tissue factor pathway inhibitor (hTFPI) or hirudin fusion protein on CD31+ cells, including vascular endothelial cells (ECs). ALI was induced by intratracheal injection of LPS, and after 24 h the expression of TF and protease-activated receptors (PARs) on EC in lungs were assessed, alongside the extent of inflammation and injury. The expression of TF and PARs on the EC in lungs was upregulated after ALI. In the two strains of transgenic mice, expression of either of hTFPI or hirudin by EC was associated with significant reduction of inflammation, as assessed by the extent of leukocyte infiltration or the levels of proinflammatory cytokines, and promoted survival after LPS-induced ALI. The beneficial outcomes were associated with inhibition of the expression of chemokine CCL2 in lung tissues. The protection observed in the CD31-TFPI-transgenic strain was abolished by injection of an anti-hTFPI antibody, but not by prior engraftment of the transgenic strains with WT bone marrow, confirming that the changes observed were a specific transgenic expression of anticoagulants by EC. These results demonstrate that the inflammation in ALI is TF and thrombin dependent, and that expression of anticoagulants by EC significantly inhibits the development of ALI via repression of leukocyte infiltration, most likely via inhibition of chemokine gradients. These data enhance our understanding of the pathology of ALI and suggest a novel therapeutic strategy for treatment.
Collapse
|
17
|
Festoff BW, Citron BA. Thrombin and the Coag-Inflammatory Nexus in Neurotrauma, ALS, and Other Neurodegenerative Disorders. Front Neurol 2019; 10:59. [PMID: 30804878 PMCID: PMC6371052 DOI: 10.3389/fneur.2019.00059] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/17/2019] [Indexed: 12/15/2022] Open
Abstract
This review details our current understanding of thrombin signaling in neurodegeneration, with a focus on amyotrophic lateral sclerosis (ALS, Lou Gehrig's disease) as well as future directions to be pursued. The key factors are multifunctional and involved in regulatory pathways, namely innate immune and the coagulation cascade activation, that are essential for normal nervous system function and health. These two major host defense systems have a long history in evolution and include elements and regulators of the coagulation pathway that have significant impacts on both the peripheral and central nervous system in health and disease. The clotting cascade responds to a variety of insults to the CNS including injury and infection. The blood brain barrier is affected by these responses and its compromise also contributes to these detrimental effects. Important molecules in signaling that contribute to or protect against neurodegeneration include thrombin, thrombomodulin (TM), protease activated receptor 1 (PAR1), damage associated molecular patterns (DAMPs), such as high mobility group box protein 1 (HMGB1) and those released from mitochondria (mtDAMPs). Each of these molecules are entangled in choices dependent upon specific signaling pathways in play. For example, the particular cleavage of PAR1 by thrombin vs. activated protein C (APC) will have downstream effects through coupled factors to result in toxicity or neuroprotection. Furthermore, numerous interactions influence these choices such as the interplay between HMGB1, thrombin, and TM. Our hope is that improved understanding of the ways that components of the coagulation cascade affect innate immune inflammatory responses and influence the course of neurodegeneration, especially after injury, will lead to effective therapeutic approaches for ALS, traumatic brain injury, and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Barry W Festoff
- pHLOGISTIX LLC, Fairway, KS, United States.,Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Bruce A Citron
- Laboratory of Molecular Biology Research & Development, VA New Jersey Health Care System, East Orange, NJ, United States.,Department of Pharmacology, Physiology & Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, United States
| |
Collapse
|
18
|
Li WZ, Yang Y, Liu K, Long R, Jin N, Huang SY, You Y, Dai J, Fan C, Wang J, Wang ZH. FGL2 prothrombinase contributes to the early stage of coronary microvascular obstruction through a fibrin-dependent pathway. Int J Cardiol 2019; 274:27-34. [DOI: 10.1016/j.ijcard.2018.09.051] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/27/2018] [Accepted: 09/12/2018] [Indexed: 12/20/2022]
|
19
|
Jaberi N, Soleimani A, Pashirzad M, Abdeahad H, Mohammadi F, Khoshakhlagh M, Khazaei M, Ferns GA, Avan A, Hassanian SM. Role of thrombin in the pathogenesis of atherosclerosis. J Cell Biochem 2018; 120:4757-4765. [DOI: 10.1002/jcb.27771] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 09/06/2018] [Indexed: 01/22/2023]
Affiliation(s)
- Najmeh Jaberi
- Department of Medical Biochemistry Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | - Atena Soleimani
- Department of Medical Biochemistry Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | - Mehran Pashirzad
- Department of Medical Biochemistry Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | - Hosein Abdeahad
- Department of Medical Biochemistry Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | - Fariba Mohammadi
- Department of Medical Biochemistry Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | - Mahdieh Khoshakhlagh
- Department of Medical Biochemistry Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | - Majid Khazaei
- Department of Medical Physiology Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | - Gordon A Ferns
- Division of Medical Education Brighton and Sussex Medical School Sussex UK
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences Mashhad Iran
- Department of Modern Sciences and Technologies School of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | - Seyed Mahdi Hassanian
- Department of Medical Biochemistry Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
20
|
Lessard AJ, LeBel M, Egarnes B, Préfontaine P, Thériault P, Droit A, Brunet A, Rivest S, Gosselin J. Triggering of NOD2 Receptor Converts Inflammatory Ly6C high into Ly6C low Monocytes with Patrolling Properties. Cell Rep 2018; 20:1830-1843. [PMID: 28834747 DOI: 10.1016/j.celrep.2017.08.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/16/2017] [Accepted: 07/28/2017] [Indexed: 01/09/2023] Open
Abstract
The signals that regulate the fate of circulating monocytes remain unknown. In the present study, we demonstrate that triggering of the NOD2 receptor by muramyl dipeptide (MDP) converts inflammatory Ly6Chigh monocytes into patrolling Ly6Clow monocytes. Administration of MDP to Nr4a1-/- mice, which lack Ly6Clow monocytes, or to Ly6Clow-depleted mice led to the emergence of blood-patrolling monocytes with a profile similar to that of Ly6Clow monocytes, including high expression of CX3CR1 and LFA1. Using intravital microscopy in animal models of inflammatory diseases, we also found that converted Ly6Chigh monocytes patrol the endothelium of blood vessels and that their presence contributes to a reduction in the inflammatory response following MDP injection. Our results demonstrate that NOD2 contributes to the regulation of blood monocytes and suggest that it could be therapeutically targeted to treat inflammatory diseases.
Collapse
Affiliation(s)
- Anne-Julie Lessard
- Laboratory of Innate Immunology, Centre de recherche du CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
| | - Manon LeBel
- Laboratory of Innate Immunology, Centre de recherche du CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
| | - Benoit Egarnes
- Laboratory of Innate Immunology, Centre de recherche du CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
| | - Paul Préfontaine
- Centre de recherche du CHU de Québec, Université Laval, Québec, QC G1V 4G2, Canada
| | - Peter Thériault
- Centre de recherche du CHU de Québec, Université Laval, Québec, QC G1V 4G2, Canada
| | - Arnaud Droit
- Department of Molecular Medicine, Université Laval, Québec, QC G1V 4G2, Canada; Centre de recherche du CHU de Québec, Université Laval, Québec, QC G1V 4G2, Canada
| | - Alexandre Brunet
- Department of Molecular Medicine, Université Laval, Québec, QC G1V 4G2, Canada; Centre de recherche du CHU de Québec, Université Laval, Québec, QC G1V 4G2, Canada
| | - Serge Rivest
- Department of Molecular Medicine, Université Laval, Québec, QC G1V 4G2, Canada; Centre de recherche du CHU de Québec, Université Laval, Québec, QC G1V 4G2, Canada
| | - Jean Gosselin
- Laboratory of Innate Immunology, Centre de recherche du CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada; Department of Molecular Medicine, Université Laval, Québec, QC G1V 4G2, Canada.
| |
Collapse
|
21
|
Salehi S, Sosa RA, Jin YP, Kageyama S, Fishbein MC, Rozengurt E, Kupiec-Weglinski JW, Reed EF. Outside-in HLA class I signaling regulates ICAM-1 clustering and endothelial cell-monocyte interactions via mTOR in transplant antibody-mediated rejection. Am J Transplant 2018; 18:1096-1109. [PMID: 29045076 PMCID: PMC5904014 DOI: 10.1111/ajt.14544] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 10/05/2017] [Accepted: 10/07/2017] [Indexed: 01/25/2023]
Abstract
Antibody-mediated rejection (AMR) resulting in transplant allograft vasculopathy (TAV) is the major obstacle for long-term survival of solid organ transplants. AMR is caused by donor-specific antibodies to HLA, which contribute to TAV by initiating outside-in signaling transduction pathways that elicit monocyte recruitment to activated endothelium. Mechanistic target of rapamycin (mTOR) inhibitors can attenuate TAV; therefore, we sought to understand the mechanistic underpinnings of mTOR signaling in HLA class I Ab-mediated endothelial cell activation and monocyte recruitment. We used an in vitro model to assess monocyte binding to HLA I Ab-activated endothelial cells and found mTOR inhibition reduced ezrin/radixin/moesin (ERM) phosphorylation, intercellular adhesion molecule 1 (ICAM-1) clustering, and monocyte firm adhesion to HLA I Ab-activated endothelium. Further, in a mouse model of AMR, in which C57BL/6. RAG1-/- recipients of BALB/c cardiac allografts were passively transferred with donor-specific MHC I antibodies, mTOR inhibition significantly reduced vascular injury, ERM phosphorylation, and macrophage infiltration of the allograft. Taken together, these studies indicate mTOR inhibition suppresses ERM phosphorylation in endothelial cells, which impedes ICAM-1 clustering in response to HLA class I Ab and prevents macrophage infiltration into cardiac allografts. These findings indicate a novel therapeutic application for mTOR inhibitors to disrupt endothelial cell-monocyte interactions during AMR.
Collapse
Affiliation(s)
- Sahar Salehi
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Rebecca A. Sosa
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yi-Ping Jin
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Shoichi Kageyama
- Department of Surgery, University of California, Los Angeles, CA, USA
| | - Michael C. Fishbein
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Enrique Rozengurt
- Department of Medicine, University of California, Los Angeles, CA, USA
| | - Jerzy W. Kupiec-Weglinski
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Surgery, University of California, Los Angeles, CA, USA
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
22
|
The Role of NK Cells in Pig-to-Human Xenotransplantation. J Immunol Res 2017; 2017:4627384. [PMID: 29410970 PMCID: PMC5749293 DOI: 10.1155/2017/4627384] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 10/31/2017] [Indexed: 02/07/2023] Open
Abstract
Recruitment of human NK cells to porcine tissues has been demonstrated in pig organs perfused ex vivo with human blood in the early 1990s. Subsequently, the molecular mechanisms leading to adhesion and cytotoxicity in human NK cell-porcine endothelial cell (pEC) interactions have been elucidated in vitro to identify targets for therapeutic interventions. Specific molecular strategies to overcome human anti-pig NK cell responses include (1) blocking of the molecular events leading to recruitment (chemotaxis, adhesion, and transmigration), (2) expression of human MHC class I molecules on pECs that inhibit NK cells, and (3) elimination or blocking of pig ligands for activating human NK receptors. The potential of cell-based strategies including tolerogenic dendritic cells (DC) and regulatory T cells (Treg) and the latest progress using transgenic pigs genetically modified to reduce xenogeneic NK cell responses are discussed. Finally, we present the status of phenotypic and functional characterization of nonhuman primate (NHP) NK cells, essential for studying their role in xenograft rejection using preclinical pig-to-NHP models, and summarize key advances and important perspectives for future research.
Collapse
|
23
|
Hughes EL, Becker F, Flower RJ, Buckingham JC, Gavins FNE. Mast cells mediate early neutrophil recruitment and exhibit anti-inflammatory properties via the formyl peptide receptor 2/lipoxin A 4 receptor. Br J Pharmacol 2017; 174:2393-2408. [PMID: 28471519 DOI: 10.1111/bph.13847] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 04/19/2017] [Accepted: 04/21/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE In recent years, studies have focused on the resolution of inflammation, which can be achieved by endogenous anti-inflammatory agonists such as Annexin A1 (AnxA1). Here, we investigated the effects of mast cells (MCs) on early LPS-induced neutrophil recruitment and the involvement of the AnxA1-formyl peptide receptor 2/ALX (FPR2/ALX or lipoxin A4 receptor) pathway. EXPERIMENTAL APPROACH Intravital microscopy (IVM) was used to visualize and quantify the effects of LPS (10 μg per mouse i.p.) on murine mesenteric cellular interactions. Furthermore, the role that MCs play in these inflammatory responses was determined in vivo and in vitro, and effects of AnxA1 mimetic peptide Ac2-26 were assessed. KEY RESULTS LPS increased both neutrophil endothelial cell interactions within the mesenteric microcirculation and MC activation (determined by IVM and ruthenium red dye uptake), which in turn lead to the early stages of neutrophil recruitment. MC recruitment of neutrophils could be blocked by preventing the pro-inflammatory activation (using cromolyn sodium) or enhancing an anti-inflammatory phenotype (using Ac2-26) in MCs. Furthermore, MCs induced neutrophil migration in vitro, and MC stabilization enhanced the release of AnxA1 from neutrophils. Pharmacological approaches (such as the administration of FPR pan-antagonist Boc2, or the FPR2/ALX antagonist WRW4) revealed neutrophil FPR2/ALX to be important in this process. CONCLUSIONS AND IMPLICATIONS Data presented here provide evidence for a role of MCs, which are ideally positioned in close proximity to the vasculature, to act as sentinel cells in neutrophil extravasation and resolution of inflammation via the AnxA1-FPR2/ALX pathway.
Collapse
Affiliation(s)
- Ellen L Hughes
- Centre for Brain Sciences, Department of Medicine, Imperial College London, London, W12 0NN, UK
| | - Felix Becker
- Department for General and Visceral Surgery, University Hospital Muenster, 48149, Muenster, Germany
| | - Roderick J Flower
- Centre of Biochemical Pharmacology, Queen Mary University, London, EC1V 3AJ, UK
| | | | - Felicity N E Gavins
- Centre for Brain Sciences, Department of Medicine, Imperial College London, London, W12 0NN, UK.,Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Centre Shreveport, Shreveport, LA, 71130, USA
| |
Collapse
|
24
|
Laurance S, Bertin FR, Ebrahimian T, Kassim Y, Rys RN, Lehoux S, Lemarié CA, Blostein MD. Gas6 Promotes Inflammatory (CCR2 hiCX3CR1 lo) Monocyte Recruitment in Venous Thrombosis. Arterioscler Thromb Vasc Biol 2017; 37:1315-1322. [PMID: 28450294 DOI: 10.1161/atvbaha.116.308925] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 04/17/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Coagulation and inflammation are inter-related. Gas6 (growth arrest-specific 6) promotes venous thrombosis and participates to inflammation through endothelial-innate immune cell interactions. Innate immune cells can provide the initiating stimulus for venous thrombus development. We hypothesize that Gas6 promotes monocyte recruitment during venous thrombosis. APPROACH AND RESULTS Deep venous thrombosis was induced in wild-type and Gas6-deficient (-/-) mice using 5% FeCl3 and flow reduction in the inferior vena cava. Total monocyte depletion was achieved by injection of clodronate before deep venous thrombosis. Inflammatory monocytes were depleted using an anti-C-C chemokine receptor type 2 (CCR2) antibody. Similarly, injection of an anti-chemokine ligand 2 (CCL2) antibody induced CCL2 depletion. Flow cytometry and immunofluorescence were used to characterize the monocytes recruited to the thrombus. In vivo, absence of Gas6 was associated with a reduction of monocyte recruitment in both deep venous thrombosis models. Global monocyte depletion by clodronate leads to smaller thrombi in wild-type mice. Compared with wild type, the thrombi from Gas6-/- mice contain less inflammatory (CCR2hiCX3CR1lo) monocytes, consistent with a Gas6-dependent recruitment of this monocyte subset. Correspondingly, selective depletion of CCR2hiCX3CR1lo monocytes reduced the formation of venous thrombi in wild-type mice demonstrating a predominant role of the inflammatory monocytes in thrombosis. In vitro, the expression of both CCR2 and CCL2 were Gas6 dependent in monocytes and endothelial cells, respectively, impacting monocyte migration. Moreover, Gas6-dependent CCL2 expression and monocyte migration were mediated via JNK (c-Jun N-terminal kinase). CONCLUSIONS This study demonstrates that Gas6 specifically promotes the recruitment of inflammatory CCR2hiCX3CR1lo monocytes through the regulation of both CCR2 and CCL2 during deep venous thrombosis.
Collapse
MESH Headings
- Animals
- CX3C Chemokine Receptor 1
- Cells, Cultured
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Chemotaxis, Leukocyte/drug effects
- Clodronic Acid/pharmacology
- Disease Models, Animal
- Endothelial Cells/metabolism
- Genetic Predisposition to Disease
- Inflammation/genetics
- Inflammation/metabolism
- Inflammation/pathology
- Inflammation/prevention & control
- Intercellular Signaling Peptides and Proteins/deficiency
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/metabolism
- JNK Mitogen-Activated Protein Kinases/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Monocytes/drug effects
- Monocytes/metabolism
- Paracrine Communication
- Phenotype
- Receptors, CCR2/genetics
- Receptors, CCR2/metabolism
- Receptors, Chemokine/metabolism
- Signal Transduction
- Vena Cava, Inferior/drug effects
- Vena Cava, Inferior/metabolism
- Vena Cava, Inferior/pathology
- Venous Thrombosis/genetics
- Venous Thrombosis/metabolism
- Venous Thrombosis/pathology
- Venous Thrombosis/prevention & control
Collapse
Affiliation(s)
- Sandrine Laurance
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| | - François-René Bertin
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| | - Talin Ebrahimian
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| | - Yusra Kassim
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| | - Ryan N Rys
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| | - Stéphanie Lehoux
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| | - Catherine A Lemarié
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada.
| | - Mark D Blostein
- From the Lady Davis Institute for Medical Research (S.L., F.-R.B., T.E., Y.K., R.N.R., S.L., C.A.L., M.D.B.); and Department of Medicine (S.L., C.A.L., M.D.B.), Jewish General Hospital, McGill University, Montreal, Québec, Canada
| |
Collapse
|
25
|
RNA-Seq analysis of chikungunya virus infection and identification of granzyme A as a major promoter of arthritic inflammation. PLoS Pathog 2017; 13:e1006155. [PMID: 28207896 PMCID: PMC5312928 DOI: 10.1371/journal.ppat.1006155] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 12/28/2016] [Indexed: 02/07/2023] Open
Abstract
Chikungunya virus (CHIKV) is an arthritogenic alphavirus causing epidemics of acute and chronic arthritic disease. Herein we describe a comprehensive RNA-Seq analysis of feet and lymph nodes at peak viraemia (day 2 post infection), acute arthritis (day 7) and chronic disease (day 30) in the CHIKV adult wild-type mouse model. Genes previously shown to be up-regulated in CHIKV patients were also up-regulated in the mouse model. CHIKV sequence information was also obtained with up to ≈8% of the reads mapping to the viral genome; however, no adaptive viral genome changes were apparent. Although day 2, 7 and 30 represent distinct stages of infection and disease, there was a pronounced overlap in up-regulated host genes and pathways. Type I interferon response genes (IRGs) represented up to ≈50% of up-regulated genes, even after loss of type I interferon induction on days 7 and 30. Bioinformatic analyses suggested a number of interferon response factors were primarily responsible for maintaining type I IRG induction. A group of genes prominent in the RNA-Seq analysis and hitherto unexplored in viral arthropathies were granzymes A, B and K. Granzyme A-/- and to a lesser extent granzyme K-/-, but not granzyme B-/-, mice showed a pronounced reduction in foot swelling and arthritis, with analysis of granzyme A-/- mice showing no reductions in viral loads but reduced NK and T cell infiltrates post CHIKV infection. Treatment with Serpinb6b, a granzyme A inhibitor, also reduced arthritic inflammation in wild-type mice. In non-human primates circulating granzyme A levels were elevated after CHIKV infection, with the increase correlating with viral load. Elevated granzyme A levels were also seen in a small cohort of human CHIKV patients. Taken together these results suggest granzyme A is an important driver of arthritic inflammation and a potential target for therapy. Trial Registration: ClinicalTrials.gov NCT00281294 The largest chikungunya virus (CHIKV) epidemic ever recorded began in 2004 in Africa and spread across Asia reaching Europe and recently the Americas, with millions of cases reported. We undertook a detailed analysis of the mRNA expression profile during acute and chronic arthritis in an adult wild-type mouse model of CHIKV infection and disease. Gene induction profiles showed a high concordance with published human data, providing some validation of the mouse model. The host response was overwhelmingly dominated by type I interferon response genes, even after type I interferon induction was lost. The analysis also provided information on CHIKV RNA, with no adaptive viral genome changes identified. An important goal of the analysis was to identify new players in arthritic inflammation. Granzyme A was prominent in the RNA-Seq data and granzyme A deficient mice showed reduced arthritis, with no effects on viral loads. Arthritic disease could also be ameliorated in wild-type mice with a granzyme A inhibitor. Elevated circulating granzyme A levels were seen in non-human primates infected with CHIKV and in human CHIKV patients. Granzyme A thus emerges to be a major driver of CHIKV-mediated arthritic inflammation and a potential target for anti-inflammatory interventions.
Collapse
|
26
|
Karegli J, Melchionna T, Farrar CA, Greenlaw R, Smolarek D, Horsfield C, Charif R, McVey JH, Dorling A, Sacks SH, Smith RAG. Thrombalexins: Cell-Localized Inhibition of Thrombin and Its Effects in a Model of High-Risk Renal Transplantation. Am J Transplant 2017; 17:272-280. [PMID: 27376583 DOI: 10.1111/ajt.13951] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 06/13/2016] [Accepted: 06/28/2016] [Indexed: 01/25/2023]
Abstract
Allograft transplantation into sensitized recipients with antidonor antibodies results in accelerated antibody-mediated rejection (AMR), complement activation, and graft thrombosis. We have developed a membrane-localizing technology of wide applicability that enables therapeutic agents, including anticoagulants, to bind to cell surfaces and protect the donor endothelium. We describe here how this technology has been applied to thrombin inhibitors to generate a novel class of drugs termed thrombalexins (TLNs). Using a rat model of hyperacute rejection, we investigated the potential of one such inhibitor (thrombalexin-1 [TLN-1]) to prevent acute antibody-mediated thrombosis in the donor organ. TLN-1 alone was able to reduce intragraft thrombosis and significantly delay rejection. The results confirm a pivotal role for thrombin in AMR in vivo. This approach targets donor organs rather than the recipient and is intended to be directly translatable to clinical use.
Collapse
Affiliation(s)
- J Karegli
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - T Melchionna
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - C A Farrar
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - R Greenlaw
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - D Smolarek
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - C Horsfield
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - R Charif
- West London Renal and Transplantation Centre, Imperial College Healthcare NHS Trust, Hammersmith Hospital, London, UK
| | - J H McVey
- School of Bioscience & Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - A Dorling
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - S H Sacks
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - R A G Smith
- MRC Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| |
Collapse
|
27
|
Ebrahimi S, Jaberi N, Avan A, Ryzhikov M, Keramati MR, Parizadeh MR, Hassanian SM. Role of thrombin in the pathogenesis of central nervous system inflammatory diseases. J Cell Physiol 2016; 232:482-485. [PMID: 27458694 DOI: 10.1002/jcp.25501] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 07/25/2016] [Indexed: 12/13/2022]
Abstract
Thrombin initiates proinflammatory signaling responses through activation of protease-activated receptors (PARs) in in vitro and in vivo systems. Proinflammatory signaling function of thrombin increases secretion of proinflammatory cytokines and chemokines, triggers vascular permeability, promotes leukocyte migration, and induces adhesion molecule expression. Thrombin as a potent signaling molecule is strongly implicated in a number of proinflammatory disorders including severe sepsis, cancer, cardiovascular disease, and of special interest in this review neurodegenerative disorders. This review summarizes the role of thrombin in the pathogenesis of central nervous system (CNS) inflammatory diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD), promoting greater understanding and clinical management of these diseases. J. Cell. Physiol. 232: 482-485, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Safieh Ebrahimi
- Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Najme Jaberi
- Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Molecular Medicine Group, Department of Modern Sciences and Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Cancer Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mikhail Ryzhikov
- Department of Biochemistry and Molecular Biology, St. Louis University School of Medicine, Saint Louis, Missouri
| | - Mohammad Reza Keramati
- Cancer Molecular Pathology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Parizadeh
- Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Biochemistry of Nutrition Research Center, 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.,Department of Biochemistry and Molecular Biology, St. Louis University School of Medicine, Saint Louis, Missouri.,Microanatomy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
28
|
Long R, You Y, Li W, Jin N, Huang S, Li T, Liu K, Wang Z. Sodium tanshinone IIA sulfonate ameliorates experimental coronary no-reflow phenomenon through down-regulation of FGL2. Life Sci 2015; 142:8-18. [PMID: 26482204 DOI: 10.1016/j.lfs.2015.10.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 10/03/2015] [Accepted: 10/15/2015] [Indexed: 02/06/2023]
Abstract
AIMS The effects of sodium tanshinone IIA sulfonate (STS) on coronary no-reflow (CNR) relevant to microvascular obstruction (MVO) remain unknown. Studies had shown that fibrinogen-like protein 2 (FGL2) expressed in microvascular endothelial cells (MECs) is a key mediator in MVO. Thus, we aimed to elucidate the roles of STS in CNR and relations between STS and FGL2. MAIN METHODS Myocardial ischemia/reperfusion was selected to represent CNR model. The no-reflow zone and infarct area were assessed using Thioflavin S and TTC staining, and cardiac functional parameters were detected using echocardiography. Western blot was used to detected FGL2 level, fibrin level, protease-activated receptor-1 (PAR-1) activation and inflammation cells infiltration. FGL2 and inflammation cells were also identified by IHC. Microthrombus was detected by Carstairs' and MSB staining. We also detected the roles of STS on FGL2 expression, thrombin generation, phospho-Akt and NF-κB levels in MECs. KEY FINDINGS Upon treatment with STS in CNR model, the no-reflow and infarct areas decreased significantly and cardiac function improved. The FGL2 expression was inhibited by STS in vivo as well as in vitro with thrombin generation inhibition. In addition, STS up-regulates Akt phosphorylation and suppressed NF-κB expression in activated MECs. Furthermore, fibrin deposition, PAR-1 activation and inflammatory response were inhibited with STS administration in CNR model. SIGNIFICANCE Our results displayed a novel pharmacological action of STS on CNR. STS is able to ameliorate CNR through inhibition of FGL2 expression mediated by Akt and NF-κB pathways as well as prevention of MVO by suppressing fibrin deposition and inflammation.
Collapse
Affiliation(s)
- Rui Long
- Department of Geriatrics, Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya You
- Department of Geriatrics, Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenzhu Li
- Department of Geriatrics, Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nan Jin
- Department of Geriatrics, Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiyuan Huang
- Department of Geriatrics, Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Li
- Department of Geriatrics, Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Liu
- Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Zhaohui Wang
- Department of Geriatrics, Institute of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
29
|
Kaplan ZS, Zarpellon A, Alwis I, Yuan Y, McFadyen J, Ghasemzadeh M, Schoenwaelder SM, Ruggeri ZM, Jackson SP. Thrombin-dependent intravascular leukocyte trafficking regulated by fibrin and the platelet receptors GPIb and PAR4. Nat Commun 2015. [PMID: 26204458 DOI: 10.1038/ncomms8835] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Thrombin is a central regulator of leukocyte recruitment and inflammation at sites of vascular injury, a function thought to involve primarily endothelial PAR cleavage. Here we demonstrate the existence of a distinct leukocyte-trafficking mechanism regulated by components of the haemostatic system, including platelet PAR4, GPIbα and fibrin. Utilizing a mouse endothelial injury model we show that thrombin cleavage of platelet PAR4 promotes leukocyte recruitment to sites of vascular injury. This process is negatively regulated by GPIbα, as seen in mice with abrogated thrombin-platelet GPIbα binding (hGPIbα(D277N)). In addition, we demonstrate that fibrin limits leukocyte trafficking by forming a physical barrier to intravascular leukocyte migration. These studies demonstrate a distinct 'checkpoint' mechanism of leukocyte trafficking involving balanced thrombin interactions with PAR4, GPIbα and fibrin. Dysregulation of this checkpoint mechanism is likely to contribute to the development of thromboinflammatory disorders.
Collapse
Affiliation(s)
- Zane S Kaplan
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria 3004, Australia
| | - Alessandro Zarpellon
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Imala Alwis
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria 3004, Australia.,Heart Research Institute &Charles Perkins Centre, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Yuping Yuan
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria 3004, Australia
| | - James McFadyen
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria 3004, Australia
| | - Mehran Ghasemzadeh
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria 3004, Australia.,Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Simone M Schoenwaelder
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria 3004, Australia.,Heart Research Institute &Charles Perkins Centre, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Zaverio M Ruggeri
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Shaun P Jackson
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria 3004, Australia.,Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California 92037, USA.,Heart Research Institute &Charles Perkins Centre, The University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
30
|
Kinetics of lung tissue factor expression and procoagulant activity in bleomycin induced acute lung injury. Clin Transl Med 2015; 4:63. [PMID: 26154059 PMCID: PMC4495096 DOI: 10.1186/s40169-015-0063-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/31/2015] [Indexed: 01/11/2023] Open
Abstract
Background Activation of coagulation by expression of tissue factor (TF) in the airspace is a hallmark of acute lung injury (ALI) but the timing of TF activation in relationship to increases in lung permeability and inflammation are unknown. Methods To test the hypothesis that TF is upregulated early in the course of acute bleomycin lung injury and precedes increased permeability and inflammation we studied the early course of bleomycin-induced ALI in mice. Mice were treated with 0.04U intratracheal bleomycin or vehicle control and bronchoalveolar lavage (BAL) and lung tissue were collected daily for 7 days. Whole lung TF mRNA was determined by QT-PCR. TF protein was assessed by ELISA and immunostaining. BAL procoagulant activity was measured by BAL clot time and thrombin-antithrombin complexes. Inflammation was assessed by BAL cell count, differentials and CXCL1/KC concentration. Lung permeability was assessed by BAL protein and lung wet to dry weight ratio. Results Expression of CXCL1 occurred by day 1. BAL protein and lung wet-to-dry weight ratio increased significantly by day 3. TF mRNA and BAL procoagulant activity peaked on day 4 while whole lung TF protein peaked on day 6. Changes in permeability and procoagulant activity preceded inflammatory cell influx which was maximal at day 6 while whole lung TF protein peaked along with inflammation. Conclusion These data demonstrate that cytokine upregulation is the earliest response to bleomycin administration, followed by increased lung permeability, upregulation of TF, and recruitment of inflammatory cells.
Collapse
|
31
|
Sartim MA, Costa TR, Laure HJ, Espíndola MS, Frantz FG, Sorgi CA, Cintra ACO, Arantes EC, Faccioli LH, Rosa JC, Sampaio SV. Moojenactivase, a novel pro-coagulant PIIId metalloprotease isolated from Bothrops moojeni snake venom, activates coagulation factors II and X and induces tissue factor up-regulation in leukocytes. Arch Toxicol 2015; 90:1261-78. [PMID: 26026608 DOI: 10.1007/s00204-015-1533-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 05/12/2015] [Indexed: 11/28/2022]
Abstract
Coagulopathies following snakebite are triggered by pro-coagulant venom toxins, in which metalloproteases play a major role in envenomation-induced coagulation disorders by acting on coagulation cascade, platelet function and fibrinolysis. Considering this relevance, here we describe the isolation and biochemical characterization of moojenactivase (MooA), a metalloprotease from Bothrops moojeni snake venom, and investigate its involvement in hemostasis in vitro. MooA is a glycoprotein of 85,746.22 Da, member of the PIIId group of snake venom metalloproteases, composed of three linked disulfide-bonded chains: an N-glycosylated heavy chain, and two light chains. The venom protease induced human plasma clotting in vitro by activating on both blood coagulation factors II (prothrombin) and X, which in turn generated α-thrombin and factor Xa, respectively. Additionally, MooA induced expression of tissue factor (TF) on the membrane surface of peripheral blood mononuclear cells (PBMC), which led these cells to adopt pro-coagulant characteristics. MooA was also shown to be involved with production of the inflammatory mediators TNF-α, IL-8 and MCP-1, suggesting an association between MooA pro-inflammatory stimulation of PBMC and TF up-regulation. We also observed aggregation of washed platelets when in presence of MooA; however, the protease had no effect on fibrinolysis. Our findings show that MooA is a novel hemostatically active metalloprotease, which may lead to the development of coagulopathies during B. moojeni envenomation. Moreover, the metalloprotease may contribute to the development of new diagnostic tools and pharmacological approaches applied to hemostatic disorders.
Collapse
Affiliation(s)
- Marco A Sartim
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14040-903, Brazil
| | - Tassia R Costa
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14040-903, Brazil
| | - Helen J Laure
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos e Centro de Química de Proteínas, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14040-903, Brazil
| | - Milena S Espíndola
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14040-903, Brazil
| | - Fabiani G Frantz
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14040-903, Brazil
| | - Carlos A Sorgi
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14040-903, Brazil
| | - Adélia C O Cintra
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14040-903, Brazil
| | - Eliane C Arantes
- Departamento de Física e Química, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14040-903, Brazil
| | - Lucia H Faccioli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14040-903, Brazil
| | - José C Rosa
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos e Centro de Química de Proteínas, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14040-903, Brazil
| | - Suely V Sampaio
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14040-903, Brazil.
| |
Collapse
|
32
|
Thrombin Receptor Protease-Activated Receptor 4 Is a Key Regulator of Exaggerated Intimal Thickening in Diabetes Mellitus. Circulation 2014; 130:1700-11. [DOI: 10.1161/circulationaha.113.007590] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background—
Diabetes mellitus predisposes to thrombotic and proliferative vascular remodeling, to which thrombin contributes via activation of protease-activated receptor (PAR) 1. However, the use of PAR-1 inhibitors to suppress remodeling may be limited by severe bleeding. We recently reported upregulation of an additional thrombin receptor, PAR-4, in human vascular smooth muscle cells exposed to high glucose and have now examined PAR-4 as a novel mediator linking hyperglycemia, hypercoagulation, and vascular remodeling in diabetes mellitus.
Methods and Results—
PAR-4 expression was increased in carotid atherectomies and saphenous vein specimens from diabetic versus nondiabetic patients and in aorta and carotid arteries from streptozotocin-diabetic versus nondiabetic C57BL/6 mice. Vascular PAR-1 mRNA was not increased in diabetic mice. Ligated carotid arteries from diabetic mice developed more extensive neointimal hyperplasia and showed greater proliferation than arteries from nondiabetic mice. The augmented remodeling response was absent in diabetic mice deficient in PAR-4. At the cellular level, PAR-4 expression was controlled via the mRNA stabilizing actions of human antigen R, which accounted for the stimulatory actions of high glucose, angiotensin II, and H
2
O
2
on PAR-4 expression, whereas cicaprost via protein kinase A activation counteracted this effect.
Conclusions—
PAR-4 appears to play a hitherto unsuspected role in diabetic vasculopathy. The development of PAR-4 inhibitors might serve to limit mainly proliferative processes in restenosis-prone diabetic patients, particularly those patients in whom severe bleeding attributed to selective PAR-1 blockade or complete thrombin inhibition must be avoided or those who do not require anticoagulation.
Collapse
|
33
|
Carmo AAF, Costa BRC, Vago JP, de Oliveira LC, Tavares LP, Nogueira CRC, Ribeiro ALC, Garcia CC, Barbosa AS, Brasil BSAF, Dusse LM, Barcelos LS, Bonjardim CA, Teixeira MM, Sousa LP. Plasmin induces in vivo monocyte recruitment through protease-activated receptor-1-, MEK/ERK-, and CCR2-mediated signaling. THE JOURNAL OF IMMUNOLOGY 2014; 193:3654-63. [PMID: 25165151 DOI: 10.4049/jimmunol.1400334] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The plasminogen (Plg)/plasmin (Pla) system is associated with a variety of biological activities beyond the classical dissolution of fibrin clots, including cell migration, tissue repair, and inflammation. Although the capacity of Plg/Pla to induce cell migration is well defined, the mechanism underlying this process in vivo is elusive. In this study, we show that Pla induces in vitro migration of murine fibroblasts and macrophages (RAW 264.7) dependent on the MEK/ERK pathway and by requiring its proteolytic activity and lysine binding sites. Plasmin injection into the pleural cavity of BALB/c mice induced a time-dependent influx of mononuclear cells that was associated with augmented ERK1/2 and IκB-α phosphorylation and increased levels of CCL2 and IL-6 in pleural exudates. The inhibition of protease activity by using a serine protease inhibitor leupeptin or two structurally different protease-activated receptor-1 antagonists (SCH79797 and RWJ56110) abolished Pla-induced mononuclear recruitment and ERK1/2 and IκB-α phosphorylation. Interestingly, inhibition of the MEK/ERK pathway abolished Pla-induced CCL2 upregulation and mononuclear cell influx. In agreement with a requirement for the CCL2/CCR2 axis to Pla-induced cell migration, the use of a CCR2 antagonist (RS504393) prevented the Plg/Pla-induced recruitment of mononuclear cells to the pleural cavity and migration of macrophages at transwell plates. Therefore, Pla-induced mononuclear cell recruitment in vivo was dependent on protease-activated receptor-1 activation of the MEK/ERK/NF-κB pathway, which led to the release of CCL2 and activation of CCR2.
Collapse
Affiliation(s)
- Aline A F Carmo
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Programa de Pós-Graduação em Biologia Celular, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Bruno R C Costa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Juliana P Vago
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Programa de Pós-Graduação em Biologia Celular, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Leonardo C de Oliveira
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; and
| | - Luciana P Tavares
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Camila R C Nogueira
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Ana Luíza C Ribeiro
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Cristiana C Garcia
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Alan S Barbosa
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | | | - Luci M Dusse
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Lucíola S Barcelos
- Programa de Pós-Graduação em Biologia Celular, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Cláudio A Bonjardim
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; and
| | - Mauro M Teixeira
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil;
| | - Lirlândia P Sousa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Programa de Pós-Graduação em Biologia Celular, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil; Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil;
| |
Collapse
|
34
|
Gomides LF, Lima OCO, Matos NA, Freitas KM, Francischi JN, Tavares JC, Klein A. Blockade of proteinase-activated receptor 4 inhibits neutrophil recruitment in experimental inflammation in mice. Inflamm Res 2014; 63:935-41. [PMID: 25118784 DOI: 10.1007/s00011-014-0767-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 07/14/2014] [Accepted: 08/05/2014] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE AND DESIGN The activation of proteinase-activated receptors (PARs) has been implicated in the development of important hallmarks of inflammation, including in vivo leukocyte recruitment; however, its role in the regulation of leukocyte migration in response to inflammatory stimuli has not been elucidated until now. Here, we examined the effects of the PAR4 antagonist YPGKF-NH 2 (tcY-NH2) on neutrophil recruitment in experimentally induced inflammation. METHODS BALB/c mice were intrapleurally injected with tcY-NH2 (40 ng/kg) prior to intrapleural injection of carrageenan (Cg) or neutrophil chemoattractant CXCL8; the number of infiltrating neutrophils was evaluated after 4 h, and KC production was assessed at different times after Cg injection. Neutrophil adhesion and rolling cells were studied using a brain circulation preparation 4 h after the Cg or CXCL8 challenge in tcY-NH2-treated mice. RESULTS PAR4 blockade inhibited CXCL8- and Cg-induced neutrophil migration into the pleural cavity of BALB/c mice and reduced neutrophil rolling and adherence. Surprisingly, PAR4 blockade increased the level of KC in response to carrageenan. CONCLUSION These results demonstrated that PAR4 blockade impairs neutrophil migration in vivo, suggesting that PAR4 plays an important role in the regulation of inflammation, at least in part because of its ability to inhibit the actions of the neutrophil chemoattractant CXCL8.
Collapse
Affiliation(s)
- Lindisley F Gomides
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627 Pampulha, 31270-901, Belo Horizonte, MG, Brazil
| | | | | | | | | | | | | |
Collapse
|
35
|
Matos NA, Silva JF, Damasceno KA, Cassali GD, Lemos VS, Duarte IDG, Klein A. Proteinase-activated receptor 2 blockade impairs CCL11- or allergen-induced eosinophil recruitment in experimental pleurisy. Eur J Pharmacol 2014; 740:627-33. [PMID: 24972241 DOI: 10.1016/j.ejphar.2014.06.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 06/12/2014] [Accepted: 06/17/2014] [Indexed: 01/05/2023]
Abstract
Although proteinase-activated receptor (PAR)-2 has been implicated in inflammatory diseases, its role in regulating eosinophil recruitment in response to chemoattractants remains unclear. Here, we investigated the role of PAR-2 and PAR-2-activating Mast Cell (MC) tryptase on chemokine C-C motif ligand (CCL)11- and antigen-induced eosinophil recruitment to the pleural cavity of BALB/c mice. The PAR-2-activating peptide H-Ser-Leu-Ile-Gly-Arg-Leu-NH2 (SLIGRL-NH2) induced eosinophil recruitment whereas PAR-2 blockade inhibited ovalbumin (OVA)- or CCL11-induced eosinophil recruitment. Moreover, OVA and CCL11 induced PAR-2 expression in pleural leukocytes, and the MC tryptase inhibitor APC 366 ([N-(1-hydroxy-2-napthoyl)-l-arginyl-l-prolinamide hydrochloride]) abolished CCL11-induced eosinophil recruitment. These results suggest a pro inflammatory effect of PAR-2 and support a role for MC tryptase mediating eosinophil migration via PAR-2 signaling. Taken together, our results suggest that PAR-2 activation through endogenous MC tryptase activity could be required, at least partially, to mediate CCL11-induced eosinophil migration.
Collapse
Affiliation(s)
- Natália A Matos
- Department of Pharmacology, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627 Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| | - Josiane F Silva
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Karine A Damasceno
- Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Geovanni D Cassali
- Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Virginia S Lemos
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Igor D G Duarte
- Department of Pharmacology, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627 Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| | - André Klein
- Department of Pharmacology, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, 6627 Pampulha, 31270-901 Belo Horizonte, MG, Brazil.
| |
Collapse
|
36
|
Kallis YN, Scotton CJ, MacKinnon AC, Goldin RD, Wright NA, Iredale JP, Chambers RC, Forbes SJ. Proteinase activated receptor 1 mediated fibrosis in a mouse model of liver injury: a role for bone marrow derived macrophages. PLoS One 2014; 9:e86241. [PMID: 24475094 PMCID: PMC3903514 DOI: 10.1371/journal.pone.0086241] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Accepted: 12/10/2013] [Indexed: 01/02/2023] Open
Abstract
Liver fibrosis results from the co-ordinated actions of myofibroblasts and macrophages, a proportion of which are of bone marrow origin. The functional effect of such bone marrow-derived cells on liver fibrosis is unclear. We examine whether changing bone marrow genotype can down-regulate the liver's fibrotic response to injury and investigate mechanisms involved. Proteinase activated receptor 1 (PAR1) is up-regulated in fibrotic liver disease in humans, and deficiency of PAR1 is associated with reduced liver fibrosis in rodent models. In this study, recipient mice received bone marrow transplantation from PAR1-deficient or wild-type donors prior to carbon tetrachloride-induced liver fibrosis. Bone marrow transplantation alone from PAR1-deficient mice was able to confer significant reductions in hepatic collagen content and activated myofibroblast expansion on wild-type recipients. This effect was associated with a decrease in hepatic scar-associated macrophages and a reduction in macrophage recruitment from the bone marrow. In vitro, PAR1 signalling on bone marrow-derived macrophages directly induced their chemotaxis but did not stimulate proliferation. These data suggest that the bone marrow can modulate the fibrotic response of the liver to recurrent injury. PAR1 signalling can contribute to this response by mechanisms that include the regulation of macrophage recruitment.
Collapse
Affiliation(s)
- Yiannis N. Kallis
- Department of Hepatology, St. Mary's Hospital Campus, Imperial College London, London, United Kingdom
| | - Christopher J. Scotton
- Centre for Inflammation & Tissue Repair, University College London, London, United Kingdom
| | - Alison C. MacKinnon
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Robert D. Goldin
- Department of Histopathology, St. Mary's Hospital Campus, Imperial College London, London, United Kingdom
| | - Nicholas A. Wright
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, London, United Kingdom
| | - John P. Iredale
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Regenerative Medicine, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Rachel C. Chambers
- Centre for Inflammation & Tissue Repair, University College London, London, United Kingdom
| | - Stuart J. Forbes
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Regenerative Medicine, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
37
|
Costa L, Roth M, Miglino N, Keglowich L, Zhong J, Lardinois D, Tamm M, Borger P. Tiotropium sustains the anti-inflammatory action of olodaterol via the cyclic AMP pathway. Pulm Pharmacol Ther 2013; 27:29-37. [PMID: 24269928 DOI: 10.1016/j.pupt.2013.11.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 11/07/2013] [Accepted: 11/10/2013] [Indexed: 11/29/2022]
Abstract
Mesenchymal cells (fibroblasts) of the airway wall respond to cholinergic stimulation by releasing pro-inflammatory and chemotactic cytokines and may thus contribute to chronic inflammation of the lung. Here, we studied the anti-inflammatory potential of olodaterol, a long acting β2-adrenergic receptor agonist, and tiotropium, a long-acting muscarinic receptor antagonist, and whether they interact at the level of the cyclic AMP dependent signaling pathway. Pulmonary fibroblasts of asthmatic (n = 9) and non-asthmatic (n = 8) subjects were stimulated with the muscarinic receptor agonist carbachol and interleukin-1β (IL-1 beta) in presence or absence of tiotropium or olodaterol alone, or their combination. We also measured cAMP levels and phosphorylation of the cAMP response element binding protein (CREB). As single components, carbachol, olodaterol and tiotropium did not affect IL-6 and IL-8 release. Carbachol concentration-dependently enhanced the production of IL-1β-induced IL-6 and IL-8, which was blocked by the simultaneous addition of tiotropium. The combination of olodaterol plus tiotropium further reduced IL-6 and IL-8 release. Olodaterol induced cAMP and the phosphorylation of CREB, an effect counteracted by carbachol, but rescued by tiotropium. We conclude that olodaterol plus tiotropium cooperate to decrease the inflammatory response in pulmonary fibroblasts in vitro.
Collapse
Affiliation(s)
- Luigi Costa
- Pulmonary Cell Research, Department of Biomedicine, University Hospital Basel, Switzerland
| | - Michael Roth
- Pulmonology, Department of Internal medicine, University Hospital Basel, Switzerland
| | - Nicola Miglino
- Pulmonary Cell Research, Department of Biomedicine, University Hospital Basel, Switzerland
| | - Laura Keglowich
- Pulmonary Cell Research, Department of Biomedicine, University Hospital Basel, Switzerland
| | - Jun Zhong
- Pulmonary Cell Research, Department of Biomedicine, University Hospital Basel, Switzerland
| | - Didier Lardinois
- Department of Thoracic Surgery, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Michael Tamm
- Pulmonology, Department of Internal medicine, University Hospital Basel, Switzerland
| | - Pieter Borger
- Pulmonary Cell Research, Department of Biomedicine, University Hospital Basel, Switzerland.
| |
Collapse
|
38
|
Enhanced effect of inhibition of thrombin on endothelium in murine endotoxaemia: specific inhibition of thrombocytopenia. Thromb Res 2013; 132:750-6. [PMID: 24161661 DOI: 10.1016/j.thromres.2013.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 07/07/2013] [Accepted: 10/01/2013] [Indexed: 11/21/2022]
Abstract
INTRODUCTION In systemic endotoxaemia, bacterial lipopolysaccharide causes the rapid expression of tissue factor (TF) and disseminated intravascular coagulation and in animal models, anticoagulants limit pathology and promote survival. Recent studies have emphasised the importance of TF expressed by mononuclear cells for initiating thrombin generation during endotoxaemia and suggested that endothelial cell TF is of little relevance. However, the precise importance of endothelium for intravascular thrombin generation has not been established. In this study, we compared the effect of equivalent levels of hirudin tethered to either endothelium or platelets and monocytes. MATERIALS AND METHODS CD31-Hir-Tg mice express a vesicle-targeted, membrane-tethered hirudin fusion protein on endothelium, platelets and monocytes. Bone marrow chimeras between these mice and C57BL/6 were generated The level of intravascular hirudin expressed during endotoxaemia was quantified by inhibition studies using an anti-hirudin antibody and reference to the circulating thrombin anti-thrombin complexes generated in control mice given soluble hirudin. RESULTS AND CONCLUSIONS Antibody inhibition studies indicated that individual chimeras expressed similar levels of hirudin fusion protein on endothelium alone as on platelets and leukocytes combined and accordingly, the levels of thrombin anti-thrombin complexes and fibrinogen in each chimera were similar, indicating equivalent inhibition of thrombin generation. However, mice with hirudin on endothelium alone developed significantly less thrombocytopenia. These results suggest a hitherto unrecognized role of endothelium in thrombin-dependent platelet sequestration during endotoxaemia. The data have implications for the development of therapeutic strategies based on targeted anticoagulation to limit disseminated intravascular coagulation.
Collapse
|
39
|
Yang L, Zhou X, Guo R, Shi Y, Liang X, Heng X. Role of Krüppel-Like Factor 2 and Protease-Activated Receptor-1 in Vulnerable Plaques of ApoE−/− Mice and Intervention With Statin. Can J Cardiol 2013; 29:997-1005. [DOI: 10.1016/j.cjca.2012.11.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 11/02/2012] [Accepted: 11/03/2012] [Indexed: 01/06/2023] Open
|
40
|
Pflücke D, Hackel D, Mousa SA, Partheil A, Neumann A, Brack A, Rittner HL. The molecular link between C-C-chemokine ligand 2-induced leukocyte recruitment and hyperalgesia. THE JOURNAL OF PAIN 2013; 14:897-910. [PMID: 23683582 DOI: 10.1016/j.jpain.2013.02.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 02/17/2013] [Accepted: 02/26/2013] [Indexed: 12/27/2022]
Abstract
UNLABELLED The chemokine C-C-chemokine ligand 2 (CCL2) (formerly known as MCP, macrophage chemotactic protein) is one of the important genes upregulated in different types of pain both in animals and humans. CCL2 governs the recruitment of C-C chemokine receptor 2-expressing monocytes into inflamed tissue. In contrast to neutrophilic chemokines, intraplantar injection of CCL2 in Wistar rats recruited macrophages and neutrophils and simultaneously lowered nociceptive thresholds. CCL2-induced hyperalgesia was abolished by prior systemic leukocyte depletion by cyclophosphamide and was reconstituted by local adoptive transfer of donor macrophages but not of neutrophils. Antagonists against transient receptor potential vannilloid 1 inhibited thermal and against transient receptor potential ankyrin 1 blocked mechanical hyperalgesia. Peripheral but not central activation of cyclooxygenase-2 (Cox-2) were critical for CCL2-induced hyperalgesia. In vitro CCL2 did not directly stimulate Cox-2 expression or prostaglandin E2 formation but slightly enhanced the formation of reactive oxygen species in monocytes and macrophages. In vivo, increased immunoreactivity for 4-hydroxy-2-nonenal (4-HNE), a downstream product of reactive oxygen species and known inducer of Cox-2, was observed and colocalized with Cox-2 in ED1 (CD68) positive infiltrating cells. No hyperalgesia, 4-HNE, or Cox-2 immunoreactivity was seen in leukocyte-depleted rats that were reconstituted with macrophages in the absence of CCL2, supporting the important role of CCL2. PERSPECTIVE CCL2 plays a dual role: 1) promoting monocyte/macrophage recruitment into tissue; and 2) potentially stimulating macrophages in the tissue to produce 4-HNE and subsequently Cox-2, all resulting in the induction of hyperalgesia via transient receptor potential vannilloid 1 and transient receptor potential ankyrin 1. This encourages pharmacological efforts targeting CCL2/C-C chemokine receptor 2 and macrophages for treatment of inflammatory pain.
Collapse
Affiliation(s)
- Diana Pflücke
- Department of Anesthesiology, University Hospital of Würzburg, Würzburg, Germany
| | | | | | | | | | | | | |
Collapse
|
41
|
Chen D, Ma L, Tham EL, Maresh S, Lechler RI, McVey JH, Dorling A. Fibrocytes mediate intimal hyperplasia post-vascular injury and are regulated by two tissue factor-dependent mechanisms. J Thromb Haemost 2013; 11:963-74. [PMID: 23516969 DOI: 10.1111/jth.12198] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Accepted: 03/11/2013] [Indexed: 01/22/2023]
Abstract
BACKGROUND CD34(+) α-smooth muscle actin (SMA)(+) cells mediate intimal hyperplasia (IH) after mechanical endoluminal injury. We previously found that IH is tissue factor (TF) dependent. The precise phenotype of the CD34(+) cells mediating IH is unknown and the mechanisms of TF are also unknown. OBJECTIVE To define the phenotype of cells mediating IH and compare the effects of inhibiting TF on different subsets of CD34(+) cells. METHODS Endoluminal injury was induced in C57BL/6 and two strains of mice expressing a human tissue factor pathway inhibitor (hTFPI) fusion protein on different subsets of CD34(+) cells. Confocal microscopy, immunocytofluorescence and real-time PCR were used to determine phenotype. RESULTS Neointimal cells in C57BL/6 mice were defined as a subset of fibrocytes (CD34(+) CD45(+) collagen-1(+) ) expressing SMA, CD31, TIE-2, CXCR4 and CXCL12. Similar cells circulated post-injury and were also found in mice expressing hTFPI on CD34(+) CD31(+) cells, though in these mice, hTFPI inhibited CD31(+) fibrocyte hyperplasia, so no IH developed. Mice with hTFPI on all CD34(+) α-SMA(+) cells repaired arteries back to a pre-injured state. No CD31(+) fibrocytes were found in these mice unless an anti-hTFPI antibody was administered. Similar findings in protease activated receptor (PAR)-1-deficient mice suggested hTFPI prevented thrombin signaling through PAR-1. In vitro, thrombin increased the number of CD31(+) fibrocytes. CONCLUSIONS Inhibition of TF on CD31(+) fibrocytes inhibits IH whereas inhibition on all CD34(+) α-SMA(+) cells (or PAR-1 deficiency) inhibits the appearance of CD31(+) fibrocytes and promotes repair. These data enhance our understanding of IH and suggest novel ways to promote regenerative repair.
Collapse
Affiliation(s)
- D Chen
- Medical Research Council (MRC) Centre for Transplantation, King's College London, King's Health Partners, Guy's Hospital, London, UK
| | | | | | | | | | | | | |
Collapse
|
42
|
Asehnoune K, Moine P. Protease-activated receptor-1: key player in the sepsis coagulation-inflammation crosstalk. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2013; 17:119. [PMID: 23448515 PMCID: PMC4057503 DOI: 10.1186/cc12502] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Protease-activated receptors (PARs) belong to the family of G protein-coupled receptors. Among the four members, PAR1 plays a major role in orchestrating the interactions between coagulation and inflammation. PAR1 has opposing functions during sepsis, and PAR1 blockade or activation may be alternatively beneficial at early or late stages of different sepsis models. Studying molecular mechanisms of the crosstalk between inflammation and coagulation may lead to the identification of new targets for therapies in sepsis. However, the time-dependent switch of PAR1 from an exacerbating proinflammatory receptor to a protective anti-inflammatory receptor needs to be investigated before clinical trials can be recommended. Finally, as PAR1 seems to play a singular role in Streptococcus pneumoniae-induced sepsis through a crosstalk between PAR1 and platelet-activating factor receptor, the exact role of PAR1 needs to be investigated in other models of sepsis.
Collapse
|
43
|
Antoniak S, Owens AP, Baunacke M, Williams JC, Lee RD, Weithäuser A, Sheridan PA, Malz R, Luyendyk JP, Esserman DA, Trejo J, Kirchhofer D, Blaxall BC, Pawlinski R, Beck MA, Rauch U, Mackman N. PAR-1 contributes to the innate immune response during viral infection. J Clin Invest 2013; 123:1310-22. [PMID: 23391721 DOI: 10.1172/jci66125] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 12/10/2012] [Indexed: 01/25/2023] Open
Abstract
Coagulation is a host defense system that limits the spread of pathogens. Coagulation proteases, such as thrombin, also activate cells by cleaving PARs. In this study, we analyzed the role of PAR-1 in coxsackievirus B3-induced (CVB3-induced) myocarditis and influenza A infection. CVB3-infected Par1(-/-) mice expressed reduced levels of IFN-β and CXCL10 during the early phase of infection compared with Par1(+/+) mice that resulted in higher viral loads and cardiac injury at day 8 after infection. Inhibition of either tissue factor or thrombin in WT mice also significantly increased CVB3 levels in the heart and cardiac injury compared with controls. BM transplantation experiments demonstrated that PAR-1 in nonhematopoietic cells protected mice from CVB3 infection. Transgenic mice overexpressing PAR-1 in cardiomyocytes had reduced CVB3-induced myocarditis. We found that cooperative signaling between PAR-1 and TLR3 in mouse cardiac fibroblasts enhanced activation of p38 and induction of IFN-β and CXCL10 expression. Par1(-/-) mice also had decreased CXCL10 expression and increased viral levels in the lung after influenza A infection compared with Par1(+/+) mice. Our results indicate that the tissue factor/thrombin/PAR-1 pathway enhances IFN-β expression and contributes to the innate immune response during single-stranded RNA viral infection.
Collapse
Affiliation(s)
- Silvio Antoniak
- Department of Medicine, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Expression, function and cooperating partners of protease-activated receptor type 3 in vascular endothelial cells and B lymphocytes studied with specific monoclonal antibody. Mol Immunol 2013; 54:319-26. [PMID: 23352962 DOI: 10.1016/j.molimm.2012.12.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 12/26/2012] [Indexed: 01/01/2023]
Abstract
Receptor-specific antibodies can both prevent ligand-receptor interaction and initiate receptor signaling. Previously we generated monoclonal antibody 8E8 (mAb 8E8) against protease-activated receptor type 3 (PAR3) which inhibited proliferation of B cell hybridoma. Here we used mAb 8E8 and PAR1-specific polyclonal antibody to reveal the functions and cooperating partners of PAR3 in endothelial cells and in B lymphocytes. MAb 8E8 or PAR1 agonist peptide stimulated IL-6 and IL-8 production and VCAM-1 expression in HPMEC-ST1.6R cells. PAR1 antibody stimulated only VCAM-1 expression, while ICAM-1 expression was stimulated with mAB 8E8 or PAR3 peptide. MAb 8E8 stimulated weak mitogenic response, while PAR1 antibody inhibited it in normal but not in malignant B lymphocytes. Sandwich ELISA assay demonstrated the interaction of PAR3 with PAR1 in malignant cell lines and with IgM in normal B lymphocytes. It is concluded that PAR3 cooperates with PAR1 to mediate the effect of thrombin on cytokine production and VCAM-1 expression in endothelial cells and on cell proliferation in malignant B cells. ICAM-1 expression in endothelial cells requires PAR3 without PAR1. The inhibitory effect of thrombin in normal B lymphocytes is mediated by PAR1 alone, while mitogenic and pro-survival signaling in B lymphocytes is provided through PAR3 in cooperation with BCR.
Collapse
|
45
|
Khoufache K, Berri F, Nacken W, Vogel AB, Delenne M, Camerer E, Coughlin SR, Carmeliet P, Lina B, Rimmelzwaan GF, Planz O, Ludwig S, Riteau B. PAR1 contributes to influenza A virus pathogenicity in mice. J Clin Invest 2012. [PMID: 23202729 DOI: 10.1172/jci61667] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Influenza causes substantial morbidity and mortality, and highly pathogenic and drug-resistant strains are likely to emerge in the future. Protease-activated receptor 1 (PAR1) is a thrombin-activated receptor that contributes to inflammatory responses at mucosal surfaces. The role of PAR1 in pathogenesis of virus infections is unknown. Here, we demonstrate that PAR1 contributed to the deleterious inflammatory response after influenza virus infection in mice. Activating PAR1 by administering the agonist TFLLR-NH2 decreased survival and increased lung inflammation after influenza infection. Importantly, both administration of a PAR1 antagonist and PAR1 deficiency protected mice from infection with influenza A viruses (IAVs). Treatment with the PAR1 agonist did not alter survival of mice deficient in plasminogen (PLG), which suggests that PLG permits and/or interacts with a PAR1 function in this model. PAR1 antagonists are in human trials for other indications. Our findings suggest that PAR1 antagonism might be explored as a treatment for influenza, including that caused by highly pathogenic H5N1 and oseltamivir-resistant H1N1 viruses.
Collapse
Affiliation(s)
- Khaled Khoufache
- Virologie et Pathologie Humaine, EA 4610, Université Lyon1, Faculté de Médecine RTH Laennec, Lyon, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Xu H, Yan J, Zhu Z, Hussain LR, Huang Y, Ding C, Bozulic LD, Wen Y, Ildstad ST. A critical role for the TLR4/TRIF pathway in allogeneic hematopoietic cell rejection by innate immune cells. Cell Transplant 2012; 22:2367-80. [PMID: 23146386 DOI: 10.3727/096368912x658881] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
We show for the first time that signaling through the TLR4/TRIF pathway plays a critical role in allogeneic bone marrow cell (BMC) rejection. This appears to be unique to BMCs as organ allografts are rejected mainly via MyD88 signaling. Using T- or T-/B-cell-deficient mice, we found that BMC allorejection occurred early before T-cell activation and was T- and B-cell independent, suggesting an effector role for innate immune cells in BMC rejection. We further demonstrated the innate immune signaling in BMC allorejection by showing superior engraftment in mice deficient in TRIF or TLR4 but not in MyD88 or TLR3. The restored cytotoxicity in TRIF-deficient recipients transferred with wild-type F4/80(+) or NK1.1(+) cells suggests TRIF signaling dependence on macrophages or NK cells in early BMC rejection. Production of the proinflammatory cytokine IL-6 and TRIF relevant chemokine MCP-1 was significantly increased early after bone marrow transplantation. In vivo specific depletion of macrophages or NK innate immune cells in combination with anti-CD154/rapamycin resulted in additive-enhanced allogeneic engraftment. The requirement for irradiation was completely eliminated when both macrophages and NK cells were depleted in combination with anti-CD154/rapamycin to target T- and B-cells, supporting the hypothesis that two barriers involving innate and adaptive immunity exist in mediating the rejection of allogeneic BMCs. In summary, our results clearly demonstrate a previously unappreciated role for innate immunity in BMC allorejection via signaling through a unique MyD88-independent TLR4/TRIF mechanism. These findings may have direct clinical impact on strategies for conditioning recipients for stem cell transplantation.
Collapse
Affiliation(s)
- Hong Xu
- Institute for Cellular Therapeutics, University of Louisville, Louisville, KY, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
|
48
|
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
|
49
|
Koo BH, Kim YH, Han JH, Kim DS. Dimerization of matrix metalloproteinase-2 (MMP-2): functional implication in MMP-2 activation. J Biol Chem 2012; 287:22643-53. [PMID: 22577146 DOI: 10.1074/jbc.m111.337949] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Matrix metalloproteinase-2 (MMP-2) functions in diverse biological processes through the degradation of extracellular and non-extracellular matrix molecules. Because of its potential for tissue damage, there are several ways to regulate MMP-2 activity, including gene expression, compartmentalization, zymogen activation, and enzyme inactivation by extracellular inhibitors. Enzyme regulation through zymogen activation is important for the regulation of MMP-2 activity. In our previous studies, we showed that thrombin directly cleaved the propeptide of MMP-2 at specific sites for enzyme activation. We also demonstrated that heparan sulfate was required for thrombin-mediated activation of pro-MMP-2 by binding to thrombin, presumably through conformational changes at the active site of the enzyme. This suggests a regulatory mechanism for thrombin-mediated activation of pro-MMP-2. In this study, we found that MMP-2 formed a reduction-sensitive homodimer in a controlled manner and that Ca(2+) ion was essential for homodimerization of MMP-2. Homodimerization was not associated with protein kinase C-mediated phosphorylation of MMP-2. MMP-2 formed a homodimer through an intermolecular disulfide bond between Cys(102) and the neighboring Cys(102). Homodimerization of MMP-2 enhanced thrombin-mediated activation of pro-MMP-2. Moreover, the MMP-2 homodimer could cleave a small peptide substrate without removal of the propeptide. Taken together, our experimental data suggest a novel regulatory mechanism for pro-MMP-2 activation that is modulated through homodimerization of MMP-2.
Collapse
Affiliation(s)
- Bon-Hun Koo
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea.
| | | | | | | |
Collapse
|
50
|
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]
|