1
|
Wu Y, Wagner WD. Syndecan-4 Functionalization Reduces the Thrombogenicity of Engineered Vascular Biomaterials. Ann Biomed Eng 2024; 52:1873-1882. [PMID: 37071281 PMCID: PMC11169030 DOI: 10.1007/s10439-023-03199-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/27/2023] [Indexed: 04/19/2023]
Abstract
Blood-biomaterial compatibility is essential for tissue repair especially for endovascular biomaterials where small-diameter vessel patency and endothelium formation is crucial. To address this issue, a composite biomaterial termed PFC fabricated from poly (glycerol sebacate), silk fibroin, and collagen was used to determine if functionalization with syndecan-4 (SYN4) would reduce thrombogenesis through the action of heparan sulfate. The material termed, PFC_SYN4, has structure and composition similar to native arterial tissue and has been reported to facilitate the binding and differentiation of endothelial colony-forming cells (ECFCs). In this study, the hemocompatibility of PFC_SYN4 was evaluated and compared with non-functionalized PFC, electrospun collagen, ePTFE, and bovine pericardial patch (BPV). Ultrastructurally, platelets were less activated when cultured on PFC and PFC_SYN4 compared to collagen where extensive platelet degranulation was observed. Quantitatively, 31% and 44% fewer platelets adhered to PFC_SYN4 compared to non-functionalized PFC and collagen, respectively. Functionalization of PFC resulted in reduced levels of complement activation compared to PFC, collagen, and BPV. Whole blood clotting times indicated that PFC_SYN4 was less thrombogenic compared with PFC, collagen, and BPV. These results suggest that syndecan-4 functionalization of blood-contacting biomaterials provides a novel solution for generating a reduced thrombogenic surface.
Collapse
Affiliation(s)
- Yidi Wu
- Department of Plastic & Reconstructive Surgery, Medical Center Boulevard, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
- Virginia Tech - Wake Forest University School of Biomedical Engineering and Sciences, Winston-Salem, NC, USA
| | - William D Wagner
- Department of Plastic & Reconstructive Surgery, Medical Center Boulevard, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
- Virginia Tech - Wake Forest University School of Biomedical Engineering and Sciences, Winston-Salem, NC, USA.
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way, Winston-Salem, NC, USA.
| |
Collapse
|
2
|
Kunnathattil M, Rahul P, Skaria T. Soluble vascular endothelial glycocalyx proteoglycans as potential therapeutic targets in inflammatory diseases. Immunol Cell Biol 2024; 102:97-116. [PMID: 37982607 DOI: 10.1111/imcb.12712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/21/2023]
Abstract
Reducing the activity of cytokines and leukocyte extravasation is an emerging therapeutic strategy to limit tissue-damaging inflammatory responses and restore immune homeostasis in inflammatory diseases. Proteoglycans embedded in the vascular endothelial glycocalyx, which regulate the activity of cytokines to restrict the inflammatory response in physiological conditions, are proteolytically cleaved in inflammatory diseases. Here we critically review the potential of proteolytically shed, soluble vascular endothelial glycocalyx proteoglycans to modulate pathological inflammatory responses. Soluble forms of the proteoglycans syndecan-1, syndecan-3 and biglycan exert beneficial anti-inflammatory effects by the removal of chemokines, suppression of proinflammatory cytokine expression and leukocyte migration, and induction of autophagy of proinflammatory M1 macrophages. By contrast, soluble versikine and decorin enhance proinflammatory responses by increasing inflammatory cytokine synthesis and leukocyte migration. Endogenous syndecan-2 and mimecan exert proinflammatory effects, syndecan-4 and perlecan mediate beneficial anti-inflammatory effects and glypican regulates Hh and Wnt signaling pathways involved in systemic inflammatory responses. Taken together, targeting the vascular endothelial glycocalyx-derived, soluble syndecan-1, syndecan-2, syndecan-3, syndecan-4, biglycan, versikine, mimecan, perlecan, glypican and decorin might be a potential therapeutic strategy to suppress overstimulated cytokine and leukocyte responses in inflammatory diseases.
Collapse
Affiliation(s)
- Maneesha Kunnathattil
- Department of Zoology, Government College Madappally, University of Calicut, Calicut, Kerala, India
| | - Pedapudi Rahul
- School of Biotechnology, National Institute of Technology Calicut, Calicut, Kerala, India
| | - Tom Skaria
- School of Biotechnology, National Institute of Technology Calicut, Calicut, Kerala, India
| |
Collapse
|
3
|
Wiedermann CJ. Antithrombin as Therapeutic Intervention against Sepsis-Induced Coagulopathy and Disseminated Intravascular Coagulation: Lessons Learned from COVID-19-Associated Coagulopathy. Int J Mol Sci 2022; 23:ijms232012474. [PMID: 36293332 PMCID: PMC9604230 DOI: 10.3390/ijms232012474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 12/04/2022] Open
Abstract
Recent research has contributed significantly to our understanding of the pathogenesis of acute disseminated intravascular coagulation. COVID-19 can be considered as a new underlying condition of disseminated intravascular coagulation. In this narrative review, current evidence is presented regarding biomarker differences between sepsis-induced and COVID-19-associated coagulopathies, supporting the importance of acquired antithrombin deficiency in the early differential diagnosis of septic coagulopathy and its potential impact on treatment with endogenous anticoagulants. Establishing new scoring systems for septic coagulopathy in combination with endogenous anticoagulant biomarker activities may allow for the identification of those in the heterogeneous population of sepsis patients who are more likely to benefit from targeted specific treatment interventions.
Collapse
Affiliation(s)
- Christian J. Wiedermann
- Institute of General Practice, Claudiana—College of Health Professions, 39100 Bolzano, Italy;
- Department of Public Health, Medical Decision Making and HTA, University of Health Sciences, Medical Informatics and Technology—Tyrol, 6060 Hall in Tyrol, Austria
| |
Collapse
|
4
|
Popescu NI, Lupu C, Lupu F. Disseminated intravascular coagulation and its immune mechanisms. Blood 2022; 139:1973-1986. [PMID: 34428280 PMCID: PMC8972096 DOI: 10.1182/blood.2020007208] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/02/2021] [Indexed: 11/26/2022] Open
Abstract
Disseminated intravascular coagulation (DIC) is a syndrome triggered by infectious and noninfectious pathologies characterized by excessive generation of thrombin within the vasculature and widespread proteolytic conversion of fibrinogen. Despite diverse clinical manifestations ranging from thrombo-occlusive damage to bleeding diathesis, DIC etiology commonly involves excessive activation of blood coagulation and overlapping dysregulation of anticoagulants and fibrinolysis. Initiation of blood coagulation follows intravascular expression of tissue factor or activation of the contact pathway in response to pathogen-associated or host-derived, damage-associated molecular patterns. The process is further amplified through inflammatory and immunothrombotic mechanisms. Consumption of anticoagulants and disruption of endothelial homeostasis lower the regulatory control and disseminate microvascular thrombosis. Clinical DIC development in patients is associated with worsening morbidities and increased mortality, regardless of the underlying pathology; therefore, timely recognition of DIC is critical for reducing the pathologic burden. Due to the diversity of triggers and pathogenic mechanisms leading to DIC, diagnosis is based on algorithms that quantify hemostatic imbalance, thrombocytopenia, and fibrinogen conversion. Because current diagnosis primarily assesses overt consumptive coagulopathies, there is a critical need for better recognition of nonovert DIC and/or pre-DIC states. Therapeutic strategies for patients with DIC involve resolution of the eliciting triggers and supportive care for the hemostatic imbalance. Despite medical care, mortality in patients with DIC remains high, and new strategies, tailored to the underlying pathologic mechanisms, are needed.
Collapse
Affiliation(s)
| | - Cristina Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK; and
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK; and
- Department of Cell Biology
- Department of Pathology, and
- Department of Internal Medicine, Oklahoma University Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
5
|
Pérez LA, Leyton L, Valdivia A. Thy-1 (CD90), Integrins and Syndecan 4 are Key Regulators of Skin Wound Healing. Front Cell Dev Biol 2022; 10:810474. [PMID: 35186924 PMCID: PMC8851320 DOI: 10.3389/fcell.2022.810474] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
Acute skin wound healing is a multistage process consisting of a plethora of tightly regulated signaling events in specialized cells. The Thy-1 (CD90) glycoprotein interacts with integrins and the heparan sulfate proteoglycan syndecan 4, generating a trimolecular complex that triggers bi-directional signaling to regulate diverse aspects of the wound healing process. These proteins can act either as ligands or receptors, and they are critical for the successful progression of wound healing. The expression of Thy-1, integrins, and syndecan 4 is controlled during the healing process, and the lack of expression of any of these proteins results in delayed wound healing. Here, we review and discuss the roles and regulatory events along the stages of wound healing that support the relevance of Thy-1, integrins, and syndecan 4 as crucial regulators of skin wound healing.
Collapse
Affiliation(s)
- Leonardo A. Pérez
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Universidad de Chile, Santiago, Chile
- *Correspondence: Lisette Leyton, ; Alejandra Valdivia,
| | - Alejandra Valdivia
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States
- *Correspondence: Lisette Leyton, ; Alejandra Valdivia,
| |
Collapse
|
6
|
Fang XZ, Wang YX, Xu JQ, He YJ, Peng ZK, Shang Y. Immunothrombosis in Acute Respiratory Dysfunction of COVID-19. Front Immunol 2021; 12:651545. [PMID: 34149692 PMCID: PMC8207198 DOI: 10.3389/fimmu.2021.651545] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 05/12/2021] [Indexed: 01/10/2023] Open
Abstract
COVID-19 is an acute, complex disorder that was caused by a new β-coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Based on current reports, it was surprising that the characteristics of many patients with COVID-19, who fulfil the Berlin criteria for acute respiratory distress syndrome (ARDS), are not always like those of patients with typical ARDS and can change over time. While the mechanisms of COVID-19–related respiratory dysfunction in COVID-19 have not yet been fully elucidated, pulmonary microvascular thrombosis is speculated to be involved. Considering that thrombosis is highly related to other inflammatory lung diseases, immunothrombosis, a two-way process that links coagulation and inflammation, seems to be involved in the pathophysiology of COVID-19, including respiratory dysfunction. Thus, the current manuscript will describe the proinflammatory milieu in COVID-19, summarize current evidence of thrombosis in COVID-19, and discuss possible interactions between these two.
Collapse
Affiliation(s)
- Xiang-Zhi Fang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Xin Wang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ji-Qain Xu
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Jun He
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhe-Kang Peng
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Antithrombin and Its Role in Host Defense and Inflammation. Int J Mol Sci 2021; 22:ijms22084283. [PMID: 33924175 PMCID: PMC8074369 DOI: 10.3390/ijms22084283] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 12/12/2022] Open
Abstract
Antithrombin (AT) is a natural anticoagulant that interacts with activated proteases of the coagulation system and with heparan sulfate proteoglycans (HSPG) on the surface of cells. The protein, which is synthesized in the liver, is also essential to confer the effects of therapeutic heparin. However, AT levels drop in systemic inflammatory diseases. The reason for this decline is consumption by the coagulation system but also by immunological processes. Aside from the primarily known anticoagulant effects, AT elicits distinct anti-inflammatory signaling responses. It binds to structures of the glycocalyx (syndecan-4) and further modulates the inflammatory response of endothelial cells and leukocytes by interacting with surface receptors. Additionally, AT exerts direct antimicrobial effects: depending on AT glycosylation it can bind to and perforate bacterial cell walls. Peptide fragments derived from proteolytic degradation of AT exert antibacterial properties. Despite these promising characteristics, therapeutic supplementation in inflammatory conditions has not proven to be effective in randomized control trials. Nevertheless, new insights provided by subgroup analyses and retrospective trials suggest that a recommendation be made to identify the patient population that would benefit most from AT substitution. Recent experiment findings place the role of various AT isoforms in the spotlight. This review provides an overview of new insights into a supposedly well-known molecule.
Collapse
|
8
|
Okimoto S, Tashiro H, Iwako H, Kuroda S, Kobayashi T, Hinoi T, Ohdan H. Antithrombin attenuates the progression of hepatocellular carcinoma by regulating neutrophil/interleukin-8 signaling. Hepatol Res 2020; 50:1284-1296. [PMID: 32893935 DOI: 10.1111/hepr.13558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/22/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022]
Abstract
AIM Inflammation plays an important role in hepatocellular carcinoma (HCC) progression. Here, we examined whether antithrombin (AT) plays a role in attenuating HCC progression, via its anti-inflammatory effects. METHODS HCCs were developed in AT-insufficient (AT+/- ) mice and wild-type (AT+/+ ) mice treated with diethyl nitrosamine and carbon tetrachloride. AT was administered to AT+/- mice. The development of HCC was compared between the three groups. In vitro study, migration assay was performed. The association of the prognosis of patients with HCC and plasma AT values was clinically examined. RESULTS AT suppressed the release of interleukin (IL)-8 from lipopolysaccharide (LPS)-stimulated human neutrophils in vitro. Huh-7 cells that were co-cultured with neutrophils and stimulated with LPS showed significantly enhanced migration; however, Huh-7 cells co-cultured with LPS/AT-stimulated neutrophils showed significantly decreased migration. Moreover, the addition of anti-IL-8 antibodies to LPS-stimulated Huh-7 cells co-cultured with neutrophils also suppressed migration. AT+/- mice (AT plasma activity: 64%) promoted liver cancer, as compared with wild-type mice (AT plasma activity: 135%); AT administration attenuated liver cancer in AT+/- mice. Patients with HCC with a preoperative AT level of ≥70% showed better outcomes after liver resection, as compared with those with an AT level of <70%. IL-8 expression and neutrophil infiltration in HCC tissues were negatively correlated with the AT level. CONCLUSIONS AT attenuates HCC progression by regulating neutrophil/IL-8 signaling.
Collapse
Affiliation(s)
- Sho Okimoto
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Kasumi, Hiroshima, Japan
| | - Hirotaka Tashiro
- Department of Surgery, National Hospital Organization Kure Medical Center, Kure City, Hiroshima, Japan
| | - Hiroshi Iwako
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Kasumi, Hiroshima, Japan
| | - Shintaro Kuroda
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Kasumi, Hiroshima, Japan
| | - Tsuyoshi Kobayashi
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Kasumi, Hiroshima, Japan
| | - Takao Hinoi
- Department of clinical and molecular Genetics, Hiroshima University Hospital, Kasumi, Hiroshima, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Kasumi, Hiroshima, Japan
| |
Collapse
|
9
|
Juschten J, Ingelse SA, Maas MAW, Girbes ARJ, Juffermans NP, Schultz MJ, Tuinman PR. Antithrombin plus alpha-1 protease inhibitor does not affect coagulation and inflammation in two murine models of acute lung injury. Intensive Care Med Exp 2019; 7:36. [PMID: 31346884 PMCID: PMC6658634 DOI: 10.1186/s40635-019-0240-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 03/07/2019] [Indexed: 01/23/2023] Open
Abstract
Background In acute respiratory distress syndrome (ARDS), uncontrolled production of activators of coagulation and proinflammatory mediators results in a shift from an adequate local innate immune response to hypercoagulability and inflammation. This study aimed to investigate whether the protease inhibitors antithrombin (AT) and alpha-1 protease inhibitor (A1PI) may attenuate an exaggerated pulmonary immune response. Methods Lung injury was induced either by single intranasal administration of lipopolysaccharide (LPS) (5 mg/kg) in BALB/c mice or by combination of an intravenous injection of LPS (10 mg/kg) with subsequent injurious ventilation using high tidal volumes (12–15 ml/kg) for 4 h in RccHan Wistar rats. Animals received either a single bolus of AT (250 IU/kg) or A1PI (60 mg/kg) alone or in combination, with or without intravenous low-dose heparin (100 U/kg). Control animals received saline. Additional controls received neither LPS, nor ventilation, nor treatment. Endpoints were local and systemic markers of coagulation, e.g., thrombin–antithrombin complexes (TATc), and inflammation, e.g., interleukin-6. Results Both lung injury models resulted in a pronounced immune response within the pulmonary compartment shown by elevated levels of markers of coagulation and inflammation. The two-hit lung injury model also induced profound systemic coagulopathy and inflammation. Monotherapy with AT or A1PI did not reduce pulmonary coagulopathy or inflammation in any lung injury model. Nor did combination therapy with AT and A1PI result in a decrease of coagulation or inflammatory parameters. AT markedly reduced systemic levels of TATc in the two-hit lung injury model. Systemic inflammation was not affected by the different interventions. Additional administration of heparin did not lead to macroscopic bleeding incidences. Conclusions In two different murine models of acute lung injury, neither single therapy with AT or A1PI nor combination of both agents attenuates the pronounced pulmonary coagulation or inflammatory response.
Collapse
Affiliation(s)
- Jenny Juschten
- Department of Intensive Care, Amsterdam UMC, VU Medical Center, Amsterdam, Netherlands. .,Research VUmc Intensive Care (REVIVE), Amsterdam UMC, VU Medical Center, Amsterdam, Netherlands. .,Department of Intensive Care, Amsterdam UMC, Academic Medical Center, Amsterdam, Netherlands. .,Laboratory of Experimental Intensive Care and Anesthesiology (L·E·I·C·A), Amsterdam UMC, Academic Medical Center, Amsterdam, Netherlands.
| | - Sarah Anne Ingelse
- Laboratory of Experimental Intensive Care and Anesthesiology (L·E·I·C·A), Amsterdam UMC, Academic Medical Center, Amsterdam, Netherlands.,Emma Children's Hospital-Pediatric Intensive Care Unit, Amsterdam UMC, Academic Medical Center, Amsterdam, Netherlands
| | - Martinus Adrianus Wilhelmus Maas
- Laboratory of Experimental Intensive Care and Anesthesiology (L·E·I·C·A), Amsterdam UMC, Academic Medical Center, Amsterdam, Netherlands
| | - Armand Roelof Johan Girbes
- Department of Intensive Care, Amsterdam UMC, VU Medical Center, Amsterdam, Netherlands.,Research VUmc Intensive Care (REVIVE), Amsterdam UMC, VU Medical Center, Amsterdam, Netherlands
| | - Nicole Petra Juffermans
- Department of Intensive Care, Amsterdam UMC, Academic Medical Center, Amsterdam, Netherlands.,Laboratory of Experimental Intensive Care and Anesthesiology (L·E·I·C·A), Amsterdam UMC, Academic Medical Center, Amsterdam, Netherlands
| | - Marcus Josephus Schultz
- Department of Intensive Care, Amsterdam UMC, Academic Medical Center, Amsterdam, Netherlands.,Mahidol Oxford Tropical Medicine Research Unit (MORU), Mahidol University, Bangkok, Thailand
| | - Pieter Roel Tuinman
- Department of Intensive Care, Amsterdam UMC, VU Medical Center, Amsterdam, Netherlands.,Research VUmc Intensive Care (REVIVE), Amsterdam UMC, VU Medical Center, Amsterdam, Netherlands
| |
Collapse
|
10
|
Smits NC, Kobayashi T, Srivastava PK, Skopelja S, Ivy JA, Elwood DJ, Stan RV, Tsongalis GJ, Sellke FW, Gross PL, Cole MD, DeVries JT, Kaplan AV, Robb JF, Williams SM, Shworak NW. HS3ST1 genotype regulates antithrombin's inflammomodulatory tone and associates with atherosclerosis. Matrix Biol 2017; 63:69-90. [PMID: 28126521 DOI: 10.1016/j.matbio.2017.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 01/19/2017] [Accepted: 01/20/2017] [Indexed: 12/21/2022]
Abstract
The HS3ST1 gene controls endothelial cell production of HSAT+ - a form of heparan sulfate containing a specific pentasaccharide motif that binds the anticoagulant protein antithrombin (AT). HSAT+ has long been thought to act as an endogenous anticoagulant; however, coagulation was normal in Hs3st1-/- mice that have greatly reduced HSAT+ (HajMohammadi et al., 2003). This finding indicates that HSAT+ is not essential for AT's anticoagulant activity. To determine if HSAT+ is involved in AT's poorly understood inflammomodulatory activities, Hs3st1-/- and Hs3st1+/+ mice were subjected to a model of acute septic shock. Compared with Hs3st1+/+ mice, Hs3st1-/- mice were more susceptible to LPS-induced death due to an increased sensitivity to TNF. For Hs3st1+/+ mice, AT treatment reduced LPS-lethality, reduced leukocyte firm adhesion to endothelial cells, and dilated isolated coronary arterioles. Conversely, for Hs3st1-/- mice, AT induced the opposite effects. Thus, in the context of acute inflammation, HSAT+ selectively mediates AT's anti-inflammatory activity; in the absence of HSAT+, AT's pro-inflammatory effects predominate. To explore if the anti-inflammatory action of HSAT+ also protects against a chronic vascular-inflammatory disease, atherosclerosis, we conducted a human candidate-gene association study on >2000 coronary catheterization patients. Bioinformatic analysis of the HS3ST1 gene identified an intronic SNP, rs16881446, in a putative transcriptional regulatory region. The rs16881446G/G genotype independently associated with the severity of coronary artery disease and atherosclerotic cardiovascular events. In primary endothelial cells, the rs16881446G allele associated with reduced HS3ST1 expression. Together with the mouse data, this leads us to conclude that the HS3ST1 gene is required for AT's anti-inflammatory activity that appears to protect against acute and chronic inflammatory disorders.
Collapse
Affiliation(s)
- Nicole C Smits
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Takashi Kobayashi
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Pratyaksh K Srivastava
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Sladjana Skopelja
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Julianne A Ivy
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Dustin J Elwood
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Radu V Stan
- Department of Pathology, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Gregory J Tsongalis
- Department of Pathology, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Brown Medical School, Providence, RI, USA
| | - Peter L Gross
- Department of Medicine, Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Michael D Cole
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - James T DeVries
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Aaron V Kaplan
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - John F Robb
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Scott M Williams
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Nicholas W Shworak
- Section of Cardiology, Department of Medicine, Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA; Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
| |
Collapse
|
11
|
Urbinati C, Grillo E, Chiodelli P, Tobia C, Caccuri F, Fiorentini S, David G, Rusnati M. Syndecan-1 increases B-lymphoid cell extravasation in response to HIV-1 Tat via α vβ 3/pp60src/pp125FAK pathway. Oncogene 2016; 36:2609-2618. [PMID: 27819680 DOI: 10.1038/onc.2016.420] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 09/21/2016] [Accepted: 09/30/2016] [Indexed: 01/11/2023]
Abstract
Syndecan-1 is a heparan sulfate proteoglycan (HSPG) commonly upregulated in AIDS-related B lymphoid malignancies. Tat is the main HIV-1 transactivating factor that has a major role in the pathogenesis of AIDS-related lymphomas (ARL) by engaging heparan sulfate proteoglycans (HSPGs), chemokine receptors and integrins at the lymphoid cell (LC) surface. Here B-lymphoid Namalwa cell clones that do not express or overexpress syndecan-1 (EV-Ncs and SYN-Ncs, respectively) were compared for their responsiveness with Tat: in the absence of syndecan-1, Tat induces a limited EV-Nc migration via C-X-C motif chemokine receptor 4 (CXCR4), G-proteins and Rac. Syndecan-1 overexpression increases SYN-Nc responsiveness to Tat and makes this response independent from CXCR4 and G-protein and dependent instead on pp60src phosphorylation. Tat-induced SYN-Nc migration and pp60src phosphorylation require the engagement of αvβ3 integrin and consequent pp125FAK phosphorylation. This complex set of Tat-driven activations is orchestrated by the direct interaction of syndecan-1 with pp60src and its simultaneous coupling with αvβ3. The Tat/syndecan-1/αvβ3 interplay is retained in vivo and is shared also by other syndecan-1+ B-LCs, including BJAB cells, whose responsiveness to Tat is inhibited by syndecan-1 knockdown. In conclusion, overexpression of syndecan-1 confers to B-LCs an increased capacity to migrate in response to Tat, owing to a switch from a CXCR4/G-protein/Rac to a syndecan-1/αvβ3/pp60src/pp125FAK signal transduction pathway that depends on the formation of a complex in which syndecan-1 interacts with Tat via its HS-chains, with αvβ3 via its core protein ectodomain and with pp60src via its intracellular tail. These findings have implications in ARL progression and may help in identifying new therapeutical targets for the treatment of AIDS-associated neoplasia.
Collapse
Affiliation(s)
- C Urbinati
- Section of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - E Grillo
- Section of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - P Chiodelli
- Section of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - C Tobia
- Section of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - F Caccuri
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - S Fiorentini
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - G David
- Department of Human Genetics, University of Leuven and Flanders Institute for Biotechnology, Leuven, Belgium
| | - M Rusnati
- Section of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
12
|
The PDZ-binding domain of syndecan-2 inhibits LFA-1 high-affinity conformation. Cell Signal 2014; 26:1489-99. [DOI: 10.1016/j.cellsig.2014.03.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 03/16/2014] [Indexed: 01/13/2023]
|
13
|
Ornaghi S, Barnhart KT, Frieling J, Streisand J, Paidas MJ. Clinical syndromes associated with acquired antithrombin deficiency via microvascular leakage and the related risk of thrombosis. Thromb Res 2014; 133:972-84. [PMID: 24593911 DOI: 10.1016/j.thromres.2014.02.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 01/25/2014] [Accepted: 02/11/2014] [Indexed: 12/17/2022]
Abstract
Antithrombin (AT) is a 65kDa glycoprotein belonging to a group of inhibitory factors known as serpins (serine protease inhibitors). It plays a critical role in the inhibition of coagulation and inflammation processes within the environment of the vascular endothelium. Inadequate levels of functional AT in plasma results in an increased risk of thrombotic events, both venous and arterial. AT deficiency can be inherited or acquired. Congenital AT deficiency is the most severe inherited thrombophilic condition with an odds ratio of 20 for the increased risk of venous thrombosis. Acquired AT deficiency occurs in a variety of physiologic and pathologic medical conditions with similar risks of increased thrombosis. In this article, we review clinical settings characterized by an acquired AT deficiency largely or partly subsequent to protein microvascular leakage. Other different mechanisms of AT depletion are implied in some clinical conditions together with endothelial loss, and, therefore, outlined. In addition, we provide a description of the current knowledge on the specific mechanisms underlying endothelial AT leakage and on the consequences of this protein decrease, specifically looking at thrombosis. We identify potential directions of research that might prove useful in patients with acquired AT deficiency.
Collapse
Affiliation(s)
- Sara Ornaghi
- Yale Women and Children's Center For Blood Disorders, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, USA; Department of Obstetrics and Gynecology, University of Milan-Bicocca, via Pergolesi 33, Monza, MB, Italy.
| | - Kurt T Barnhart
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| | - Johan Frieling
- rEVO Biologics 175 Crossing Boulevard, Framingham, MA 01702, USA
| | - James Streisand
- rEVO Biologics 175 Crossing Boulevard, Framingham, MA 01702, USA
| | - Michael J Paidas
- Yale Women and Children's Center For Blood Disorders, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, USA
| |
Collapse
|
14
|
de Morais-Zani K, Nunes FPB, da Silva JB, Ferreira MJ, Grego KF, Lopes-Ferreira M, Tanaka AS, Tanaka-Azevedo AM. The anti-inflammatory action of Bothrops jararaca snake antithrombin on acute inflammation induced by carrageenan in mice. Inflamm Res 2013; 62:733-42. [PMID: 23665851 DOI: 10.1007/s00011-013-0628-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 02/18/2013] [Accepted: 04/23/2013] [Indexed: 10/26/2022] Open
Abstract
OBJECTIVE AND DESIGN Antithrombin is known as the most important natural coagulation inhibitor and has been shown to have anti-inflammatory properties. The present study aimed to investigate the effects of Bothrops jararaca antithrombin on acute inflammation induced by carrageenan in mice. METHODS We evaluated the anti-inflammatory activity of antithrombin on models of paw edema formation, cell migration and leukocyte-endothelium interaction in mice (Swiss; n = 5). Acute inflammation was induced by the administration of carrageenan (15 mg kg⁻¹). RESULTS Treatment with B. jararaca antithrombin (1 mg kg⁻¹) 1 h before or after carrageenan administration significantly inhibited paw edema formation, reduced cell influx to the peritoneal cavity due to reduction in the migration of polymorphonuclear cells, and attenuated leukocyte rolling in the microcirculation of the cremaster muscle.The effects of antithrombin on vascular and cellular events of inflammation were completely abolished by treatment with the cyclo-oxygenase inhibitor indomethacin (4 mg kg⁻¹), suggesting the involvement of prostacyclin in the mechanism of inflammation inhibition by B. jararaca antithrombin. CONCLUSION This work showed for the first time the anti-inflammatory properties of B. jararaca antithrombin on vascular and cellular events of inflammation. These findings suggest that antithrombin is effective in preventing paw edema formation, cell migration and leukocyte rolling induced by carrageenan in mice.
Collapse
Affiliation(s)
- Karen de Morais-Zani
- Laboratório de Herpetologia, Instituto Butantan, Avenida Vital Brazil, 1500, CEP 05503-900, São Paulo, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Antithrombin Reduces Inflammation and Microcirculatory Perfusion Failure in Closed Soft-Tissue Injury and Endotoxemia. Crit Care Med 2013; 41:867-73. [DOI: 10.1097/ccm.0b013e3182742d2c] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
16
|
Asmal M, Whitney JB, Luedemann C, Carville A, Steen R, Letvin NL, Geiben-Lynn R. In vivo anti-HIV activity of the heparin-activated serine protease inhibitor antithrombin III encapsulated in lymph-targeting immunoliposomes. PLoS One 2012; 7:e48234. [PMID: 23133620 PMCID: PMC3487854 DOI: 10.1371/journal.pone.0048234] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 09/28/2012] [Indexed: 11/23/2022] Open
Abstract
Endogenous serine protease inhibitors (serpins) are anti-inflammatory mediators with multiple biologic functions. Several serpins have been reported to modulate HIV pathogenesis, or exhibit potent anti-HIV activity in vitro, but the efficacy of serpins as therapeutic agents for HIV in vivo has not yet been demonstrated. In the present study, we show that heparin-activated antithrombin III (hep-ATIII), a member of the serpin family, significantly inhibits lentiviral replication in a non-human primate model. We further demonstrate greater than one log(10) reduction in plasma viremia in the nonhuman primate system by loading of hep-ATIII into anti-HLA-DR immunoliposomes, which target tissue reservoirs of viral replication. We also demonstrate the utility of hep-ATIIII as a potential salvage agent for HIV strains resistant to standard anti-retroviral treatment. Finally, we applied gene-expression arrays to analyze hep-ATIII-induced host cell interactomes and found that downstream of hep-ATIII, two independent gene networks were modulated by host factors prostaglandin synthetase-2, ERK1/2 and NFκB. Ultimately, understanding how serpins, such as hep-ATIII, regulate host responses during HIV infection may reveal new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Mohammed Asmal
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America.
| | | | | | | | | | | | | |
Collapse
|
17
|
O'Brien M. The reciprocal relationship between inflammation and coagulation. Top Companion Anim Med 2012; 27:46-52. [PMID: 23031455 DOI: 10.1053/j.tcam.2012.06.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 06/18/2012] [Indexed: 12/21/2022]
Abstract
Inflammation and coagulation constitute two host defense systems with complementary roles in eliminating invading pathogens, limiting tissue damage, and restoring homeostasis. Extensive cross talk exists between these 2 systems, whereby inflammation leads to activation of coagulation, and coagulation considerably affects inflammatory activity. Infection leads to the production of proinflammatory cytokines that, in turn, stimulate the production of tissue factor. Activation of the coagulation system and ensuing thrombin generation are dependent on the expression of tissue factor. Conversely, activated coagulation proteases may affect specific receptors on inflammatory cells and endothelial cells and thereby modulate the inflammatory response. Activation of coagulation with the simultaneous down-regulation of endothelial-bound anticoagulant mechanisms and endogenous fibrinolysis characterizes the pathophysiology of sepsis. The mechanisms by which these highly complex and codependent defense strategies are linked together both in health and disease is the focus of this review.
Collapse
Affiliation(s)
- Mauria O'Brien
- University of Illinois Urbana-Champaign, College of Veterinary Medicine, Urbana, IL 61802, USA.
| |
Collapse
|
18
|
Prevention of High-Mobility Group Box 1-Mediated Early Loss of Transplanted Mouse Islets in the Liver by Antithrombin III. Transplantation 2012; 93:983-8. [DOI: 10.1097/tp.0b013e31824d3508] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
19
|
Kubier A, O'Brien M. Endogenous Anticoagulants. Top Companion Anim Med 2012; 27:81-7. [DOI: 10.1053/j.tcam.2012.07.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 07/17/2012] [Indexed: 12/21/2022]
|
20
|
Suga N, Sugimura M, Koshiishi T, Yorifuji T, Makino S, Takeda S. Heparin/heparan sulfate/CD44-v3 enhances cell migration in term placenta-derived immortalized human trophoblast cells. Biol Reprod 2012; 86:134, 1-8. [PMID: 22321833 DOI: 10.1095/biolreprod.111.093690] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The function of CD44-v3 and heparin/heparan sulfate (HS) signaling was investigated during trophoblast cell migration to identify their role in the renewal of syncytial layer damage caused by increased hemodynamic turbulence in the intervillous space and maintenance of syncytial integrity in pre-eclampsia. We evaluated the effect of heparin/HS/CD44-v3-mediated processes during scratch wound closure in monolayer immortalized human trophoblast cells derived from term placenta (TCL-1 cells). Western blot analysis showed that these cultured human trophoblast cells express the epidermal growth factor receptor and CD44-v3 but do not express syndecan 4. An in vitro scratch wound healing assay showed enhanced migration of trophoblast cells in a dose-dependent manner in the presence of heparin compared with controls when cultured under serum-free conditions. Conversely, an anti-CD44 function-blocking antibody and CD44 siRNA suppressed the migration of trophoblast cells in the presence of heparin in a similar scratch assay. Furthermore, both heparin treatment and in vitro scratch wounding induced the phosphorylation of p21-activated kinase 1 (PAK1), whereas the anti-CD44-v3 antibody suppressed the heparin-induced phosphorylation of PAK1 in trophoblast cells. These results indicate that heparin/HS/CD44-v3-mediated signaling, in the absence of growth factor networks, enhances the direct repair of the damaged trophoblast layer through the migration of trophoblast cells. This renewed cell coverage may lead to the maintenance of syncytiotrophoblast cell function and an associated reduction in pathogenic soluble factors derived from the damaged trophoblast cells.
Collapse
Affiliation(s)
- Naoko Suga
- Department of Obstetrics and Gynecology, School of Medicine, Juntendo University, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
21
|
Huang JY, Cheng YJ, Lin YP, Lin HC, Su CC, Juliano R, Yang BC. Extracellular matrix of glioblastoma inhibits polarization and transmigration of T cells: the role of tenascin-C in immune suppression. THE JOURNAL OF IMMUNOLOGY 2010; 185:1450-9. [PMID: 20622113 DOI: 10.4049/jimmunol.0901352] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Dense accumulations of T cells are often found in peritumoral areas, which reduce the efficiency of contact-dependent lysis of tumor cells. We demonstrate in this study that the extracellular matrix (ECM) produced by tumors can directly regulate T cell migration. The transmigration rate of several T cells including peripheral blood primary T cell, Jurkat, and Molt-4 measured for glioma cells or glioma ECM was consistently low. Jurkat cells showed reduced amoeba-like shape formation and delayed ERK activation when they were in contact with monolayers or ECM of glioma cells as compared with those in contact with HepG2 and MCF-7 cells. Phospho-ERK was located at the leading edge of migrating Jurkat cells. Glioma cells, but not MCF-7 and HepG2 cells, expressed tenascin-C. Knocking down the tenascin-C gene using the short hairpin RNA strategy converted glioma cells to a transmigration-permissive phenotype for Jurkat cells regarding ERK activation, transmigration, and amoeba-like shape formation. In addition, exogenous tenascin-C protein reduced the amoeba-like shape formation and transmigration of Jurkat cells through MCF-7 and HepG2 cell monolayers. A high level of tenascin-C was visualized immunohistochemically in glioma tumor tissues. CD3(+) T cells were detected in the boundary tumor area and stained strongly positive for tenascin-C. In summary, glioma cells can actively paralyze T cell migration by the expression of tenascin-C, representing a novel immune suppressive mechanism achieved through tumor ECM.
Collapse
Affiliation(s)
- Jyun-Yuan Huang
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | | | | | |
Collapse
|
22
|
Update on Antithrombin for the Treatment of Burn Trauma and Smoke Inhalation Injury. Intensive Care Med 2010. [DOI: 10.1007/978-1-4419-5562-3_27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
23
|
Shworak NW, Kobayashi T, de Agostini A, Smits NC. Anticoagulant heparan sulfate to not clot--or not? PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010; 93:153-78. [PMID: 20807645 DOI: 10.1016/s1877-1173(10)93008-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Vascular endothelial cells (ECs) produce anticoagulant heparan sulfate (HSAT+)-a small subpopulation of heparan sulfate (HS) containing a specific pentasaccharide motif with high affinity for plasma antithrombin (AT). This pentasaccharide is responsible for the anticoagulant action of therapeutic heparin, which dramatically catalyzes AT neutralization of coagulation proteases. Consequently, HSAT+ has been designated as "anticoagulant HS," and has long been thought to convey antithrombotic properties to the blood vessel wall. The Hs3st1 gene encodes HS 3-O-sulfotransferase-1, whose rate limiting action regulates EC production of HSAT+. To elucidate the biologic role of HSAT+, we generated Hs3st1-/- knock-out mice that have undetectable EC HSAT+. Despite long held historic expectations, hemostasis was unaffected in Hs3st1-/- mice. In light of this surprising finding, herein we evaluate historic, biochemical, kinetic, physiologic, and molecular genetic studies of AT, heparin, and HSAT+. We find that a hemostatic role for HSAT+ cannot presently be excluded; however, HSAT+ may well not be essential for AT's anticoagulant function. Specifically, in the absence of glycosaminoglycans, physiologic levels of AT can neutralize coagulation proteases at a sufficiently high throughput to account for most of AT's anticoagulant function. Moreover, at the vessel wall surface, glycosaminoglycans distinct from HSAT+ may be the predominant catalysts of AT's anticoagulant activity. We then explore the possibility that HSAT+ regulates a less well known function of AT, anti-inflammatory activity. We find that Hs3st1-/- mice exhibit a strong proinflammatory phenotype that is unresponsive to AT's anti-inflammatory activity. We conclude that the predominant function of HSAT+ is to mediate AT's anti-inflammatory activity.
Collapse
Affiliation(s)
- Nicholas W Shworak
- Department of Medicine, Dartmouth Medical School, Hanover, New Hampshire, USA
| | | | | | | |
Collapse
|
24
|
Kumarasuriyar A, Lee I, Nurcombe V, Cool SM. De-sulfation of MG-63 cell glycosaminoglycans delays in vitro osteogenesis, up-regulates cholesterol synthesis and disrupts cell cycle and the actin cytoskeleton. J Cell Physiol 2009; 219:572-83. [PMID: 19142873 DOI: 10.1002/jcp.21700] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glycosaminoglycan (GAG) sugars are largely responsible for the bioactivity of the proteoglycan proteins they decorate, and are particularly important for mediating the processes of cell attachment and growth factor signaling. Here, we show that chlorate-induced de-sulfation of GAGs expressed by MG-63 osteosarcoma cells results in delayed cell proliferation when the cells are exposed to chlorate for short or medium periods, but a disrupted mineralization without altered cell proliferation in response to long-term chlorate exposure. Analysis of GAG-binding growth factor activity indicated that chlorate disrupted BMP2/noggin signaling, but not FGF2 activity. Microarray analyses, which were confirmed by subsequent cell-based assays, indicated that chlorate predominantly disrupted the cell cycle and actin cytoskeleton and upregulated cholesterol synthesis, without affecting cell migration or attachment. Furthermore, we observed that disruption of the functions of the proteoglycan syndecan-4 replicated phenotypes induced by chlorate, implicating a primary role for this proteoglycan in providing bioactivity for these cells. J. Cell. Physiol. 219: 572-583, 2009. (c) 2009 Wiley-Liss, Inc.
Collapse
|
25
|
Rodgers KD, San Antonio JD, Jacenko O. Heparan sulfate proteoglycans: a GAGgle of skeletal-hematopoietic regulators. Dev Dyn 2008; 237:2622-42. [PMID: 18629873 DOI: 10.1002/dvdy.21593] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
This review summarizes our current understanding of the presence and function of heparan sulfate proteoglycans (HSPGs) in skeletal development and hematopoiesis. Although proteoglycans (PGs) comprise a large and diverse group of cell surface and matrix molecules, we chose to focus on HSPGs owing to their many proposed functions in skeletogenesis and hematopoiesis. Specifically, we discuss how HSPGs play predominant roles in establishing and regulating niches during skeleto-hematopoietic development by participating in distinct developmental processes such as patterning, compartmentalization, growth, differentiation, and maintenance of tissues. Special emphasis is placed on our novel hypothesis that mechanistically links endochondral skeletogenesis to the establishment of the hematopoietic stem cell (HSC) niche in the marrow. HSPGs may contribute to these developmental processes through their unique abilities to establish and mediate morphogen, growth factor, and cytokine gradients; facilitate signaling; provide structural stability to tissues; and act as molecular filters and barriers.
Collapse
Affiliation(s)
- Kathryn D Rodgers
- Department of Animal Biology, Division of Biochemistry, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania 19104-6046, USA.
| | | | | |
Collapse
|
26
|
Komura H, Uchiba M, Mizuochi Y, Arai M, Harada N, Katsuya H, Okajima K. Antithrombin inhibits lipopolysaccharide-induced tumor necrosis factor-alpha production by monocytes in vitro through inhibition of Egr-1 expression. J Thromb Haemost 2008; 6:499-507. [PMID: 18088351 DOI: 10.1111/j.1538-7836.2007.02869.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Antithrombin (AT) improves the outcome of septic patients with intravascular coagulation. However, the mechanisms underlying the therapeutic benefits of AT are not fully understood. Tumor necrosis factor-alpha (TNF-alpha) plays a critical role in the development of organ failure and intravascular coagulation in sepsis. AIM This study aimed to elucidate a molecular mechanism by which AT inhibits TNF-alpha production. METHODS Human peripheral monocyte was stimulated by lipopolysaccharide (LPS) and TNF-alpha concentration in media was measured. Levels of phosphorylation of extracellular signal-regulated protein kinases (ERK) 1/2 and early growth response factor-1 (Egr-1) were estimated by western blotting or by electrophoretic mobility shift assay. RESULTS Antithrombin (3 U mL(-1)) inhibited TNF-alpha production by monocytes stimulated with LPS. Conversely, chemically modified AT that lacks affinity for heparin did not. AT inhibited the phosphorylation of ERK 1/2 and decreased the expression of Egr-1 in LPS-stimulated monocytes. However, it did not affect the activation of either nuclear factor-kappaB or activator protein-1. Pretreatment with KT5720, a protein kinase A inhibitor, reversed the inhibitory effect of AT on the LPS-induced phosphorylation of ERK1/2. Although 2 U mL(-1) AT slightly inhibited TNF-alpha production by LPS-stimulated monocytes, it significantly inhibited TNF-alpha production in the presence of a low concentration of beraprost, a stable derivative of prostacyclin. CONCLUSIONS These observations suggest that AT might inhibit LPS-induced production of TNF-alpha by inhibiting the increase in Egr-1 expression in monocytes via interaction with heparin-like substances expressed on the cell surface.
Collapse
Affiliation(s)
- H Komura
- Department of Anaesthesiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Combined anticoagulants ameliorate acute lung injury in sheep after burn and smoke inhalation. Clin Sci (Lond) 2008; 114:321-9. [PMID: 17927568 DOI: 10.1042/cs20070254] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Burn and smoke inhalation-related multiple organ dysfunction is associated with a severe fall in the plasma concentration of antithrombin. Therefore the aim of the present study was to test the hypothesis that intravenous administration of recombinant human antithrombin in combination with aerosolized heparin will ameliorate acute lung injury in sheep exposed to cutaneous burn and smoke inhalation. Sheep were prepared operatively for study and, 7 days post-surgery, sheep were given a cutaneous burn (40% of total body surface area, third-degree burn) and insufflated with cotton smoke (48 breaths, <40 degrees C) under halothane anaesthesia. After injury, sheep were placed on a ventilator and resuscitated with Ringer's lactate solution. The animals were divided into three groups: sham group (non-injured and non-treated; n=6), saline group (injured and received saline; n=6) and rhAT.iv.+Hep group [injured and treated with rhAT (recombinant human antithrombin) and heparin; n=6]. In the rhAT.iv.+Hep group, rhAT was infused continuously for 48 h starting 1 h post-injury with a dose of 0.34 mg.h(-1).kg(-1) of body weight and heparin (10000 units) was aerosolized every 4 h starting at 1 h post-injury. The experiment lasted 48 h. Haemodynamics were stable in sham group, whereas the saline-treated sheep developed multiple signs of acute lung injury, including decreased pulmonary gas exchange, increased inspiratory pressures, extensive airway obstruction and increased pulmonary oedema. These pathological changes were associated with a severe fall in plasma antithrombin concentration, lung tissue accumulation of leucocytes and excessive production of NO. Treatment of injured sheep with anticoagulants attenuated all of the pulmonary pathophysiology observed. In conclusion, the results provide definitive evidence that anticoagulant therapy may be a novel and effective treatment tool in the management of burn patients with concomitant smoke inhalation injury.
Collapse
|
28
|
Importance of Airway Management in Burn and Smoke Inhalation-induced Acute Lung Injury. Intensive Care Med 2007. [DOI: 10.1007/0-387-35096-9_47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
29
|
Feistritzer C, Wiedermann CJ. Effects of anticoagulant strategies on activation of inflammation and coagulation. Expert Opin Biol Ther 2007; 7:855-70. [PMID: 17555371 DOI: 10.1517/14712598.7.6.855] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Acute inflammatory events, such as those that occur in sepsis, lead to dysregulation of the coagulation cascade. The hemostatic imbalance in sepsis, characterized by the excessive activation of procoagulant pathways and the impairment of anticoagulant activity, leads to disseminated intravascular coagulation and results in microvascular thrombosis, tissue hypoperfusion and, ultimately, multiple organ failure and death. Furthermore, natural anti-inflammatory mechanisms of the endogenous anticoagulants are diminished by the impaired coagulation. Supportive strategies aiming at inhibiting activation of coagulation and inflammation by treatment with exogenous anticoagulants have been found to be beneficial in experimental and initial clinical studies. This review summarizes the available experimental and clinical data regarding the interaction between coagulation and inflammation, focusing on the two anticoagulants which are in clinical use, antithrombin and activated protein C. Identification of the different biological mechanisms of the two endogenous anticoagulants might help to determine target patient populations as well as to develop new anticoagulant analogs that differ in there respective effects in coagulation and inflammation.
Collapse
Affiliation(s)
- Clemens Feistritzer
- Medical University of Innsbruck, Department of Internal Medicine, Innsbruck (Tyrol), Austria
| | | |
Collapse
|
30
|
Pakula R, Melchior A, Denys A, Vanpouille C, Mazurier J, Allain F. Syndecan-1/CD147 association is essential for cyclophilin B-induced activation of p44/42 mitogen-activated protein kinases and promotion of cell adhesion and chemotaxis. Glycobiology 2007; 17:492-503. [PMID: 17267519 DOI: 10.1093/glycob/cwm009] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Many of the biological functions attributed to cell surface proteoglycans are dependent on the interaction with extracellular mediators through their heparan sulphate (HS) moieties and the participation of their core proteins in signaling events. A class of recently identified inflammatory mediators is secreted cyclophilins, which are mostly known as cyclosporin A-binding proteins. We previously demonstrated that cyclophilin B (CyPB) triggers chemotaxis and integrin-mediated adhesion of T lymphocytes mainly of the CD4+/CD45RO+ phenotype. These activities are related to interactions with two types of binding sites, CD147 and cell surface HS. Here, we demonstrate that CyPB-mediated adhesion of CD4+/CD45RO+ T cells is related to p44/42 mitogen-activated protein kinase (MAPK) activation by a mechanism involving CD147 and HS proteoglycans (HSPG). Although HSPG core proteins are represented by syndecan-1, -2, -4, CD44v3 and betaglycan in CD4+/CD45RO+ T cells, we found that only syndecan-1 is physically associated with CD147. The intensity of the heterocomplex increased in response to CyPB, suggesting a transient enhancement and/or stabilization in the association of CD147 to syndecan-1. Pretreatment with anti-syndecan-1 antibodies or knockdown of syndecan-1 expression by RNA interference dramatically reduced CyPB-induced p44/p42 MAPK activation and consequent migration and adhesion, supporting the model in which syndecan-1 serves as a binding subunit to form the fully active receptor of CyPB. Altogether, our findings provide a novel example of a soluble mediator in which a member of the syndecan family plays a critical role in efficient interaction with signaling receptors and initiation of cellular responses.
Collapse
Affiliation(s)
- Rachel Pakula
- Laboratory of Molecular and Cellular Biophysics, National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
The polysaccharide heparan sulphate is ubiquitously expressed as a proteoglycan in extracellular matrices and on cell surfaces. Heparan sulphate has marked sequence diversity that allows it to specifically interact with many proteins. This Review focuses on the multiple roles of heparan sulphate in inflammatory responses and, in particular, on its participation in almost every stage of leukocyte transmigration through the blood-vessel wall. Heparan sulphate is involved in the initial adhesion of leukocytes to the inflamed endothelium, the subsequent chemokine-mediated transmigration through the vessel wall and the establishment of both acute and chronic inflammatory reactions.
Collapse
Affiliation(s)
- Christopher R Parish
- Division of Immunology and Genetics, John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia.
| |
Collapse
|
32
|
Rinder CS, Rinder HM, Smith MJ, Fitch JCK, Tracey JB, Chandler WL, Rollins SA, Smith BR. Antithrombin reduces monocyte and neutrophil CD11b up regulation in addition to blocking platelet activation during extracorporeal circulation. Transfusion 2006; 46:1130-7. [PMID: 16836559 DOI: 10.1111/j.1537-2995.2006.00861.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Patients undergoing cardiac surgery requiring cardiopulmonary bypass develop a systemic inflammatory reaction. Antithrombin III (AT) has anticoagulant effects but also shows evidence of anti-inflammatory activity. The aim of this study was to examine whether exogenous AT could reduce white blood cell activation (CD11b up regulation or elastase release), in addition to inhibiting platelet (PLT) activation and fibrin generation, during simulated cardiopulmonary bypass (sCPB), undertaken in the absence of endothelium. STUDY DESIGN AND METHODS sCPB was carried out with minimally heparinized (2 U/mL) human blood for 90 minutes in controls and with supplementation by low-dose (1 U/mL) and high-dose (5 U/mL) AT. RESULTS High-dose AT blunted thrombin generation during sCPB (prothrombin fragment 1.2); both doses significantly inhibited thrombin activity (fibrinopeptide A). Complement activation (C3a and C5b-9) was unaffected by AT. High-dose AT inhibited PLT activation (P-selectin expression and P-selectin-dependent monocyte-PLT conjugate formation). AT supplementation at the higher dose significantly abrogated monocyte and neutrophil CD11b up regulation and neutrophil elastase release. CONCLUSION In addition to anticoagulant and anti-PLT effects, pharmacologic AT doses significantly blunted monocyte and neutrophil CD11b up regulation and neutrophil elastase release during sCPB, independent of endothelial effects. These data provide evidence for the direct anti-inflammatory activity of AT that has clinical relevance for CPB complications.
Collapse
Affiliation(s)
- Christine S Rinder
- Department of Anesthesiology, Yale University School of Medicine, New Haven, Connecticut 06520-8051, USA.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Borozan I, Chen L, Sun J, Tannis LL, Guindi M, Rotstein OD, Heathcote J, Edwards AM, Grant D, McGilvray ID. Gene expression profiling of acute liver stress during living donor liver transplantation. Am J Transplant 2006; 6:806-24. [PMID: 16539639 DOI: 10.1111/j.1600-6143.2006.01254.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
During liver transplantation, the donor graft is subjected to a number of acute stresses whose molecular basis is not well-understood. The effects of surgical stress, preservation and reperfusion injury were studied in 24 consecutive living donor liver transplant (LDLT) operations. Liver biopsies were taken early in the donor operation (OPENING), after transection of the donor liver (PRECLAMP) and following implantation of the graft (post hepatic artery, [PHA]); these were evaluated for histology, tissue glutathione content and gene expression using a 19K-human cDNA microarray. LDLT was associated with an ischemia/reperfusion injury, with accumulation of small numbers of neutrophils and decreased glutathione in the PHA biopsies. Following reperfusion, the expression of 129 genes increased and 106 genes decreased when compared to OPENING levels (> or <2-fold, p < 0.01). By real-time PCR a subset of 25 genes was verified (15 increased, 10 decreased). These genes were similarly altered in another condition of acute liver stress (the response to brain-death), but not in three chronic liver disease states (HCV, HBV and PBC). This study has identified a set of genes whose expression is altered in acute, but not chronic, liver stress, likely to play a central role in the pathogenesis of acute liver injury of liver transplantation.
Collapse
Affiliation(s)
- I Borozan
- Banting and Best Department of Medical Research, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Mana M, Cole M, Cox S, Tawil B. Human U937 monocyte behavior and protein expression on various formulations of three-dimensional fibrin clots. Wound Repair Regen 2006. [DOI: 10.1111/j.1524-475x.2005.00091.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
35
|
Jalil J, Lavandero S, Chiong M, Ocaranza MP. [Rho/Rho kinase signal transduction pathway in cardiovascular disease and cardiovascular remodeling]. Rev Esp Cardiol 2005. [PMID: 16053829 DOI: 10.1157/13078132] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The small guanosine triphosphatase Rho and its target, Rho kinase, play important roles in both blood pressure regulation and vascular smooth muscle contraction. Rho is activated by agonists of receptors coupled to cell membrane G protein, such as angiotensin II and phenylephrine. Once Rho is activated, it translocates to the cell membrane where it, in turn, activates Rho kinase. Activated Rho kinase phosphorylates myosin light chain phosphatase, which is then inhibited. This sequence stimulates vascular smooth muscle contraction, stress fiber formation,and cell migration. In this way, Rho and Rho kinase activation have important effects on several cardiovascular diseases. Currently available substances that specifically inhibit this signaling pathway could offer clinical benefits in several cardiovascular, as well as noncardiovascular diseases, such as arterial hypertension, pulmonary hypertension, cerebral or coronary spasm, post-angioplasty restenosis, and erectile dysfunction.
Collapse
Affiliation(s)
- Jorge Jalil
- Departamento de Enfermedades Cardiovasculares, Hospital Clínico, Pontificia Universidad Católica de Chile, Chile.
| | | | | | | |
Collapse
|
36
|
Saito H, Minamiya Y, Kalina U, Saito S, Ogawa JI. Effect of antithrombin III on neutrophil deformability. J Leukoc Biol 2005; 78:777-84. [PMID: 16000388 DOI: 10.1189/jlb.1004589] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
As the spherical diameter of pulmonary capillaries is smaller than that of neutrophils, increased neutrophil stiffness or conversely, decreased neutrophil deformability is a key step in the initial sequestration of neutrophils within the lungs during inflammatory processes. Antithrombin III (AT) is known to exert a therapeutic effect against disseminated intravascular coagulation, and accumulating evidence suggests that AT also has anti-inflammatory properties. The mechanisms of its anti-inflammatory effects remain unclear, but in a rat endotoxin model, AT apparently inhibited neutrophil sequestration in the lung. In the present in vitro study, therefore, we examined the effect of AT on the deformability of human neutrophils and correlated those findings with their F-actin content. Isolated human neutrophils were stimulated with formyl-Met-Leu-Phe (1 muM, 2 min) in the presence or absence of the alpha, beta, or low heparin-affinity isoforms of AT (1 IU/ml, 20 min), and deformability was evaluated using a filter assay system. Neutrophils were also stained with fluorescein isothiocyanate-phalloidin and subjected to a fluorescein-activated cell sorter scan to assess F-actin content. The results showed that pretreatment with any of the three AT isoforms similarly inhibited the decreased neutrophil deformability and increased F-actin content of stimulated cells. Notably, heparinase had no effect on deformability or F-actin content in the presence or absence of AT, which was somewhat unexpected, as heparin sulfate proteoglycans likely function as AT receptors. These findings suggested that AT inhibits the increase in neutrophil stiffness seen during inflammatory processes by inhibiting actin polymerization via a heparin-independent pathway.
Collapse
Affiliation(s)
- Hajime Saito
- Division of Thoracic Surgery, Akita University School of Medicine, 1-1-1 Hondo, Akita City 010-8543, Japan.
| | | | | | | | | |
Collapse
|
37
|
Liu CY, Battaglia M, Lee SH, Sun QH, Aster RH, Visentin GP. Platelet factor 4 differentially modulates CD4+CD25+ (regulatory) versus CD4+CD25- (nonregulatory) T cells. THE JOURNAL OF IMMUNOLOGY 2005; 174:2680-6. [PMID: 15728475 DOI: 10.4049/jimmunol.174.5.2680] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Active suppression mediated by CD4(+)CD25(+) T regulatory (Tr) cells plays an important role in the down-regulation of T cell responses to both foreign and self-Ags. Platelet factor 4 (PF4), a platelet-derived CXC chemokine, has been shown to strongly inhibit T cell proliferation as well as IFN-gamma and IL-2 release by isolated T cells. In this report we show that human PF4 stimulates proliferation of the naturally anergic human CD4(+)CD25(+) Tr cells while inhibiting proliferation of CD4(+)CD25(-) T cells. In coculture experiments we found that CD4(+)CD25(+) Tr cells exposed to PF4 lose the ability to inhibit the proliferative response of CD4(+)CD25(-) T cells. Our findings suggest that human PF4, by inducing Tr cell proliferation while impairing Tr cell function, may play a previously unrecognized role in the regulation of human immune responses. Because platelets are the sole source of PF4 in the circulation, these findings may be relevant to the pathogenesis of certain immune-mediated disorders associated with platelet activation, such as heparin-induced thrombocytopenia and autoimmune thrombocytopenic purpura.
Collapse
Affiliation(s)
- Chao Yan Liu
- Blood Research Institute, The Blood Center of Southeastern Wisconsin, Milwaukee, WI 53226, USA
| | | | | | | | | | | |
Collapse
|
38
|
Gritti D, Malinverno A, Gasparetto C, Wiedermann CJ, Ricevuti R. Attenuation of leukocyte beta 2-integrin expression by antithrombin-III. Int J Immunopathol Pharmacol 2004; 17:27-32. [PMID: 15000863 DOI: 10.1177/039463200401700104] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Antithrombin-III exerts antiinflammatory effects via ligation of heparan sulfate proteoglycans. Here we show in vitro that recombinant human antithrombin-III attenuates CD11b/CD18 expression of activated neutrophils and monocytes in whole blood ex vivo. As leukocyte integrin expression is triggered by extracorporeal circulation, this observation may be of relevance for pharmacological treatment during cardiopulmonary bypass.
Collapse
Affiliation(s)
- D Gritti
- Department of Internal Medicine and Therapeutics, IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | | | | | | | | |
Collapse
|
39
|
Rops ALWMM, van der Vlag J, Lensen JFM, Wijnhoven TJM, van den Heuvel LPWJ, van Kuppevelt TH, Berden JHM. Heparan sulfate proteoglycans in glomerular inflammation. Kidney Int 2004; 65:768-85. [PMID: 14871397 DOI: 10.1111/j.1523-1755.2004.00451.x] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Heparan sulfate proteoglycans (HSPGs) are glycoproteins consisting of a core protein to which linear heparan sulfate side chains are covalently attached. These heparan sulfate side chains can be modified at different positions by several enzymes, which include N-deacetylases, N- and O-sulfotransferases, and an epimerase. These heparan sulfate modifications give rise to an enormous structural diversity, which corresponds to the variety of biologic functions mediated by heparan sulfate, including its role in inflammation. The HSPGs in the glomerular basement membrane (GBM), perlecan, agrin, and collagen XVIII, play an important role in the charge-selective permeability of the glomerular filter. In addition to these HSPGs, various cell types express HSPGs at their cell surface, which include syndecans, glypicans, CD44, and betaglycan. During inflammation, HSPGs, especially heparan sulfate, in the extracellular matrix (ECM) and at the surface of endothelial cells bind chemokines, which establishes a local concentration gradient recruiting leukocytes. Endothelial and leukocyte cell surface HSPGs also play a role in their direct adhesive interactions via other cell surface adhesion molecules, such as selectins and beta2 integrin. Activated leukocytes and endothelial cells exert heparanase activity, resulting in degradation of heparan sulfate moieties in the ECM, which facilitates leukocyte passage into tissues and the release of heparan sulfate-bound factors. In various renal inflammatory diseases the expression of agrin and GBM-associated heparan sulfate is decreased, while the expression of CD44 is increased. Heparan sulfate or heparin preparations affect inflammatory cell behavior and have promising therapeutic, anti-inflammatory properties by preventing leukocyte adhesion/influx and tissue damage.
Collapse
Affiliation(s)
- Angelique L W M M Rops
- Nephrology Research Laboratory, Nijmegen Centre for Molecular Life Sciences, University Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
40
|
|
41
|
Trocho C, Escolà-Gil JC, Ribas V, Benítez S, Martín-Campos JM, Rotllan N, Osaba L, Ordóñez-Llanos J, González-Sastre F, Blanco-Vaca F. Phenytoin treatment reduces atherosclerosis in mice through mechanisms independent of plasma HDL-cholesterol concentration. Atherosclerosis 2004; 174:275-85. [PMID: 15136057 DOI: 10.1016/j.atherosclerosis.2004.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2003] [Revised: 02/18/2004] [Accepted: 02/25/2004] [Indexed: 10/26/2022]
Abstract
Phenytoin (PHT) increases high density lipoprotein cholesterol (HDL-C) and reduces coronary artery disease mortality in humans. We report the results of PHT treatment on atherosclerosis susceptibility and lipid profile in four different types of mouse: control C57BL/6 mice and cholesteryl ester transfer protein transgenic mice as models of fatty streak, and LDL receptor-deficient mice and apolipoprotein E-deficient mice as models of mature atherosclerosis. Each mouse type was fed an appropriate diet to induce atherosclerosis and prevent liver toxicity. PHT treatment demonstrated a protective effect in all models. Reduction in aortic atherosclerotic area by PHT treatment was more evident in early atherosclerosis (2.3-fold) than in mature atherosclerosis (decreases of 40 and 23%, respectively, but only in mice in the upper 50% percentile of plasma PHT concentration). Atherosclerosis prevention was not concomitant with a consistent increase in HDL-C or any other protective change in the lipid profile. Different analyses of potential antiatherogenic HDL functions did not provide additional information. Microarray liver gene expression analyses identified a potential atheroprotective mechanism characterized by decreased expression of syndecan-4, RhoA2, double LIM protein-1, zeta-chain-associated protein kinase-70 and interleukin 6 receptor-alpha. However, to demonstrate that these changes are part of a PHT-antiatherogenic effect, they will need to be found also in arteries, maintained at protein level and proved to be causal rather than reactive.
Collapse
Affiliation(s)
- Carme Trocho
- Servei de Bioquímica, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Feistritzer C, Kaneider NC, Sturn DH, Wiedermann CJ. Syndecan-4-dependent migration of human eosinophils. Clin Exp Allergy 2004; 34:696-703. [PMID: 15144459 DOI: 10.1111/j.1365-2222.2004.1853.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Heparan sulphate proteoglycans (HSPGs) are important participants in cell surface signalling and critical in controlling cell behaviour. They modulate inflammatory cell maturation and activation, leucocyte rolling, adhesion to endothelium as well as extravasation and chemotaxis. Whether eosinophil's function is affected has not yet been reported. OBJECTIVE We investigated the effects of transgenic, recombinant anti-thrombin III and Kybernin P, an anti-thrombin III concentrate, as HSPG ligands on spontaneous and chemokine-triggered migration of normal eosinophils from human peripheral blood in modified Boyden chamber micropore filter assays. METHODS Eosinophils from human peripheral blood were purified using magnetic antibody cell sorting. The signalling mechanisms required for anti-thrombin-dependent migration were studied using signalling enzyme blockers. Expression of HSPG core protein mRNA was studied by PCR. RESULTS Pre-treatment of eosinophils with anti-thrombin III inhibited chemotaxis toward optimal concentrations of eotaxin or RANTES (regulated upon activation normal T cell expressed and activated). In the absence of the chemokines, direct exposure to gradients of anti-thrombin III stimulated eosinophil migration. The effects of anti-thrombin III were abolished by pre-treating cells with heparinase-1, chondroitinase, sodium chlorate and anti-syndecan-4 antibodies. Syndecan-4 gene expression in eosinophils was confirmed in PCR. In the presence of pentasaccharide, anti-thrombin III lost its effect on the cells. Functional responses were also abrogated by inhibition of protein kinase C, phosphatidylinositol 3-kinase and phosphodiesterase. CONCLUSION Data indicate that anti-thrombin III affects eosinophil motility via the effects of its heparin-binding site on cell surface syndecan-4. Ligation of syndecan-4 with anti-thrombin III induces eosinophil migration and deactivates motility toward chemokines. These observations suggest that syndecan-4-dependent signalling may control eosinophil locomotion.
Collapse
Affiliation(s)
- C Feistritzer
- Division of General Internal Medicine, Department of Internal Medicine, University of Innsbruck, Innsbruck, Austria
| | | | | | | |
Collapse
|
43
|
Kaneider NC, Dunzendorfer S, Wiedermann CJ. Heparan Sulfate Proteoglycans Are Involved in Opiate Receptor-Mediated Cell Migration. Biochemistry 2003; 43:237-44. [PMID: 14705951 DOI: 10.1021/bi035295i] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Opioid receptors are expressed in cells of the immune system, and potent immunomodulatory effects of their natural and synthetic ligands have been reported. In some studies, the opiate receptor antagonist naloxone itself displayed immunomodulatory actions. We investigated effects of naloxone on leukocyte chemotaxis. Cell migration was tested in micropore filter assays using modified Boyden chambers, and receptor expression was investigated using radiolabel binding assays. Naloxone induced peripheral blood nonadherent mononuclear cell and neutrophil chemotaxis at nanomolar concentrations and deactivated their migration toward beta-endorphin, angiotensin II, somatostatin, or interleukin-8 but not toward RANTES, vasoactive intestinal peptide, or substance P. Ligand binding studies showed no alteration in the binding of interleukin-8 to neutrophils by naloxone. Cleavage of heparan sulfate from proteoglycans on the cells' surface completely inhibited chemotactic and deactivating properties of naloxone but not other attractants. Chemotactic properties were abolished by pretreating cells with heparinase, chondroitinase, sodium chlorate, and anti-syndecan-4 antibodies, indicating the involvement of syndecan-4. The extent of migration toward naloxone was diminished by pretreatment with dimethylsphingosine, a specific sphingosine kinase inhibitor. As syndecan-4 signaling in leukocyte chemotaxis involves activation of sphingosine kinase, results indicate that naloxone interacts with syndecan-4 function in cell migration and suggest a role for heparan sulfate proteoglycans as coreceptors to members of the delta-opiate receptor family.
Collapse
Affiliation(s)
- Nicole C Kaneider
- Division of General Internal Medicine, Department of Internal Medicine, University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | | | | |
Collapse
|
44
|
Djanani A, Kaneider NC, Sturn D, Wiedermann CJ. Agonist function of the neurokinin receptor antagonist, [D-Arg1,D-Phe5,D-Trp7,9,Leu11]substance P, in monocytes. REGULATORY PEPTIDES 2003; 115:123-9. [PMID: 12972327 DOI: 10.1016/s0167-0115(03)00148-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
G-protein-coupled bombesin receptors are capable of signaling through the G(i) protein even when receptor-coupling to G(q) is blocked by [D-Arg1,D-Phe5,D-Trp7,9,Leu11]substance P (SpD), a neurokinin-1 receptor antagonist and "biased" agonist to bombesin receptors. As bombesin is a monocyte and tumor cell attractant, we were interested in the effects of SpD on cell migration. Chemotaxis of monocytes was tested in micropore filter assays. SpD was a dose-dependent agonist in monocyte migration and was not inhibited by antagonists to neurokinin-1 or -2 receptors. SpD failed to inhibit chemotaxis toward bombesin, suggesting that inhibition of bombesin receptor coupling to G(q) with SpD does not impair migratory responses elicited by bombesin. As pertussis toxin inhibited migration, coupling of receptors to G(i) may signal migration. Chemotaxis toward SpD was inhibited by bombesin receptor antagonists as well as by blocking signaling enzymes downstream of G(q) (phospholipase-3 and protein kinase C with wortmannin and bisindolylmaleimide, respectively), suggesting transactivation of G(q)-mediated chemotaxis signaling by SpD via bombesin receptors. Protein kinase C that induces sphingosine kinase activation and production of sphingosine-1-phosphate, which may lead to G(q)-dependent chemoattraction, was involved in SpD-dependent migration. Inhibition of sphingosine-1-phosphate production with dimethylsphingosine inhibited monocyte migration toward SpD. Data suggest that SpD induces migration in monocytes and signaling events involving activation of sphingosine kinase in a G(i) protein- and protein kinase C-dependent fashion. "Biased" agonism of SpD at bombesin receptors may affect normal and tumor cell migration.
Collapse
Affiliation(s)
- Angela Djanani
- Department of Internal Medicine, Division of General Internal Medicine, University of Innsbruck, Anichstrasse 35, A-6020, Innsbruck, Austria
| | | | | | | |
Collapse
|
45
|
Kaneider NC, Egger P, Djanani AM, Wiedermann CJ. Leukocyte motility in response to neuropeptides is heparan sulfate proteoglycan dependent. Peptides 2003; 24:695-700. [PMID: 12895655 DOI: 10.1016/s0196-9781(03)00129-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Activation of neuropeptide receptors on leukocytes induces chemotaxis. We determined in Boyden chambers with micropore filters, whether in human monocytes and lymphocytes this migratory response is heparan sulfate proteoglycan (HSPG) dependent. Chemotaxis toward calcitonin gene-related peptide, secretoneurin, vasoactive intestinal peptide (VIP), and substance P (SP) was abolished by removal of heparan sulfate side chains from cell surface proteoglycans or by addition of anti-syndecan-4 antibodies. Inhibition of neuropeptide-induced chemotaxis by dimethyl sphingosine (DMS), an inhibitor of sphingosine kinase, indicates transactivation of the sphingosine-1-phosphate chemotaxis pathway which was previously identified as being syndecan-4-related. Data suggest that HSPGs are involved in neuropeptide-induced chemotaxis of leukocytes.
Collapse
Affiliation(s)
- Nicole C Kaneider
- Division of General Internal Medicine, Department of Internal Medicine, University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | | | | | | |
Collapse
|
46
|
Abstract
Cell surface heparan sulfate (HS) influences a multitude of molecules, cell types, and processes relevant to inflammation. HS binds to cell surface and matrix proteins, cytokines, and chemokines. These interactions modulate inflammatory cell maturation and activation, leukocyte rolling, and tight adhesion to endothelium, as well as extravasation and chemotaxis. The syndecan family of transmembrane proteoglycans is the major source of cell surface HS on all cell types. Recent in vitro and in vivo data suggest the involvement of syndecans in the modulation of leukocyte-endothelial interactions and extravasation, the formation of chemokine and kininogen gradients, participation in chemokine and growth factor signaling, as well as repair processes. Thus, the complex role of HS in inflammation is reflected by multiple functions of its physiological carriers, the syndecans. Individual and common functions of the four mammalian syndecan family members can be distinguished. Recently generated transgenic and knockout mouse models will facilitate analysis of the individual processes that each syndecan is involved in.
Collapse
|
47
|
Högerkorp CM, Bilke S, Breslin T, Ingvarsson S, Borrebaeck CAK. CD44-stimulated human B cells express transcripts specifically involved in immunomodulation and inflammation as analyzed by DNA microarrays. Blood 2003; 101:2307-13. [PMID: 12411303 DOI: 10.1182/blood-2002-06-1837] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A number of studies have implicated a role for the cell surface glycoprotein CD44 in several biologic events, such as lymphopoiesis, homing, lymphocyte activation, and apoptosis. We have earlier reported that signaling via CD44 on naive B cells in addition to B-cell receptor (BCR) and CD40 engagement generated a germinal center-like phenotype. To further characterize the global role of CD44 in B differentiation, we examined the expression profile of human B cells cultured in vitro in the presence or absence of CD44 ligation, together with anti-immunoglobulin (anti-Ig) and anti-CD40 antibodies. The data sets derived from DNA microarrays were analyzed using a novel statistical analysis scheme created to retrieve the most likely expression pattern of CD44 ligation. Our results show that genes such as interleukin-6 (IL-6), IL-1alpha, and beta(2)-adrenergic receptor (beta(2)-AR) were specifically up-regulated by CD44 ligation, suggesting a novel role for CD44 in immunoregulation and inflammation.
Collapse
|
48
|
Endogenous Anticoagulants and the Role of Heparin in the Treatment of Severe Sepsis. Intensive Care Med 2003. [DOI: 10.1007/978-1-4757-5548-0_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
49
|
Shirotani-Ikejima H, Kokame K, Hamuro T, Bu G, Kato H, Miyata T. Tissue factor pathway inhibitor induces expression of JUNB and GADD45B mRNAs. Biochem Biophys Res Commun 2002; 299:847-52. [PMID: 12470656 DOI: 10.1016/s0006-291x(02)02759-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Tissue factor pathway inhibitor (TFPI) is a Kunitz-type protease inhibitor that regulates tissue factor-triggered blood coagulation. It has previously been reported that TFPI inhibits the proliferation of human umbilical vein endothelial cells (HUVECs), suggesting that TFPI may act as more than just a mediator of coagulation through changes in gene expression. By using DNA-array techniques and Northern blot analysis, we here revealed that TFPI transiently induced the mRNA expression of JUNB and GADD45B. The inducible effects were not observed in TFPIdeltaC (lacking the C-terminal basic region) or antithrombin (heparin-binding anticoagulant protease inhibitor). Moreover, the TFPI-induced expression of GADD45B was blocked by receptor-associated protein, which masks the ligand-binding domain of very low density lipoprotein receptor (VLDL-R). In conclusion, this is the first report to show an effect of TFPI on mRNA expression, and suggests that TFPI modulates cellular functions by inducing JUNB and GADD45B expression through binding to VLDL-R.
Collapse
Affiliation(s)
- Hiroko Shirotani-Ikejima
- National Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | | | | | | | | | | |
Collapse
|
50
|
Roemisch J, Gray E, Hoffmann JN, Wiedermann CJ. Antithrombin: a new look at the actions of a serine protease inhibitor. Blood Coagul Fibrinolysis 2002; 13:657-70. [PMID: 12441904 DOI: 10.1097/00001721-200212000-00001] [Citation(s) in RCA: 149] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Antithrombin (AT) is a plasma-derived, single-chain glycoprotein with a molecular weight of 58 kDa. It is a serine protease inhibitor (serpin), sharing about 30% homology in amino acid sequence with other serpins. AT is a complex molecule with multiple biologically important properties. It is a potent anticoagulant that has been demonstrated to provide benefit in animal models and small cohorts of patients with coagulation disorders. AT also has remarkable anti-inflammatory properties, several of which result from its actions in the coagulation cascade. Activated coagulation proteases like activated factor X and thrombin contribute to inflammation; for instance, by the release of pro-inflammatory mediators. Inhibition of these proteases by AT prevents their specific interaction with cells and subsequent reactions. Anti-inflammatory properties of AT independent of coagulation involve direct interactions with cells leading to the release of, for instance, prostacyclin. Binding of AT to a recently identified cellular receptor, syndecan-4, leads to the interference with the intracellular signal induced by mediators like lipopolysaccharides and, thereby, to a down-modulation of the inflammatory response. AT has been shown to be effective in prospective and well-controlled small-scale studies of patients with inflammatory conditions, including sepsis. Although AT did not decrease overall patient mortality in a double-blind, placebo-controlled, phase III trial of patients with sepsis, it is important to note that AT improved the survival of individuals in this study not receiving heparin as a prophylactic regimen, which can be explained by the impaired interaction of AT with its cellular receptor in the presence of heparin, resulting in the reduction of the anti-inflammatory properties. Accordingly, the supplementation of AT without concomitant heparin may be beneficial in disorders with inflammatory characteristics, which has to be demonstrated in further clinical studies. Finally, recent results suggest that latent AT can induce apoptosis of endothelial cells by disrupting cell-matrix interactions. Further investigations will have to demonstrate whether latent and/or cleaved AT are physiological means to control angiogenesis. A potential prophylactic or therapeutic use as an anti-angiogenic and antitumor agent merits further exploration, including whether the growth of vessels in tumor tissues or close to tumors can be controlled by latent AT without affecting the formation of blood vessels during wound healing processes.
Collapse
Affiliation(s)
- J Roemisch
- Aventis Behring GmbH, Research, Marburg, Germany
| | | | | | | |
Collapse
|