1
|
Ku CJ, Yu X, Zhao QY, Grzegorski SJ, Daniel JG, Ferguson AC, Shavit JA. Loss of protein C vs protein S results in discrepant thrombotic phenotypes. Blood Adv 2025; 9:545-557. [PMID: 39657127 PMCID: PMC11821410 DOI: 10.1182/bloodadvances.2024013237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 10/25/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024] Open
Abstract
ABSTRACT Venous thrombosis is a leading cause of morbidity/mortality and associated with deficiencies of the anticoagulant protein C (PC; PROC) and its cofactor, protein S (PS; PROS1). Heterozygous mutations increase the risk of adult-onset thrombosis, whereas homozygous mutations result in pre/neonatal lethal thrombosis. Phenotypes of patients with PC and PS deficiency are generally considered clinically indistinguishable. Here, we generate proc (zebrafish PROC ortholog) and pros1 knockouts through genome editing in zebrafish and uncover partially discordant phenotypes. proc-/- mutants exhibited ∼70% lethality at 1 year of age, whereas pros1-/- survival was unaffected. Induced venous endothelial injury in both mutants revealed reduced occlusive thrombus formation. This is consistent with the consumptive coagulopathy of zebrafish antithrombin 3 knockouts, which also results in spontaneous venous thrombosis. However, proc and pros1 mutants revealed a discrepancy. Although both mutants demonstrated spontaneous thrombosis, proc-/- was localized to the cardiac and venous systems, whereas pros1-/- was intracardiac. Aside from coagulation, PC has been shown to have PS-independent roles in inflammation. proc mutants displayed altered inflammatory markers and defects in neutrophil migration independent of pros1. Transcriptomic analysis and gene knockdown identified novel proc genetic interactions with adgrf7, a G protein-coupled receptor (GPCR) not previously known to be involved in coagulation. In summary, our data reveal differences between PC- and PS-deficient thrombosis, with cardiovascular tissue-specific phenotypes and survival differences, suggesting the possibility of underlying clinical differences in affected patients. This model of complete proc-/- deficiency in an accessible organism will facilitate further in vivo study of these distinctions, as well as PS-dependent and -independent functions of PC.
Collapse
Affiliation(s)
- Chia-Jui Ku
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Xinge Yu
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Queena Y. Zhao
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | | | | | | | - Jordan A. Shavit
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
- Department of Human Genetics, University of Michigan, Ann Arbor, MI
| |
Collapse
|
2
|
Sharma D, Thomas S, Moody TB, Taylor M, Ituarte B, Georgeson CJ, Barrett CD, Wei EX. Laboratory and clinical haemostatic aberrations in primary dermatologic disease: A review. Thromb J 2024; 22:101. [PMID: 39533305 PMCID: PMC11558853 DOI: 10.1186/s12959-024-00665-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Inflammatory dermatologic diseases have long been viewed as a "skin limited" disease process. Current literature on inflammatory dermatologic diseases investigates their relationship and influence on thromboembolic states and thromboembolic complications and the understanding of their pathophysiology and molecular mechanisms.Studies specifically discuss known inflammatory skin diseases including alopecia areata, vitiligo, psoriasis, hidradenitis suppurativa, atopic dermatitis, chronic spontaneous urticaria, and autoimmune bullous diseases, and their effects on systemic inflammation, associated cardiovascular comorbidities, and thromboembolic or hypercoagulable states. The limited current literature shows potential for links between inflammatory skin diseases and hypercoagulable states. Biomarkers such as F1 + 2, D-dimer, eosinophilic cationic protein, and PAI-1 are currently being studied to outline the mechanisms connecting inflammatory skin disease to the coagulation system. Further study and larger amounts of data are needed to draw definitive conclusions, especially when interpreting biomarkers alone such as PAI-1.The mechanisms, rates of systemic inflammation, and clinical outcomes of traditionally "skin limited" inflammatory diseases remain chronically understudied in dermatology. Many organ systems have well established connections between inflammatory disease and hypercoagulable states, but there are significant gaps in the literature regarding skin diseases. There is a significant need for comprehensive investigation of molecular mechanisms behind inflammatory dermatologic disease and hypercoagulability, how hypercoagulability effects clinical outcomes, and proper intervention to optimize patient outcomes.
Collapse
Affiliation(s)
- Divya Sharma
- Department of Dermatology, University of Nebraska Medical Center, 985645, Omaha, NE, 68198-5645, USA
| | - Sierra Thomas
- Department of Dermatology, University of Nebraska Medical Center, 985645, Omaha, NE, 68198-5645, USA
- University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Trace B Moody
- Division of Acute Care Surgery, Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mitchell Taylor
- Department of Dermatology, University of Nebraska Medical Center, 985645, Omaha, NE, 68198-5645, USA
- Creighton University School of Medicine, Omaha, NE, USA
| | - Bianca Ituarte
- Department of Dermatology, University of Nebraska Medical Center, 985645, Omaha, NE, 68198-5645, USA
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Corey J Georgeson
- Department of Dermatology, University of Nebraska Medical Center, 985645, Omaha, NE, 68198-5645, USA
| | - Christopher D Barrett
- Division of Acute Care Surgery, Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Erin X Wei
- Department of Dermatology, University of Nebraska Medical Center, 985645, Omaha, NE, 68198-5645, USA
| |
Collapse
|
3
|
Xue M, Lin H, Lynch T, Bereza-Malcolm L, Sinnathurai P, Thomas R, Keen H, Hill C, Lester S, Wechalekar M, March L. Exploring the association between circulating endothelial protein C receptor and disease activity of rheumatoid arthritis in a pilot study. Rheumatol Adv Pract 2024; 8:rkae096. [PMID: 39184533 PMCID: PMC11343369 DOI: 10.1093/rap/rkae096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/28/2024] [Indexed: 08/27/2024] Open
Abstract
Objectives To investigate whether circulating endothelial protein C receptor (EPCR) is associated with disease activity and inflammatory markers in rheumatoid arthritis. Methods Thirty-eight RA patients and 21 healthy controls (HC) were recruited via the A3BC biobank. Peripheral blood mononuclear cells and plasma were isolated from the blood of these participants. Plasma soluble (s)EPCR, IL-6, IL-17 and sCD14 were measured by enzyme-linked immunosorbent assay, cell membrane-associated (m)EPCR by flow cytometry; EPCR gene H3 single nucleotide polymorphism (SNP), which contributes to high plasma sEPCR levels, by PCR and DNA sequencing. Data were analysed using FlowJo10 and GraphPad Prism 10. Results RA patients had higher levels of mEPCR on T cells and plasma sEPCR compared with HC. No difference in the EPCR gene H3 SNP G genotype frequency was found between RA and HC. This SNP was significantly correlated with higher sEPCR levels in HC but not in RA patients. In RA, plasma sEPCR levels were positively correlated with IL-6, IL-17, sCD14, anti-CCP and rheumatoid factor. In contrast, mEPCR levels on T cells and natural killer cells (NK) were inversely associated with disease activity measures including 28/66 swollen joint count, 28/68 tender joint count and/or DAS28-CRP/ESR scores, and positively correlated with EPCR gene H3 SNP, which was also correlated with lower disease activity measures in RA. Conclusion Our findings suggest that EPCR may play an important role in RA, with plasma sEPCR being potentially associated with inflammatory markers and mEPCR and the EPCR gene H3 SNP possibly related to disease activity measures.
Collapse
Affiliation(s)
- Meilang Xue
- Sutton Arthritis Research Laboratory, Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Haiyan Lin
- Sutton Arthritis Research Laboratory, Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Tom Lynch
- The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Lara Bereza-Malcolm
- Sutton Arthritis Research Laboratory, Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Premarani Sinnathurai
- The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
- Department of Rheumatology, Royal North Shore Hospital, Syndey, NSW, Australia
| | - Ranjeny Thomas
- Frazer Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Helen Keen
- Medical School, The University of Western Australia, Perth, WA, Australia
- Department of Rheumatology, Fiona Stanley Hospital, Murdoch, WA, Australia
| | - Catherine Hill
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Rheumatology Research Group, Paediatrics, and Paediatric Rheumatology, Basil Hetzel Institute and The Queen Elizabeth Hospital, Adelaide, SA, Australia
| | - Susan Lester
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Rheumatology Research Group, Paediatrics, and Paediatric Rheumatology, Basil Hetzel Institute and The Queen Elizabeth Hospital, Adelaide, SA, Australia
| | - Mihir Wechalekar
- Rheumatology Synovial Tissue Translational Research Group, Flinders University, Adelaide, SA, Australia
- Rheumatology Unit, Flinders Medical Centre, Adelaide, SA, Australia
| | - Lyn March
- The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
- Department of Rheumatology, Royal North Shore Hospital, Syndey, NSW, Australia
| |
Collapse
|
4
|
O’Hehir ZD, Lynch T, O’Neill S, March L, Xue M. Endothelial Protein C Receptor and Its Impact on Rheumatic Disease. J Clin Med 2024; 13:2030. [PMID: 38610795 PMCID: PMC11012567 DOI: 10.3390/jcm13072030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
Endothelial Protein C Receptor (EPCR) is a key regulator of the activated protein C anti-coagulation pathway due to its role in the binding and activation of this protein. EPCR also binds to other ligands such as Factor VII and X, γδ T-cells, plasmodium falciparum erythrocyte membrane protein 1, and Secretory group V Phospholipases A2, facilitating ligand-specific functions. The functions of EPCR can also be regulated by soluble (s)EPCR that competes for the binding sites of membrane-bound (m)EPCR. sEPCR is created when mEPCR is shed from the cell surface. The propensity of shedding alters depending on the genetic haplotype of the EPCR gene that an individual may possess. EPCR plays an active role in normal homeostasis, anti-coagulation pathways, inflammation, and cell stemness. Due to these properties, EPCR is considered a potential effector/mediator of inflammatory diseases. Rheumatic diseases such as rheumatoid arthritis and systemic lupus erythematosus are autoimmune/inflammatory conditions that are associated with elevated EPCR levels and disease activity, potentially driven by EPCR. This review highlights the functions of EPCR and its contribution to rheumatic diseases.
Collapse
Affiliation(s)
- Zachary Daniel O’Hehir
- Sutton Arthritis Research Laboratory, Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney at Royal North Shore Hospital, Sydney, NSW 2065, Australia;
| | - Tom Lynch
- The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (T.L.); (L.M.)
| | - Sean O’Neill
- Department of Rheumatology, Royal North Shore Hospital, Syndey, NSW 2065, Australia;
| | - Lyn March
- The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (T.L.); (L.M.)
- Department of Rheumatology, Royal North Shore Hospital, Syndey, NSW 2065, Australia;
| | - Meilang Xue
- Sutton Arthritis Research Laboratory, Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney at Royal North Shore Hospital, Sydney, NSW 2065, Australia;
- The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (T.L.); (L.M.)
| |
Collapse
|
5
|
Xue M, Lin H, Liang HPH, Bereza-Malcolm L, Lynch T, Sinnathurai P, Weiler H, Jackson C, March L. EPCR deficiency ameliorates inflammatory arthritis in mice by suppressing the activation and migration of T cells and dendritic cells. Rheumatology (Oxford) 2024; 63:571-580. [PMID: 37228024 PMCID: PMC10834933 DOI: 10.1093/rheumatology/kead230] [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: 02/20/2023] [Revised: 04/08/2023] [Accepted: 05/10/2023] [Indexed: 05/27/2023] Open
Abstract
OBJECTIVES Endothelial protein C receptor (EPCR) is highly expressed in synovial tissues of patients with RA, but the function of this receptor remains unknown in RA. This study investigated the effect of EPCR on the onset and development of inflammatory arthritis and its underlying mechanisms. METHODS CIA was induced in EPCR gene knockout (KO) and matched wild-type (WT) mice. The onset and development of arthritis was monitored clinically and histologically. T cells, dendritic cells (DCs), EPCR and cytokines from EPCR KO and WT mice, RA patients and healthy controls (HCs) were detected by flow cytometry and ELISA. RESULTS EPCR KO mice displayed >40% lower arthritis incidence and 50% less disease severity than WT mice. EPCR KO mice also had significantly fewer Th1/Th17 cells in synovial tissues with more DCs in circulation. Lymph nodes and synovial CD4 T cells from EPCR KO mice expressed fewer chemokine receptors CXCR3, CXCR5 and CCR6 than WT mice. In vitro, EPCR KO spleen cells contained fewer Th1 and more Th2 and Th17 cells than WT and, in concordance, blocking EPCR in WT cells stimulated Th2 and Th17 cells. DCs generated from EPCR KO bone marrow were less mature and produced less MMP-9. Circulating T cells from RA patients expressed higher levels of EPCR than HC cells; blocking EPCR stimulated Th2 and Treg cells in vitro. CONCLUSION Deficiency of EPCR ameliorates arthritis in CIA via inhibition of the activation and migration of pathogenic Th cells and DCs. Targeting EPCR may constitute a novel strategy for future RA treatment.
Collapse
Affiliation(s)
- Meilang Xue
- Sutton Arthritis Research Laboratory, Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
- Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Haiyan Lin
- Sutton Arthritis Research Laboratory, Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
- Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Hai Po Helena Liang
- Sutton Arthritis Research Laboratory, Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Lara Bereza-Malcolm
- Sutton Arthritis Research Laboratory, Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
- Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Tom Lynch
- Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Premarani Sinnathurai
- Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Hartmut Weiler
- Versiti Blood Research Institute, Versiti, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Christopher Jackson
- Sutton Arthritis Research Laboratory, Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Lyn March
- Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| |
Collapse
|
6
|
Membrane curvature and PS localize coagulation proteins to filopodia and retraction fibers of endothelial cells. Blood Adv 2022; 7:60-72. [PMID: 35849711 PMCID: PMC9827038 DOI: 10.1182/bloodadvances.2021006870] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 01/18/2023] Open
Abstract
Prior reports indicate that the convex membrane curvature of phosphatidylserine (PS)-containing vesicles enhances formation of binding sites for factor Va and lactadherin. Yet, the relationship of convex curvature to localization of these proteins on cells remains unknown. We developed a membrane topology model, using phospholipid bilayers supported by nano-etched silica substrates, to further explore the relationship between curvature and localization of coagulation proteins. Ridge convexity corresponded to maximal curvature of physiologic membranes (radii of 10 or 30 nm) and the troughs had a variable concave curvature. The benchmark PS probe lactadherin exhibited strong differential binding to the ridges, on membranes with 4% to 15% PS. Factor Va, with a PS-binding motif homologous to lactadherin, also bound selectively to the ridges. Bound factor Va supported coincident binding of factor Xa, localizing prothrombinase complexes to the ridges. Endothelial cells responded to prothrombotic stressors and stimuli (staurosporine, tumor necrosis factor-α [TNF- α]) by retracting cell margins and forming filaments and filopodia. These had a high positive curvature similar to supported membrane ridges and selectively bound lactadherin. Likewise, the retraction filaments and filopodia bound factor Va and supported assembly of prothrombinase, whereas the cell body did not. The perfusion of plasma over TNF-α-stimulated endothelia in culture dishes and engineered 3-dimensional microvessels led to fibrin deposition at cell margins, inhibited by lactadherin, without clotting of bulk plasma. Our results indicate that stressed or stimulated endothelial cells support prothrombinase activity localized to convex topological features at cell margins. These findings may relate to perivascular fibrin deposition in sepsis and inflammation.
Collapse
|
7
|
Keshava S, Magisetty J, Tucker TA, Kujur W, Mulik S, Esmon CT, Idell S, Rao LVM, Pendurthi UR. Endothelial Cell Protein C Receptor Deficiency Attenuates Streptococcus pneumoniae-induced Pleural Fibrosis. Am J Respir Cell Mol Biol 2021; 64:477-491. [PMID: 33600743 PMCID: PMC8008801 DOI: 10.1165/rcmb.2020-0328oc] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 01/19/2021] [Indexed: 12/15/2022] Open
Abstract
Streptococcus pneumoniae is the leading cause of hospital community-acquired pneumonia. Patients with pneumococcal pneumonia may develop complicated parapneumonic effusions or empyema that can lead to pleural organization and subsequent fibrosis. The pathogenesis of pleural organization and scarification involves complex interactions between the components of the immune system, coagulation, and fibrinolysis. EPCR (endothelial protein C receptor) is a critical component of the protein C anticoagulant pathway. The present study was performed to evaluate the role of EPCR in the pathogenesis of S. pneumoniae infection-induced pleural thickening and fibrosis. Our studies show that the pleural mesothelium expresses EPCR. Intrapleural instillation of S. pneumoniae impairs lung compliance and lung volume in wild-type and EPCR-overexpressing mice but not in EPCR-deficient mice. Intrapleural S. pneumoniae infection induces pleural thickening in wild-type mice. Pleural thickening is more pronounced in EPCR-overexpressing mice, whereas it is reduced in EPCR-deficient mice. Markers of mesomesenchymal transition are increased in the visceral pleura of S. pneumoniae-infected wild-type and EPCR-overexpressing mice but not in EPCR-deficient mice. The lungs of wild-type and EPCR-overexpressing mice administered intrapleural S. pneumoniae showed increased infiltration of macrophages and neutrophils, which was significantly reduced in EPCR-deficient mice. An analysis of bacterial burden in the pleural lavage, the lungs, and blood revealed a significantly lower bacterial burden in EPCR-deficient mice compared with wild-type and EPCR-overexpressing mice. Overall, our data provide strong evidence that EPCR deficiency protects against S. pneumoniae infection-induced impairment of lung function and pleural remodeling.
Collapse
Affiliation(s)
| | | | | | - Weshely Kujur
- Department of Pulmonary Immunology, The University of Texas Health Science Center at Tyler, Tyler, Texas; and
| | - Sachin Mulik
- Department of Pulmonary Immunology, The University of Texas Health Science Center at Tyler, Tyler, Texas; and
| | - Charles T. Esmon
- Coagulation Biology Laboratory, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | | | | | | |
Collapse
|
8
|
EPCR deficiency or function-blocking antibody protects against joint bleeding-induced pathology in hemophilia mice. Blood 2021; 135:2211-2223. [PMID: 32294155 DOI: 10.1182/blood.2019003824] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 03/18/2020] [Indexed: 02/06/2023] Open
Abstract
We recently showed that clotting factor VIIa (FVIIa) binding to endothelial cell protein C receptor (EPCR) induces anti-inflammatory signaling and protects vascular barrier integrity. Inflammation and vascular permeability are thought to be major contributors to the development of hemophilic arthropathy following hemarthrosis. The present study was designed to investigate the potential influence of FVIIa interaction with EPCR in the pathogenesis of hemophilic arthropathy and its treatment with recombinant FVIIa (rFVIIa). For this, we first generated hemophilia A (FVIII-/-) mice lacking EPCR (EPCR-/-FVIII-/-) or overexpressing EPCR (EPCR++ FVIII-/-). Joint bleeding was induced in FVIII-/-, EPCR-/-FVIII-/-, and EPCR++FVIII-/- mice by needle puncture injury. Hemophilic synovitis was evaluated by monitoring joint bleeding, change in joint diameter, and histopathological analysis of joint tissue sections. EPCR deficiency in FVIII-/- mice significantly reduced the severity of hemophilic synovitis. EPCR deficiency attenuated the elaboration of interleukin-6, infiltration of macrophages, and neoangiogenesis in the synovium following hemarthrosis. A single dose of rFVIIa was sufficient to fully prevent the development of milder hemophilic synovitis in EPCR-/-FVIII-/- mice. The development of hemophilic arthropathy in EPCR-overexpressing FVIII-/- mice did not significantly differ from that of FVIII-/- mice, and 3 doses of rFVIIa partly protected against hemophilic synovitis in these mice. Consistent with the data that EPCR deficiency protects against developing hemophilic arthropathy, administration of a single dose of EPCR-blocking monoclonal antibodies markedly reduced hemophilic synovitis in FVIII-/- mice subjected to joint bleeding. The present data indicate that EPCR could be an attractive new target to prevent joint damage in hemophilia patients.
Collapse
|
9
|
Kondreddy V, Keshava S, Esmon CT, Pendurthi UR, Rao LVM. A critical role of endothelial cell protein C receptor in the intestinal homeostasis in experimental colitis. Sci Rep 2020; 10:20569. [PMID: 33239717 PMCID: PMC7689504 DOI: 10.1038/s41598-020-77502-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 11/11/2020] [Indexed: 12/28/2022] Open
Abstract
Crohn’s disease and ulcerative colitis are the two forms of disorders of the human inflammatory bowel disease with unknown etiologies. Endothelial cell protein C receptor (EPCR) is a multifunctional and multiligand receptor, which is expressed on the endothelium and other cell types, including epithelial cells. Here, we report that EPCR is expressed in the colon epithelial cells, CD11c+, and CD21+/CD35+ myeloid cells surrounding the crypts in the colon mucosa. EPCR expression was markedly decreased in the colon mucosa during colitis. The loss of EPCR appeared to associate with increased disease index of the experimental colitis in mice. EPCR−/− mice were more susceptible to dextran sulfate sodium (DSS)-induced colitis, manifested by increased weight loss, macrophage infiltration, and inflammatory cytokines in the colon tissue. DSS treatment of EPCR−/− mice resulted in increased bleeding, bodyweight loss, anemia, fibrin deposition, and loss of colon epithelial and goblet cells. Administration of coagulant factor VIIa significantly attenuated the DSS-induced colon length shortening, rectal bleeding, bodyweight loss, and disease activity index in the wild-type mice but not EPCR−/− mice. In summary, our data provide direct evidence that EPCR plays a crucial role in regulating the inflammation in the colon during colitis.
Collapse
Affiliation(s)
- Vijay Kondreddy
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center At Tyler, 11937 US Highway 271, Tyler, TX, 75708-3154, USA
| | - Shiva Keshava
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center At Tyler, 11937 US Highway 271, Tyler, TX, 75708-3154, USA
| | - Charles T Esmon
- Coagulation Biology Laboratory, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Usha R Pendurthi
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center At Tyler, 11937 US Highway 271, Tyler, TX, 75708-3154, USA
| | - L Vijaya Mohan Rao
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center At Tyler, 11937 US Highway 271, Tyler, TX, 75708-3154, USA.
| |
Collapse
|
10
|
Bai L, Liu W, Guo P, Bai J, Liu Y, Hua Y, Pang C, Zhang W, Yin F, Wang Y. Elevated levels of soluble Endothelial protein C receptor in rheumatoid arthritis and block the therapeutic effect of protein C in collagen-induced arthritis. Int Immunopharmacol 2020; 81:106255. [PMID: 32007797 DOI: 10.1016/j.intimp.2020.106255] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/16/2020] [Accepted: 01/24/2020] [Indexed: 12/01/2022]
Abstract
BACKGROUND Endothelial protein C receptor (EPCR) is a membranous protein that can be combined with a variety of ligands and plays important roles in anticoagulant and anti-inflammation. Recent reports have shown that surface EPCR expression on T cells is negatively associated with Th17 differentiation and is co-expressed with other immunosuppressive molecules, such as The programmed cell death 1 (PD-1) and cytotoxic T lymphocyte-associated antigen-4 (CTLA-4). Hence, we hypothesized that EPCR may play a critical role in rheumatoid arthritis (RA) disease progression that is mediated by Th17 differentiation. In order to explore the role of EPCR on RA disease pathogenesis, we detected membranous EPCR (mEPCR) expression in CD4+ T cells and soluble EPCR (sEPCR) expression in the sera of RA patients. METHODS The proportion of CD4+/EPCR+ T cells in the peripheral blood of RA patients was detected by flow cytometry, and the expression of sEPCR in the sera of RA patients was detected by enzyme-linked immunosorbent assay (ELISA). For in vitro experiments, protein C (PC) and EPCR recombinant proteins were used to block peripheral blood mononuclear cell (PBMC) activation and to detect Th17 differentiation. For in vivo experiments in DBA/1 mice with collagen-induced arthritis (CIA), we administered PC and EPCR recombinant proteins, monitored disease progression, and evaluated the role of EPCR in disease progression. RESULTS The proportion of CD4+/EPCR+ T cells in the peripheral blood of RA patients was lower than that of osteoarthritis (OA) patients, while the expression level of sEPCR in the sera of RA patients was concomitantly higher than that in OA patients. Subsequent analysis revealed that sEPCR expression was positively correlated with rheumatoid factors (RF) and other inflammatory indicators in RA patients. Further studies confirmed that sEPCR administration alleviated the progression of collagen-induced arthritis and partially blocked the therapeutic effect of PC in CIA mice. CONCLUSION Soluble EPCR is associated with RA disease progression and induces disease remission in CIA mice by inhibiting Th17 differentiation.
Collapse
Affiliation(s)
- Li Bai
- Department of Rheumatology, The First Affiliated Hospital of Baotou Medical College, Baotou 014010, China; Institute of Immunology and Rheumatology, Baotou Medical College (Inner Mongolia Key Laboratory of Autoimmunity), Baotou 014010, China; Baotou Medical College, Baotou 014010, China
| | - Wenjia Liu
- Department of Rheumatology, The First Affiliated Hospital of Baotou Medical College, Baotou 014010, China; Baotou Medical College, Baotou 014010, China
| | - Pengnian Guo
- Department of Orthopedic, The First Affiliated Hospital of Baotou Medical College, Baotou 014010, China
| | - Jingru Bai
- Department of Rheumatology, The First Affiliated Hospital of Baotou Medical College, Baotou 014010, China; Baotou Medical College, Baotou 014010, China
| | - Yuan Liu
- Department of Rheumatology, The First Affiliated Hospital of Baotou Medical College, Baotou 014010, China; Baotou Medical College, Baotou 014010, China
| | - Yana Hua
- Department of Rheumatology, The First Affiliated Hospital of Baotou Medical College, Baotou 014010, China; Baotou Medical College, Baotou 014010, China
| | - Chunyan Pang
- Department of Rheumatology, The First Affiliated Hospital of Baotou Medical College, Baotou 014010, China; Institute of Immunology and Rheumatology, Baotou Medical College (Inner Mongolia Key Laboratory of Autoimmunity), Baotou 014010, China; Baotou Medical College, Baotou 014010, China
| | - Wei Zhang
- Department of Rheumatology, The First Affiliated Hospital of Baotou Medical College, Baotou 014010, China; Institute of Immunology and Rheumatology, Baotou Medical College (Inner Mongolia Key Laboratory of Autoimmunity), Baotou 014010, China; Baotou Medical College, Baotou 014010, China
| | - Fangrui Yin
- Department of Rheumatology, The First Affiliated Hospital of Baotou Medical College, Baotou 014010, China; Institute of Immunology and Rheumatology, Baotou Medical College (Inner Mongolia Key Laboratory of Autoimmunity), Baotou 014010, China; Baotou Medical College, Baotou 014010, China
| | - Yongfu Wang
- Department of Rheumatology, The First Affiliated Hospital of Baotou Medical College, Baotou 014010, China; Institute of Immunology and Rheumatology, Baotou Medical College (Inner Mongolia Key Laboratory of Autoimmunity), Baotou 014010, China; Baotou Medical College, Baotou 014010, China.
| |
Collapse
|
11
|
Activated Protein C in Cutaneous Wound Healing: From Bench to Bedside. Int J Mol Sci 2019; 20:ijms20040903. [PMID: 30791425 PMCID: PMC6412604 DOI: 10.3390/ijms20040903] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/14/2019] [Accepted: 02/16/2019] [Indexed: 12/12/2022] Open
Abstract
Independent of its well-known anticoagulation effects, activated protein C (APC) exhibits pleiotropic cytoprotective properties. These include anti-inflammatory actions, anti-apoptosis, and endothelial and epithelial barrier stabilisation. Such beneficial effects have made APC an attractive target of research in a plethora of physiological and pathophysiological processes. Of note, the past decade or so has seen the emergence of its roles in cutaneous wound healing-a complex process involving inflammation, proliferation and remodelling. This review will highlight APC's functions and mechanisms, and detail its pre-clinical and clinical studies on cutaneous wound healing.
Collapse
|
12
|
Chapelet A, Foucher Y, Gérard N, Rousseau C, Zambon O, Bretonnière C, Mira JP, Charreau B, Guitton C. An early increase in endothelial protein C receptor is associated with excess mortality in pneumococcal pneumonia with septic shock in the ICU. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2018; 22:251. [PMID: 30290852 PMCID: PMC6173894 DOI: 10.1186/s13054-018-2179-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 09/05/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND This study investigated changes in plasma level of soluble endothelial protein C receptor (sEPCR) in association with outcome in patients with septic shock. We explored sEPCR for early sepsis prognosis assessment and constructed a scoring system based on clinical and biological data, in order to discriminate between surviving at hospital discharge and non-surviving patients. METHODS Clinical data and samples were extracted from the prospective "STREPTOGENE" cohort. We enrolled 278 patients, from 50 intensive care units (ICUs), with septic shock caused by pneumococcal pneumonia. Patients were divided into survivors (n = 194) and non-survivors (n = 84) based on in-hospital mortality. Soluble EPCR plasma levels were quantified at day 1 (D1) and day 2 (D2) by ELISA. The EPCR gene A3 haplotype was determined. Patients were followed up until hospital discharge. Univariate and multivariate analyses were performed. A scoring system was constructed using least absolute shrinkage and selection operator (lasso) logistic regression for selecting predictive variables. RESULTS In-hospital mortality was 30.2% (n = 84). Plasma sEPCR level was significantly higher at D1 and D2 in non-surviving patients compared to patients surviving to hospital discharge (p = 0.0447 and 0.0047, respectively). Early increase in sEPCR at D2 was found in non-survivors while a decrease was observed in the survival group (p = 0.0268). EPCR A3 polymorphism was not associated with mortality. Baseline sEPCR level and its variation from D1 to D2 were independent predictors of in-hospital mortality. The scoring system including sEPCR predicted mortality with an AUC of 0.75. CONCLUSIONS Our findings confirm that high plasma sEPCR and its increase at D2 are associated with poor outcome in sepsis and thus we propose sEPCR as a key player in the pathogenesis of sepsis and as a potential biomarker of sepsis outcome.
Collapse
Affiliation(s)
- Agnès Chapelet
- Medical Intensive Care Unit, Nantes University Hospital, Nantes, France.,Centre for Research in Transplantation and Immunology (CRTI) UMR1064, INSERM, Nantes University, Nantes, France.,Institute of Transplantation Urology Nephrology (ITUN), Nantes University Hospital, Nantes, France
| | - Yohann Foucher
- INSERM, UMR 1246 - SPHERE, Nantes University, Nantes University Hospital, Nantes, France
| | - Nathalie Gérard
- Centre for Research in Transplantation and Immunology (CRTI) UMR1064, INSERM, Nantes University, Nantes, France
| | | | - Olivier Zambon
- Medical Intensive Care Unit, Nantes University Hospital, Nantes, France
| | | | - Jean-Paul Mira
- Institut Cochin, INSERM U1016, Paris, France.,Medical Intensive Care Unit, Cochin University Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Paris, France
| | - Béatrice Charreau
- Centre for Research in Transplantation and Immunology (CRTI) UMR1064, INSERM, Nantes University, Nantes, France.,Institute of Transplantation Urology Nephrology (ITUN), Nantes University Hospital, Nantes, France
| | - Christophe Guitton
- Medical Intensive Care Unit, Nantes University Hospital, Nantes, France. .,Centre for Research in Transplantation and Immunology (CRTI) UMR1064, INSERM, Nantes University, Nantes, France. .,Medical and Surgical Intensive Care Unit, Le Mans Hospital, Le Mans, France.
| |
Collapse
|
13
|
Exploring traditional and nontraditional roles for thrombomodulin. Blood 2018; 132:148-158. [DOI: 10.1182/blood-2017-12-768994] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/19/2018] [Indexed: 12/19/2022] Open
Abstract
AbstractThrombomodulin (TM) is an integral component of a multimolecular system, localized primarily to the vascular endothelium, that integrates crucial biological processes and biochemical pathways, including those related to coagulation, innate immunity, inflammation, and cell proliferation. These are designed to protect the host from injury and promote healing. The “traditional” role of TM in hemostasis was determined with its discovery in the 1980s as a ligand for thrombin and a critical cofactor for the major natural anticoagulant protein C system and subsequently for thrombin-mediated activation of the thrombin activatable fibrinolysis inhibitor (also known as procarboxypeptidase B2). Studies in the past 2 decades are redefining TM as a molecule with many properties, exhibited via its multiple domains, through its interacting partners, complex regulated expression, and synthesis by cells other than the endothelium. In this report, we review some of the recently reported diverse properties of TM and how these may impact on our understanding of the pathogenesis of several diseases.
Collapse
|
14
|
Yoshida K, Akita N, Okamoto T, Asanuma K, Uchida A, Sudo A, Shimaoka M, Suzuki K, Hayashi T. Activated protein C suppresses osteoclast differentiation via endothelial protein C receptor, protease-activated receptor-1, sphingosine 1-phosphate receptor, and apolipoprotein E receptor 2. Thromb Res 2018; 163:30-40. [PMID: 29334656 DOI: 10.1016/j.thromres.2018.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 12/24/2017] [Accepted: 01/02/2018] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Bone remodeling relies on a delicate balance between formation and resorption of bone tissues, processes in which bone-forming osteoblasts and bone-resorbing osteoclasts play central roles. Recently, we reported that anticoagulant activated protein C (APC) promotes osteoblast proliferation, but the role of the blood coagulation system in bone remodeling remains unclear. In this study, to further elucidate the relationship between bone remodeling and blood coagulation, we investigated the effect of APC on osteoclast differentiation. MATERIALS AND METHODS Normal human osteoclast precursor cells were cultured in their growth medium including soluble RANKL, M-CSF, and FBS, and on days 4 and 7, the culture medium was replaced with the same medium containing various concentrations of APC, protein C (PC), sphingosine 1-phosphate (S1P) receptor agonist, FTY720, or APC+various substances without FBS. On day 8, TRAP-positive multinucleated cells (≥3 nuclei) were counted manually using a light microscope. The effects of APC on NF-κB and NFATc1 activation were evaluated using specific ELISA. RESULTS APC suppressed RANKL-induced osteoclast differentiation, and this APC-induced suppression of osteoclast differentiation was inhibited by zymogen protein C and aprotinin, a serine protease inhibitor. Immunohistochemistry and RT-PCR analyses suggested that endothelial protein C receptor (EPCR) and protease-activated receptor-1 (PAR-1) were expressed in osteoclast precursor cells and osteoclasts. Both anti-PAR-1 antibody and anti-EPCR antibody (RCR-252), which blocks APC binding to EPCR, inhibited the APC-induced suppression of osteoclast differentiation. FTY720 had no effect on osteoclast differentiation. However, FTY 720 and S1P receptor antagonist, VP 23019, inhibited the APC-induced suppression of osteoclast differentiation. On the other hand, recombinant soluble human ApoER2 and anti-human ApoER2 inhibited the APC-induced suppression of osteoclast differentiation. Further, APC had no effect on NF-κB and NFATc1 activation. CONCLUSIONS APC suppresses human osteoclast differentiation mainly by inhibiting the formation of multinucleated cells via EPCR, PAR-1, S1P receptor, and ApoER2 in a manner that depends on APC protease activity.
Collapse
Affiliation(s)
- Kakunoshin Yoshida
- Department of Orthopedic Surgery, Mie University Graduate School of Medicine, Tsu-city, Mie, Japan
| | - Nobuyuki Akita
- Faculty of Medical Engineering, Suzuka University of Medical Science, Suzuka-city, Mie, Japan
| | - Takayuki Okamoto
- Department of Pharmacology, Faculty of Medicine, Shimane University, Shimane-city, Shimane, Japan
| | - Kunihiro Asanuma
- Department of Orthopedic Surgery, Mie University Graduate School of Medicine, Tsu-city, Mie, Japan
| | - Atsumasa Uchida
- Department of Orthopedic Surgery, Mie University Graduate School of Medicine, Tsu-city, Mie, Japan
| | - Akihiro Sudo
- Department of Orthopedic Surgery, Mie University Graduate School of Medicine, Tsu-city, Mie, Japan
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu-city, Mie, Japan
| | - Koji Suzuki
- Faculty of Pharmaceutical Science, Suzuka University of Medical Science, Suzuka-city, Mie, Japan
| | - Tatsuya Hayashi
- Faculty of Nursing Science, Mie Prefectural College of Nursing, Tsu-city, Mie, Japan.
| |
Collapse
|
15
|
|
16
|
Ma Y, Zhao Y, Zhang R, Liang X, Yin Z, Geng Y, Shu G, Song X, Zou Y, Li L, Yin L, Yue G, Li Y, Ye G, He C. α-Cyperone Inhibits PMA-Induced EPCR Shedding through PKC Pathway. Biol Pharm Bull 2017; 40:1678-1685. [PMID: 28804104 DOI: 10.1248/bpb.b17-00183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
α-Cyperone, a sesquiterpene compound represents 25.23% of the total oil and is the most abundant compound in Cyperus rotundus oil. Endothelial cell protein C receptor (EPCR) is a main member in protein C (PC) anti-coagulation system. EPCR could be shed from cell surface, and is mediated by tumor necrosis factor-α converting enzyme (TACE). Nothing that EPCR is a marker of vascular barrier integrity in vascular inflammatory disease and takes part in systemic inflammatory disease. In this study, we investigated whether α-cyperone could inhibit EPCR shedding. To observe the effect, we investigated this issue by detection the effect of α-cyperone on phorbol-12-myristate 13-acetate (PMA)-induced EPCR shedding in human umbilical vein endothelial cells (HUVECs). The cells were pretreated with α-cyperone for 12 h, and then stimulated by PMA for 1 h. The solute EPCR (sEPCR) and expression of membrane EPCR (mEPCR) were measured by enzyme-linked immunosorbent assay (ELISA) and Western blot. The mRNA, protein level and activity of TACE were tested by quantitative (q)RT-PCR, Western blot and InnoZyme TACE activity assay kit. Furthermore, we measured the protein level of mitogen-activated protein kinase (MAPK) signaling and protein kinase C (PKC) pathway under this condition by Western blot. The results showed that α-cyperone could suppress PMA-induced EPCR shedding through inhibiting the expression and activity of TACE. In addition, α-cyperone could inhibit PKC translocation, but not have an effect on phosphorylation of c-Jun N-terminal kinase (JNK), p38 and extracellular regulated protein kinases (ERK) 1/2. Given these results, α-cyperone inhibits PMA-induced EPCR shedding through PKC pathway, which will provide an experimental basis for further research on α-cyperone.
Collapse
Affiliation(s)
- Yu Ma
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University
| | - Yi Zhao
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University
| | - Ran Zhang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University
| | - Xiaoxia Liang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University
| | - Zhongqiong Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University
| | - Yi Geng
- Department of Pharmacy, Sichuan Agricultural University
| | - Gang Shu
- Department of Pharmacy, Sichuan Agricultural University
| | - Xu Song
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University
| | - Yuanfeng Zou
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University
| | - Lixia Li
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University
| | - Lizi Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University
| | - Guizhou Yue
- College of Science, Sichuan Agricultural University
| | - Yinglun Li
- Department of Pharmacy, Sichuan Agricultural University
| | - Gang Ye
- Department of Pharmacy, Sichuan Agricultural University
| | - Changliang He
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University
| |
Collapse
|
17
|
Kleine SA, Budsberg SC. Synovial membrane receptors as therapeutic targets: A review of receptor localization, structure, and function. J Orthop Res 2017; 35:1589-1605. [PMID: 28374922 DOI: 10.1002/jor.23568] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 03/28/2017] [Indexed: 02/04/2023]
Abstract
Joint pathology and degeneration is a significant cause of pain. The synovial membrane plays an important role in maintenance of the joint, contributes to the pathology of many arthropathies and may be adversely affected in joint disease. Improving knowledge of the receptors present within the synovium will aid in a better understanding of joint pathology and the development of new treatments for diseases such as osteoarthritis and rheumatoid arthritis. Knowledge of the location and function of synovial membrane receptors (both in healthy and diseased synovium) may provide important targets in the treatment of various arthropathies. Classic pain receptors such as opioid receptors in the synovium are a mainstay in local and systemic management of chronic pain in many species. In addition to these, many other receptors such as bradykinin, neurokinin, transient receptor potential vanilloid, and inflammatory receptors, such as prostanoid and interleukin receptors have been discovered within the synovial membrane. These receptors are important in pain, inflammation, and in maintenance of normal joint function and may serve as targets for pharmacologic intervention in pathologic states. The goal of this review is to outline synovial membrane receptor localization and local therapeutic modulation of these receptors, in order to stimulate further research into pharmacological management of arthropathies at the local level. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:1589-1605, 2017.
Collapse
Affiliation(s)
- Stephanie A Kleine
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, 2200 College Station Road, Athens 30602, Georgia
| | - Steven C Budsberg
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, 2200 College Station Road, Athens 30602, Georgia
| |
Collapse
|
18
|
Xue M, Dervish S, Chan B, Jackson CJ. The Endothelial Protein C Receptor Is a Potential Stem Cell Marker for Epidermal Keratinocytes. Stem Cells 2017; 35:1786-1798. [PMID: 28480559 DOI: 10.1002/stem.2630] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 03/16/2017] [Accepted: 04/06/2017] [Indexed: 11/10/2022]
Abstract
Endothelial protein C receptor (EPCR) is a specific receptor for anticoagulant protein C and expressed by human epidermis and cultured keratinocytes. Here we investigated whether: (a) the level of EPCR in keratinocytes is associated with their growth potential; and (b) EPCR is a potential marker for human epidermal stem cells. Human keratinocytes isolated from foreskins or adult skin tissues were transfected with EPCR siRNA or EPCR overexpressing plasmids. Cell proliferation, long term proliferation potential, colony forming efficiency (CFE), and in vitro epidermal regeneration ability of EPCRhigh and EPCRl °w cells were assessed. The expression and colocalization of EPCR with stem cell markers p63, integrin β1, and activation of MAP kinases were detected by flow cytometry, immunofluorescence staining, or Western blot. Results showed that EPCR was highly expressed by the basal layer of skin epidermis. EPCRhigh cells were associated with the highest levels of p63 and integrin β1. Most EPCRhigh cells were smaller in size, formed larger colonies and had a greater long term growth potential, CFE, holoclone formation, and in vitro epidermal regeneration ability when compared to EPCRl °w cells. Blocking EPCR resulted in keratinocyte apoptosis, particularly in nondifferentiated conditions. Cell proliferation and p63 expression were reduced by blocking EPCR and enhanced by overexpressing this receptor. These data indicate that EPCR can regulate p63, is associated with highly proliferative keratinocytes, and is a potential human epidermal stem cell marker. Stem Cells 2017;35:1786-1798.
Collapse
Affiliation(s)
- Meilang Xue
- Sutton Research Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, University of Sydney at Royal North Shore Hospital, Camperdown, New South Wales, Australia
| | - Suat Dervish
- Sutton Research Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, University of Sydney at Royal North Shore Hospital, Camperdown, New South Wales, Australia.,Westmead Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Benjamin Chan
- Raymond Purves Research Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, University of Sydney at Royal North Shore Hospital, Camperdown, New South Wales, Australia
| | - Christopher J Jackson
- Sutton Research Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, University of Sydney at Royal North Shore Hospital, Camperdown, New South Wales, Australia
| |
Collapse
|
19
|
Foley JH, Conway EM. Cross Talk Pathways Between Coagulation and Inflammation. Circ Res 2017; 118:1392-408. [PMID: 27126649 DOI: 10.1161/circresaha.116.306853] [Citation(s) in RCA: 409] [Impact Index Per Article: 51.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 03/21/2016] [Indexed: 02/06/2023]
Abstract
Anatomic pathology studies performed over 150 years ago revealed that excessive activation of coagulation occurs in the setting of inflammation. However, it has taken over a century since these seminal observations were made to delineate the molecular mechanisms by which these systems interact and the extent to which they participate in the pathogenesis of multiple diseases. There is, in fact, extensive cross talk between coagulation and inflammation, whereby activation of one system may amplify activation of the other, a situation that, if unopposed, may result in tissue damage or even multiorgan failure. Characterizing the common triggers and pathways are key for the strategic design of effective therapeutic interventions. In this review, we highlight some of the key molecular interactions, some of which are already showing promise as therapeutic targets for inflammatory and thrombotic disorders.
Collapse
Affiliation(s)
- Jonathan H Foley
- From the Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom (J.H.F.); Katharine Dormandy Haemophilia Centre and Thrombosis Unit, Royal Free NHS Trust, London, United Kingdom (J.H.F.); and Centre for Blood Research, Department of Medicine, University of British Columbia, Vancouver, Canada (E.M.C.)
| | - Edward M Conway
- From the Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom (J.H.F.); Katharine Dormandy Haemophilia Centre and Thrombosis Unit, Royal Free NHS Trust, London, United Kingdom (J.H.F.); and Centre for Blood Research, Department of Medicine, University of British Columbia, Vancouver, Canada (E.M.C.).
| |
Collapse
|
20
|
Ma Y, Zhao Y, Zhang R, Liang X, Yin Z, Geng Y, Shu G, Song X, Zou Y, Li L, Yin L, Yue G, Li Y, Ye G, He C. Astragaloside IV inhibits PMA-induced EPCR shedding through MAPKs and PKC pathway. Immunopharmacol Immunotoxicol 2017; 39:148-156. [PMID: 28367652 DOI: 10.1080/08923973.2017.1306868] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Astragaloside IV (AS-IV), a main active substance isolated from Astragalus membranaceus Bunge, has been shown to have multiple pharmacological effects. Endothelial cell protein C receptor (EPCR) is a marker of inflammation, and is also a major member of protein C (PC) anti-coagulation system. EPCR can be cut off from the cell surface by tumor necrosis factor-α converting enzyme (TACE), which is controlled through mitogen-activated protein kinase (MAPK) and protein kinase C (PKC) pathways. To develop novel therapeutic drug for EPCR shedding, the effect of AS-IV was studied in phorbol-12-myristate 13-acetate (PMA)-induced human umbilical vein endothelial cells (HUVECs) and the potential molecular mechanism of AS-IV action was investigated. The results showed that AS-IV could significantly inhibit PMA-induced EPCR shedding. In further study, AS-IV suppressed the expression and activity of TACE. In addition, AS-IV could decrease the phosphorylation of MAPK such as janus kinase (JNK) and p38, and inhibit activation of PKC through the prevention of non-phosphorylation and phosphorylation of specific PKC isoforms in PMA-stimulated HUVECs. These findings indicate that AS-IV may be used as a natural medicine to treat EPCR-related systemic inflammation and cardiovascular diseases by targeting MAPK and PKC pathway.
Collapse
Affiliation(s)
- Yu Ma
- a Natural Medicine Research Center, College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , PR China
| | - Yi Zhao
- a Natural Medicine Research Center, College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , PR China
| | - Ran Zhang
- a Natural Medicine Research Center, College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , PR China
| | - Xiaoxia Liang
- a Natural Medicine Research Center, College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , PR China
| | - Zhongqiong Yin
- a Natural Medicine Research Center, College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , PR China
| | - Yi Geng
- b Department of Pharmacy , Sichuan Agricultural University , Chengdu , PR China
| | - Gang Shu
- b Department of Pharmacy , Sichuan Agricultural University , Chengdu , PR China
| | - Xu Song
- a Natural Medicine Research Center, College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , PR China
| | - Yuanfeng Zou
- a Natural Medicine Research Center, College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , PR China
| | - Lixia Li
- a Natural Medicine Research Center, College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , PR China
| | - Lizi Yin
- a Natural Medicine Research Center, College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , PR China
| | - Guizhou Yue
- c College of Science , Sichuan Agricultural University , Ya'an , PR China
| | - Yinglun Li
- b Department of Pharmacy , Sichuan Agricultural University , Chengdu , PR China
| | - Gang Ye
- b Department of Pharmacy , Sichuan Agricultural University , Chengdu , PR China
| | - Changliang He
- a Natural Medicine Research Center, College of Veterinary Medicine , Sichuan Agricultural University , Chengdu , PR China
| |
Collapse
|
21
|
Keshava S, Rao LVM, Pendurthi UR. Intrapleural Adenoviral-mediated Endothelial Cell Protein C Receptor Gene Transfer Suppresses the Progression of Malignant Pleural Mesothelioma in a Mouse Model. Sci Rep 2016; 6:36829. [PMID: 27833109 PMCID: PMC5104979 DOI: 10.1038/srep36829] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/21/2016] [Indexed: 12/29/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive thoracic cancer with a high mortality rate as it responds poorly to standard therapeutic interventions. Our recent studies showed that expression of endothelial cell protein C receptor (EPCR) in MPM cells suppresses tumorigenicity. The present study was aimed to investigate the mechanism by which EPCR suppresses MPM tumor growth and evaluate whether EPCR gene therapy could suppress the progression of MPM in a mouse model of MPM. Measurement of cytokines from the pleural lavage showed that mice implanted with MPM cells expressing EPCR had elevated levels of IFNγ and TNFα compared to mice implanted with MPM cells lacking EPCR. In vitro studies demonstrated that EPCR expression renders MPM cells highly susceptible to IFNγ + TNFα-induced apoptosis. Intrapleural injection of Ad.EPCR into mice with an established MPM originating from MPM cells lacking EPCR reduced the progression of tumor growth. Ad.EPCR treatment elicited recruitment of macrophages and NK cells into the tumor microenvironment and increased IFNγ and TNFα levels in the pleural space. Ad.EPCR treatment resulted in a marked increase in tumor cell apoptosis. In summary, our data show that EPCR expression in MPM cells promotes tumor cell apoptosis, and intrapleural EPCR gene therapy suppresses MPM progression.
Collapse
Affiliation(s)
- Shiva Keshava
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Texas, USA
| | - L Vijaya Mohan Rao
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Texas, USA
| | - Usha R Pendurthi
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Texas, USA
| |
Collapse
|
22
|
Adukpo S, Gyan BA, Ofori MF, Dodoo D, Velavan TP, Meyer CG. Triggering receptor expressed on myeloid cells 1 (TREM-1) and cytokine gene variants in complicated and uncomplicated malaria. Trop Med Int Health 2016; 21:1592-1601. [PMID: 27671831 DOI: 10.1111/tmi.12787] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Malaria elicits inflammatory responses, which, if not well regulated, may exert detrimental effects. When activated, triggering receptor expressed on myeloid cells 1 (TREM-1) enhances inflammatory responses by increasing secretion of IL-8 and other Th1 cytokines. In contrast, TREM-like transcript 1 (TREML-1) promotes anti-inflammatory responses by binding to TREM-1 ligands and competing with TREM-1, thus antagonizing TREM-1 activation to reduce inflammation. Endothelial protein C receptor (EPCR) also mediates anti-inflammatory responses by activating endothelial protein C (PC). Upon microbial stimulation, soluble forms of TREM-1 (sTREM-1) and soluble EPCR (sEPCR) are released. Their plasma levels reflect the degree of inflammation and the severity of infection. METHODS In a cross-sectional study comparing patients with severe with uncomplicated malaria, sTREM-1, soluble TREML-1 (sTREML-1) and sEPCR plasma levels as well as plasma levels of sEPCR derived from convalescent patients were quantified. Samples were collected on admittance of paediatric patients infected with Plasmodium falciparum to hospitals in Accra, Ghana. Distinct genetic regions of the genes encoding TREM-1, EPCR, interleukin (IL)-8 and IL-18 encompassing known genetic polymorphisms that influence plasma levels underwent DNA sequencing. RESULTS Higher sTREM-1 levels were observed among children suffering from severe malaria compared to those with uncomplicated malaria (P = 0.049). Low TREM-1 to TREML-1 ratios were associated with uncomplicated malaria (P = 0.033). The TREM1 rs2234237T variant causing the amino acid exchange Thr25Ser, which has been associated with higher TREM-1 plasma levels, was significantly more frequent among patients with severe malaria than in those with uncomplicated malaria (P = 0.036). Low levels of sEPCR were observed in severe and uncomplicated malaria, while variant genotypes of IL8, IL18 and EPCR did not show any association. CONCLUSION Higher plasma levels of sTREM-1 alone or relative to sTREML-1 during malaria predispose to the phenotype of severe malaria. Carriage of the TREM1 rs2234237T allele appears to be a risk factor for the development of severe malaria.
Collapse
Affiliation(s)
- Selorme Adukpo
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany.,Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Ben A Gyan
- Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Michael F Ofori
- Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Daniel Dodoo
- Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Thirumalaisamy P Velavan
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany.,Fondation Congolaise pour la Recherche Médicale, Brazzaville, Republic of Congo.,Duy Tan University, Da Nang, Vietnam
| | - Christian G Meyer
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany.,Duy Tan University, Da Nang, Vietnam
| |
Collapse
|
23
|
Xue M, Jackson CJ. Novel Functions of the Anticoagulant Activated Protein C in Maintaining Skin Barrier Integrity to Impact on Skin Disease. Pathobiology 2015; 82:100-6. [PMID: 26160431 DOI: 10.1159/000430957] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 04/27/2015] [Indexed: 11/19/2022] Open
Abstract
The epidermis is the outermost skin layer and provides the first line of defence against the external environment. Keratinocytes are the most predominant cells in the epidermis and play a critical role in maintaining epidermal barrier function. When the barrier is disrupted any of a number of diseases, such as chronic wounds, psoriasis, pemphigus, atopic dermatitis or toxic epidermal necrolysis, can take hold. Activated protein C (APC) or its precursor, protein C, is abundantly expressed by skin epidermal keratinocytes and stimulates their proliferation and migration, and inhibits apoptosis and inflammation, leading to a healing phenotype. Importantly, APC also increases the barrier function of keratinocytes by promoting expression and cell-cell contact redistribution of tight junction proteins. These cytoprotective properties of APC on epidermal keratinocytes place it as an exciting new therapy for skin disorders associated with the disruption of barrier function and inflammation.
Collapse
Affiliation(s)
- Meilang Xue
- Sutton Arthritis Research Laboratories, Institute of Bone and Joint Research, Kolling Institute, University of Sydney, Royal North Shore Hospital, St Leonards, N.S.W., Australia
| | | |
Collapse
|
24
|
Magnus N, D'Asti E, Meehan B, Garnier D, Rak J. Oncogenes and the coagulation system--forces that modulate dormant and aggressive states in cancer. Thromb Res 2015; 133 Suppl 2:S1-9. [PMID: 24862126 DOI: 10.1016/s0049-3848(14)50001-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cancers arise and progress genetically amidst profound perturbations of the microenvironmental and systemic homeostasis. This includes the coagulation system, which is a part of the vascular milieu (niche) that remains under the control of molecular events occurring within the cancer cell genome. Thus, activation of several prototypic oncogenic pathways, such as RAS, EGFR, HER2, MET, SHH and loss of tumor suppressors (PTEN, TP53) alter the expression, activity and vesicular release of coagulation effectors, as exemplified by tissue factor (TF). The cancer-specific determinants of coagulopathy are also illustrated by the emerging link between the expression profiles of coagulation-related genes (coagulome) in glioblastoma multiforme (GBM), medulloblastoma (MB) and possibly other cancers and molecular subtypes of these respective tumors. The state of the coagulome is consequential for growth, metastasis and angiogenesis of established tumors, but could potentially also affect dormant cancer cells. For example, TF expression may trigger awakening of dormant glioma cells in mice in a manner involving recruitment of vascular and inflammatory cells, and resulting in lasting changes in the cancer cell genome and epigenome. Thus, coagulation system effectors could act as both targets and (indirect) inducers of genetic tumor progression, and a better understanding of this link may hold new diagnostic and therapeutic opportunities.
Collapse
Affiliation(s)
- Nathalie Magnus
- Montreal Children's Hospital, RI MUHC, McGill University, Montreal, Quebec, Canada
| | - Esterina D'Asti
- Montreal Children's Hospital, RI MUHC, McGill University, Montreal, Quebec, Canada
| | - Brian Meehan
- Montreal Children's Hospital, RI MUHC, McGill University, Montreal, Quebec, Canada
| | - Delphine Garnier
- Montreal Children's Hospital, RI MUHC, McGill University, Montreal, Quebec, Canada
| | - Janusz Rak
- Montreal Children's Hospital, RI MUHC, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
25
|
Ku SK, Han MS, Lee MY, Lee YM, Bae JS. Inhibitory effects of oroxylin A on endothelial protein C receptor shedding in vitro and in vivo. BMB Rep 2015; 47:336-41. [PMID: 24286327 PMCID: PMC4163868 DOI: 10.5483/bmbrep.2014.47.6.198] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Indexed: 11/24/2022] Open
Abstract
Endothelial cell protein C receptor (EPCR) plays important roles in blood coagulation and inflammation. EPCR activity is markedly changed by ectodomain cleavage and release as the soluble EPCR. EPCR can be shed from the cell surface, which is mediated by tumor necrosis factor-α converting enzyme (TACE). Oroxylin A (OroA), a major component of Scutellaria baicalensis Georgi, is known to exhibit anti-angiogenic, antiinflammation, and anti-invasive activities. However, little is known about the effects of OroA on EPCR shedding. Data showed that OroA induced potent inhibition of phorbol-12-myristate 13-acetate (PMA), tumor necrosis factor (TNF)-α, interleukin (IL)-1β and on cecal ligation and puncture (CLP)-induced EPCR shedding through suppression of TACE expression and activity. In addition, treatment with OroA resulted in reduced PMA-stimulated phosphorylation of p38, extracellular regulated kinases (ERK) 1/2, and c-Jun N-terminal kinase (JNK). These results demonstrate the potential of OroA as an anti-sEPCR shedding reagent against PMA and CLP-mediated EPCR shedding. [BMB Reports 2014; 47(6): 336-341]
Collapse
Affiliation(s)
- Sae-Kwang Ku
- Department of Anatomy and Histology, College of Oriental Medicine, Daegu Haany University, Gyeongsan 712-715, Korea
| | - Min-Su Han
- Laboratory for Arthritis and Bone Biology, Fatima Research Institute, Daegu Fatima Hospital, Daegu 701-724, Korea
| | - Min Young Lee
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Korea
| | - You-Mie Lee
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Korea
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Korea
| |
Collapse
|
26
|
Couzi L, Pitard V, Moreau JF, Merville P, Déchanet-Merville J. Direct and Indirect Effects of Cytomegalovirus-Induced γδ T Cells after Kidney Transplantation. Front Immunol 2015; 6:3. [PMID: 25653652 PMCID: PMC4301015 DOI: 10.3389/fimmu.2015.00003] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 01/04/2015] [Indexed: 01/30/2023] Open
Abstract
Despite effective anti-viral therapies, cytomegalovirus (CMV) is still associated with direct (CMV disease) and indirect effects (rejection and poor graft survival) in kidney transplant recipients. Recently, an unconventional T cell population (collectively designated as Vδ2neg γδ T cells) has been characterized during the anti-CMV immune response in all solid-organ and bone-marrow transplant recipients, neonates, and healthy people. These CMV-induced Vδ2neg γδ T cells undergo a dramatic and stable expansion after CMV infection, in a conventional “adaptive” manner. Similarly, as CMV-specific CD8+ αβ T cells, they exhibit an effector/memory TEMRA phenotype and cytotoxic effector functions. Activation of Vδ2neg γδ T cells by CMV-infected cells involves the γδ T cell receptor (TCR) and still ill-defined co-stimulatory molecules such as LFA-1. A multiple of Vδ2neg γδ TCR ligands are apparently recognized on CMV-infected cells, the first one identified being the major histocompatibility complex-related molecule endothelial protein C receptor. A singularity of CMV-induced Vδ2neg γδ T cells is to acquire CD16 expression and to exert an antibody-dependent cell-mediated inhibition on CMV replication, which is controlled by a specific cytokine microenvironment. Beyond the well-demonstrated direct anti-CMV effect of Vδ2neg γδ T cells, unexpected indirect effects of these cells have been also observed in the context of kidney transplantation. CMV-induced Vδ2neg γδ T cells have been involved in surveillance of malignancy subsequent to long-term immunosuppression. Moreover, CMV-induced CD16+ γδ T cells are cell effectors of antibody-mediated rejection of kidney transplants, and represent a new physiopathological contribution to the well-known association between CMV infection and poor graft survival. All these basic and clinical studies paved the road to the development of a future γδ T cell-based immunotherapy. In the meantime, γδ T cell monitoring should prove a valuable immunological biomarker in the management of CMV infection.
Collapse
Affiliation(s)
- Lionel Couzi
- Université de Bordeaux , Bordeaux , France ; UMR 5164, Centre National de la Recherche Scientifique , Bordeaux , France ; Service de Néphrologie, Transplantation, Dialyse, Centre Hospitalier Universitaire de Bordeaux , Bordeaux , France
| | - Vincent Pitard
- Université de Bordeaux , Bordeaux , France ; UMR 5164, Centre National de la Recherche Scientifique , Bordeaux , France
| | - Jean-François Moreau
- Université de Bordeaux , Bordeaux , France ; UMR 5164, Centre National de la Recherche Scientifique , Bordeaux , France ; Centre Hospitalier Universitaire de Bordeaux, Laboratoire d'immunologie , Bordeaux , France
| | - Pierre Merville
- Université de Bordeaux , Bordeaux , France ; UMR 5164, Centre National de la Recherche Scientifique , Bordeaux , France ; Service de Néphrologie, Transplantation, Dialyse, Centre Hospitalier Universitaire de Bordeaux , Bordeaux , France
| | - Julie Déchanet-Merville
- Université de Bordeaux , Bordeaux , France ; UMR 5164, Centre National de la Recherche Scientifique , Bordeaux , France
| |
Collapse
|
27
|
Differential expression of leukocyte receptors in disseminated intravascular coagulation: Prognostic value of low protein C receptor expression. Thromb Res 2014; 134:1130-4. [DOI: 10.1016/j.thromres.2014.08.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 08/09/2014] [Accepted: 08/26/2014] [Indexed: 11/17/2022]
|
28
|
Abstract
The coagulation system constitutes an important facet of the unique vascular microenvironment in which primary and metastatic brain tumors evolve and progress. While brain tumor cells express tissue factor (TF) and other effectors of the coagulation system (coagulome), their propensity to induce local and peripheral thrombosis is highly diverse, most dramatic in the case of glioblastoma multiforme (GBM), and less obvious in pediatric tumors. While the immediate medical needs often frame the discussion on current clinical challenges, the coagulation pathway may contribute to brain tumor progression through subtle, context-dependent, and non-coagulant effects, such as induction of inflammation, angiogenesis, or by responding to iatrogenic insults (e.g. surgery). In this regard, the emerging molecular diversity of brain tumor suptypes (e.g. in glioma and medulloblastoma) highlights the link between oncogenic pathways and the tumor repertoire of coagulation system regulators (coagulome). This relationship may influence the mechanisms of spontaneous and therapeutically provoked tumor cell interactions with the coagulation system as a whole. Indeed, oncogenes (EGFR, MET) and tumor suppressors (PTEN, TP53) may alter the expression, activity, and vesicular release of tissue factor (TF), and cause other changes. Conversely, the coagulant microenvironment may also influence the molecular evolution of brain tumor cells through selective and instructive cues. We suggest that effective targeting of the coagulation system in brain tumors should be explored through molecular stratification, stage-specific analysis, and more personalized approaches including thromboprophylaxis and adjuvant treatment aimed at improvement of patient survival.
Collapse
Affiliation(s)
- Esterina D'Asti
- Department of Pediatrics, McGill University. Montreal Children's Hospital, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Yi Fang
- Department of Pediatrics, McGill University. Montreal Children's Hospital, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Janusz Rak
- Department of Pediatrics, McGill University. Montreal Children's Hospital, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
29
|
Mohan Rao LV, Esmon CT, Pendurthi UR. Endothelial cell protein C receptor: a multiliganded and multifunctional receptor. Blood 2014; 124:1553-62. [PMID: 25049281 PMCID: PMC4155268 DOI: 10.1182/blood-2014-05-578328] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 07/08/2014] [Indexed: 01/10/2023] Open
Abstract
Endothelial cell protein C receptor (EPCR) was first identified and isolated as a cellular receptor for protein C on endothelial cells. EPCR plays a crucial role in the protein C anticoagulant pathway by promoting protein C activation. In the last decade, EPCR has received wide attention after it was discovered to play a key role in mediating activated protein C (APC)-induced cytoprotective effects, including antiapoptotic, anti-inflammatory, and barrier stabilization. APC elicits cytoprotective signaling through activation of protease activated receptor-1 (PAR1). Understanding how EPCR-APC induces cytoprotective effects through activation of PAR1, whose activation by thrombin is known to induce a proinflammatory response, has become a major research focus in the field. Recent studies also discovered additional ligands for EPCR, which include factor VIIa, Plasmodium falciparum erythrocyte membrane protein, and a specific variant of the T-cell receptor. These observations open unsuspected new roles for EPCR in hemostasis, malaria pathogenesis, innate immunity, and cancer. Future research on these new discoveries will undoubtedly expand our understanding of the role of EPCR in normal physiology and disease, as well as provide novel insights into mechanisms for EPCR multifunctionality. Comprehensive understanding of EPCR may lead to development of novel therapeutic modalities in treating hemophilia, inflammation, cerebral malaria, and cancer.
Collapse
Affiliation(s)
- L Vijaya Mohan Rao
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center, Tyler, TX; and
| | - Charles T Esmon
- Coagulation Biology Laboratory, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Usha R Pendurthi
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center, Tyler, TX; and
| |
Collapse
|
30
|
McKelvey K, Jackson CJ, Xue M. Activated protein C: A regulator of human skin epidermal keratinocyte function. World J Biol Chem 2014; 5:169-179. [PMID: 24921007 PMCID: PMC4050111 DOI: 10.4331/wjbc.v5.i2.169] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 04/03/2014] [Indexed: 02/05/2023] Open
Abstract
Activated protein C (APC) is a physiological anticoagulant, derived from its precursor protein C (PC). Independent of its anticoagulation, APC possesses strong anti-inflammatory, anti-apoptotic and barrier protective properties which appear to be protective in a number of disorders including chronic wound healing. The epidermis is the outermost skin layer and provides the first line of defence against the external environment. Keratinocytes are the most predominant cells in the epidermis and play a critical role in maintaining epidermal barrier function. PC/APC and its receptor, endothelial protein C receptor (EPCR), once thought to be restricted to the endothelium, are abundantly expressed by skin epidermal keratinocytes. These cells respond to APC by upregulating proliferation, migration and matrix metalloproteinase-2 activity and inhibiting apoptosis/inflammation leading to a wound healing phenotype. APC also increases barrier function of keratinocyte monolayers by promoting the expression of tight junction proteins and re-distributing them to cell-cell contacts. These cytoprotective properties of APC are mediated through EPCR, protease-activated receptors, epidermal growth factor receptor or Tie2. Future preventive and therapeutic uses of APC in skin disorders associated with disruption of barrier function and inflammation look promising. This review will focus on APC’s function in skin epidermis/keratinocytes and its therapeutical potential in skin inflammatory conditions.
Collapse
|
31
|
Ku SK, Kim JA, Bae JS. Piperlonguminine downregulates endothelial protein C receptor shedding in vitro and in vivo. Inflammation 2014; 37:435-42. [PMID: 24127121 DOI: 10.1007/s10753-013-9756-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Endothelial cell protein C receptor (EPCR) plays an important role in coagulation and inflammation. EPCR can be shed from the cell surface, and this is mediated by tumor necrosis factor-α-converting enzyme (TACE). Piperlonguminine (PL), an important component of Piper longum fruits, is known to exhibit antihyperlipidemic, antiplatelet, and antimelanogenesis activities. However, little is known about the effects of PL on EPCR shedding. Here, we investigated this issue by monitoring the effects of PL on phorbol-12-myristate 13-acetate (PMA) and on cecal ligation and puncture (CLP)-mediated EPCR shedding and underlying mechanisms. PL induced potent inhibition of PMA, and CLP induced EPCR shedding through suppression of TACE expression. And treatment with PL resulted in reduced PMA-stimulated phosphorylation of p38, extracellular regulated kinases (ERK) 1/2, and c-Jun N-terminal kinase (JNK). Given these results, PL might have potential as an anti-sEPCR shedding reagent against PMA- and CLP-mediated EPCR shedding.
Collapse
Affiliation(s)
- Sae-Kwang Ku
- Department of Anatomy and Histology, College of Oriental Medicine, Daegu Haany University, Gyeongsan, 712-715, Republic of Korea
| | | | | |
Collapse
|
32
|
Xue M, Shen K, McKelvey K, Li J, Chan YKA, Hatzis V, March L, Little CB, Tonkin M, Jackson CJ. Endothelial protein C receptor-associated invasiveness of rheumatoid synovial fibroblasts is likely driven by group V secretory phospholipase A2. Arthritis Res Ther 2014; 16:R44. [PMID: 24495480 PMCID: PMC3979138 DOI: 10.1186/ar4473] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 01/28/2014] [Indexed: 01/24/2023] Open
Abstract
INTRODUCTION Rheumatoid synovial fibroblasts (RASFs) mediate joint inflammation and destruction in rheumatoid arthritis (RA). Endothelial protein C receptor (EPCR) is a specific receptor for the natural anticoagulant activated protein C (APC). It mediates the cytoprotective properties of APC and is expressed in rheumatoid synovial tissue. A recent report shows that group V secretory phospholipase A2 (sPLA₂V) prevents APC from binding to EPCR in endothelium and inhibits EPCR/APC function. The aim of this study was to investigate the expression and function of EPCR on RASFs. METHODS Human synovial fibroblasts (SFs) were isolated from RA or osteoarthritis (OA) synovial tissues and treated with control, EPCR, or sPLA₂V small interfering RNA (siRNA); recombinant human APC, tumor necrosis factor-alpha (TNF-α), or sPLA₂V. RASF viability and migration/invasion were measured by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide (MTT) and collagen gel migration/invasion assays, respectively, and cartilage degradation by 1,9-dimethylmethylene blue (DMMB) assay in the presence of human OA articular cartilage explants. The expression or activation of cytokines, EPCR, cadherin-11, mitogen-activated protein (MAP) kinases, and nuclear factor-kappa-B (NF-κB) or both were detected by enzyme-linked immunosorbent assay, Western blotting, or immunostaining. RESULTS EPCR was expressed by both OASFs and RASFs but was markedly increased in RASFs. When EPCR was suppressed by siRNA or blocking antibody cell viability, cell invasion and cartilage degradation were reduced by more than 30%. Inflammatory mediators interleukin-1-beta (IL-1β), cadherin-11, and NF-κB were significantly reduced by EPCR suppression under control or TNF-α-stimulated conditions. The expression or activation (or both) of MAP kinases ERK, p38, and JNK were also markedly decreased in cells transfected with EPCR siRNA. Further analysis revealed that sPLA₂V co-localized with EPCR on RASFs. Suppression of sPLA₂V reduced cell viability and cartilage degradation and increased APC binding to RASFs. Conversely, recombinant sPLA₂V increased cartilage degradation, blocked APC binding to RASFs, and could not rescue the effects induced by EPCR suppression. CONCLUSIONS Our results demonstrate that EPCR is overexpressed by RASFs and mediates the aggressive behavior of RASFs. This function of EPCR is contrary to its cytoprotective role in other settings and is likely driven by sPLA₂V.
Collapse
|
33
|
Lagrange J, Li Z, Fassot C, Bourhim M, Louis H, Nguyen Dinh Cat A, Parlakian A, Wahl D, Lacolley P, Jaisser F, Regnault V. Endothelial mineralocorticoid receptor activation enhances endothelial protein C receptor and decreases vascular thrombosis in mice. FASEB J 2014; 28:2062-72. [PMID: 24451386 DOI: 10.1096/fj.13-238188] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Previous studies have shown that aldosterone, which activates the mineralocorticoid receptor (MR), promotes thrombosis in animal models. Our objective was to determine whether MR activation/expression in the vascular endothelium could modify thrombotic risk in vivo and to examine thrombin generation at the surface of aortic endothelial cells (HAECs). MR was conditionally overexpressed in vivo in vascular endothelial cells in mice (MR-EC mice) or stimulated with aldosterone in HAECs. Thrombosis after ferric chloride injury was delayed in MR-EC mice compared with controls as well as in wild-type FVB/NRj mice treated with aldosterone (60 μg/kg/d for 21 d). Thrombin generation in platelet-poor plasma did not differ between MR-EC mice and controls. In MR-EC mice, aortic endothelial cell protein C receptor (EPCR) expression was increased. Aldosterone (10(-8) M) attenuated thrombin generation at the surface of cultured HAECs, and this effect was associated with up-regulation of expression of EPCR, which promotes formation of activated protein C. Aldosterone increases EPCR expression via a transcriptional mechanism involving interaction of MR with the specificity protein 1 site. These findings demonstrate that MR activation acts on endothelial cells to protect against thrombosis in physiological conditions and that MR-mediated EPCR overexpression drives this antithrombotic property through enhancing protein C activation.
Collapse
Affiliation(s)
- Jérémy Lagrange
- 2INSERM U1116, Faculté de Médecine, 9 Avenue de la Forêt de Haye, 54500 Vandoeuvre-les-Nancy, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Xue M, Jackson CJ. Activated protein C and its potential applications in prevention of islet β-cell damage and diabetes. VITAMINS AND HORMONES 2014; 95:323-63. [PMID: 24559924 DOI: 10.1016/b978-0-12-800174-5.00013-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Activated protein C (APC) is derived from its precursor, protein C (PC). Originally thought to be synthesized exclusively by the liver, recent reports have shown that PC is also produced by many other cells including pancreatic islet β cells. APC functions as a physiological anticoagulant with anti-inflammatory, anti-apoptotic, and barrier-stabilizing properties. APC exerts its protective effects via an intriguing mechanism requiring combinations of endothelial PC receptor, protease-activated receptors, epidermal growth factor receptor, Tie2 or CD11b, depending on cell types. Diabetes is a chronic condition resulted from the body's inability to produce and/or properly use insulin. The prevalence of diabetes has risen dramatically and has become one of the major causes of premature mortality and morbidity worldwide. Diabetes prevention is an ideal approach to reduce this burden. Type 1 and type 2 diabetes are the major forms of diabetes mellitus, and both are characterized by an autoimmune response, intraislet inflammation, β-cell apoptosis, and progressive β-cell loss. Protecting β-cell from damage is critical in both prevention and treatment of diabetes. Recent in vitro and animal studies show that APC's strong anti-inflammatory and anti-apoptotic properties are beneficial in preventing β-cell destruction and diabetes in the NOD mouse model of type 1 diabetes. Future preventive and therapeutic uses of APC in diabetes look very promising.
Collapse
Affiliation(s)
- Meilang Xue
- Sutton Arthritis Research Laboratories, Kolling Institute of Medical Research, The University of Sydney at Royal North Shore Hospital, St Leonards, New South Wales, Australia.
| | - Christopher J Jackson
- Sutton Arthritis Research Laboratories, Kolling Institute of Medical Research, The University of Sydney at Royal North Shore Hospital, St Leonards, New South Wales, Australia
| |
Collapse
|
35
|
Ku SK, Lee W, Yoo H, Han CK, Bae JS. Inhibitory effects of epi-sesamin on endothelial protein C receptor shedding in vitro and in vivo. Inflamm Res 2013; 62:895-902. [PMID: 23884236 DOI: 10.1007/s00011-013-0648-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 07/16/2013] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE AND DESIGN Endothelial protein C receptor (EPCR) plays a pivotal role in augmenting Protein C activation by the thrombin-thrombomodulin complex. The activity of EPCR is markedly changed by ectodomain cleavage and release as the soluble protein (sEPCR). The EPCR shedding is mediated by the tumor necrosis factor-α converting enzyme (TACE). Epi-sesamin (ESM), from the roots of Asarum siebodlii, is known to exhibit anti-allergic and anti-fungal activities. However, little is known about the effects of ESM on EPCR shedding. METHODS We investigated this issue by monitoring the effects of ESM on phorbol-12-myristate 13-acetate (PMA), tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and cecal ligation and puncture (CLP)-mediated EPCR shedding. RESULTS Data showed that ESM induced potent inhibition of PMA, TNF-α, IL-1β, and CLP-induced EPCR shedding, likely through suppression of TACE expression. In addition, treatment with ESM resulted in a reduction of PMA-stimulated phosphorylation of p38, extracellular regulated kinases (ERK) 1/2, and c-Jun N-terminal kinase (JNK). CONCLUSIONS Given these results, ESM should be viewed as a candidate therapeutic agent for treatment of various severe vascular inflammatory diseases via inhibition of EPCR shedding.
Collapse
Affiliation(s)
- Sae-Kwang Ku
- Department of Anatomy and Histology, College of Oriental Medicine, Daegu Haany University, Gyeongsan, 712-715, Republic of Korea
| | | | | | | | | |
Collapse
|
36
|
Kallel C, Cohen W, Saut N, Blankenberg S, Schnabel R, Rupprecht HJ, Bickel C, Munzel T, Tregouet DA, Morange PE. Association of soluble endothelial protein C receptor plasma levels and PROCR rs867186 with cardiovascular risk factors and cardiovascular events in coronary artery disease patients: the Athero Gene study. BMC MEDICAL GENETICS 2012; 13:103. [PMID: 23136988 PMCID: PMC3523004 DOI: 10.1186/1471-2350-13-103] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 10/12/2012] [Indexed: 12/23/2022]
Abstract
BACKGROUND Blood coagulation is an essential determinant of coronary artery disease (CAD). Soluble Endothelial Protein C Receptor (sEPCR) may be a biomarker of a hypercoagulable state. We prospectively investigated the relationship between plasma sEPCR levels and the risk of cardiovascular events (CVE). METHODS We measured baseline sEPCR levels in 1673 individuals with CAD (521 with acute coronary syndrome [ACS] and 1152 with stable angina pectoris [SAP]) from the AtheroGene cohort. During a median follow up of 3.7 years, 136 individuals had a CVE. In addition, 891 of these CAD patients were genotyped for the PROCR rs867186 (Ser219Gly) variant. RESULTS At baseline, sEPCR levels were similar in individuals with ACS and SAP (median: 111 vs. 115 ng/mL respectively; p=0.20). Increased sEPCR levels were found to be associated with several cardiovascular risk factors including gender (p=0.006), soluble Tissue Factor levels (p=0.0001), diabetes (p=0.0005), and factors reflecting impaired renal function such as creatinine and cystatin C (p<0.0001). sEPCR levels were not significantly associated with the risk of CVE (median: 110 and 114 ng/mL in individuals with and without future CVE respectively; p=0.68). The rs867186 variant was found to explain 59% of sEPCR levels variability (p<10-200) but did not associate with CVE risk. CONCLUSION Our findings show that in patients with CAD, circulating sEPCR levels are related to classical cardiovascular risk factors and renal impairment but are not related to long-term incidence of CVE.
Collapse
|
37
|
Festoff BW, Li C, Woodhams B, Lynch S. Soluble thrombomodulin levels in plasma of multiple sclerosis patients and their implication. J Neurol Sci 2012; 323:61-5. [PMID: 22967748 DOI: 10.1016/j.jns.2012.08.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 07/27/2012] [Accepted: 08/09/2012] [Indexed: 11/28/2022]
Abstract
Thrombomodulin (TM) on the cell-surface of cerebrovascular endothelial cells (CECs) is released into blood upon CEC damage. TM promotes activation of protein C (APC), an anticoagulant, anti-inflammatory, neuroprotective molecule that protects CECs and impedes inflammatory cell migration across the blood-brain barrier (BBB). Multiple sclerosis (MS) is associated with CEC damage and BBB dysfunction. We evaluated soluble TM (sTM) levels as a biomarker of BBB integrity and whether glatiramer acetate (GA) influenced sTM levels in MS patients. sTM levels quantified by 2-site ELISA from sera of healthy controls and systemic lupus erythematosus (SLE) patients (CEC-damage positive control) were compared with levels from patients with relapsing-remitting (RRMS) or secondary-progressive MS (SPMS), stratified as: RRMS/GA/no relapse, RRMS/GA/in relapse, RRMS no GA/no relapse, RRMS/no GA/in relapse; and SPMS/no GA. Additionally, soluble endothelial protein C receptor (sEPCR) levels were assessed in the non-stratified MS group, SLE patients, and controls. sTM levels were highest in RRMS patients taking GA with or without relapse, followed in decreasing order by SLE, RRMS/no GA/in relapse, SPMS, RRMS/no GA/no relapse, healthy controls. sEPCR levels were highest in MS patients, then SLE, then controls. sTM may be a useful biomarker of BBB integrity in RRMS patients. Further evaluation of sEPCR is needed. The finding that the highest sTM levels were in RRMS patients taking GA is interesting and warrants further investigation.
Collapse
Affiliation(s)
- Barry W Festoff
- Neurobiology Research Laboratory, Veterans Affairs Medical Center, Kansas City, MO 64128, USA.
| | | | | | | |
Collapse
|
38
|
Willcox CR, Pitard V, Netzer S, Couzi L, Salim M, Silberzahn T, Moreau JF, Hayday AC, Willcox BE, Déchanet-Merville J. Cytomegalovirus and tumor stress surveillance by binding of a human γδ T cell antigen receptor to endothelial protein C receptor. Nat Immunol 2012; 13:872-9. [PMID: 22885985 DOI: 10.1038/ni.2394] [Citation(s) in RCA: 237] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Accepted: 07/12/2012] [Indexed: 12/15/2022]
Abstract
T cells bearing γδ T cell antigen receptors (TCRs) function in lymphoid stress surveillance. However, the contribution of γδ TCRs to such responses is unclear. Here we found that the TCR of a human V(γ)4V(δ)5 clone directly bound endothelial protein C receptor (EPCR), which allowed γδ T cells to recognize both endothelial cells targeted by cytomegalovirus and epithelial tumors. EPCR is a major histocompatibility complex-like molecule that binds lipids analogously to the antigen-presenting molecule CD1d. However, the V(γ)4V(δ)5 TCR bound EPCR independently of lipids, in an antibody-like way. Moreover, the recognition of target cells by γδ T cells required a multimolecular stress signature composed of EPCR and costimulatory ligand(s). Our results demonstrate how a γδ TCR mediates recognition of broadly stressed human cells by engaging a stress-regulated self antigen.
Collapse
MESH Headings
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Cytomegalovirus/immunology
- Cytomegalovirus Infections/immunology
- Endothelial Protein C Receptor
- Humans
- Immunoblotting
- Immunologic Surveillance/immunology
- Immunoprecipitation
- Neoplasms, Glandular and Epithelial/immunology
- Protein Binding
- Receptors, Antigen, T-Cell, gamma-delta/chemistry
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Cell Surface/immunology
- Receptors, Cell Surface/metabolism
- Stress, Physiological/immunology
- T-Lymphocyte Subsets/chemistry
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocytes/chemistry
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Carrie R Willcox
- Birmingham Cancer Research UK Cancer Centre, School of Cancer Sciences, University of Birmingham, Birmingham, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Ducros E, Mirshahi SS, Faussat AM, Mirshahi P, Dimicoli S, Tang R, Pardo J, Ibrahim J, Marie JP, Therwath A, Soria J, Mirshahi M. Soluble endothelial protein C receptor (sEPCR) is likely a biomarker of cancer-associated hypercoagulability in human hematologic malignancies. Cancer Med 2012; 1:261-7. [PMID: 23342274 PMCID: PMC3544449 DOI: 10.1002/cam4.11] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 05/14/2012] [Accepted: 05/16/2012] [Indexed: 11/08/2022] Open
Abstract
Elevated plasma level of soluble endothelial protein C receptor (sEPCR) may be an indicator of thrombotic risk. The present study aims to correlate leukemia-associated hypercoagulability to high level plasma sEPCR and proposes its measurement in routine clinical practice. EPCR expressions in leukemic cell lines were determined by flow cytometry, immunocytochemistry, and reverse transcription polymerase chain reaction (RT-PCR). EPCR gene sequence of a candidate cell line HL-60 was also determined. Plasma samples (n = 76) and bone marrow aspirates (n = 72) from 148 patients with hematologic malignancies and 101 healthy volunteers were analyzed by enzyme-linked immunosorbent assay (ELISA) via a retrospective study for sEPCR and D-dimer. All leukemic cell lines were found to express EPCR. Also, HL-60 EPCR gene sequence showed extensive similarities with the endothelial reference gene. All single nucleotide polymorphisms (SNPs) originally described and some new SNPs were revealed in the promoter and intronic regions. Among these patients 67% had plasma sEPCR level higher than the controls (100 ± 28 ng/mL), wherein 16.3% patients had experienced a previous thrombotic event. These patients were divided into: group-1 (n = 45) with amount of plasmatic sEPCR below 100 ng/mL, group-2 (n = 45) where the concentration of sEPCR was between 100 and 200, and group-3 (n = 20) higher than 200 ng/mL. The numbers of thrombotic incidence recorded in each group were four, six, and eight, respectively. These results reveal that EPCR is expressed not only by a wide range of human malignant hematological cells but also the detection of plasma sEPCR levels provides a powerful insight into thrombotic risk assessment in cancer patients, especially when it surpasses 200 ng/mL.
Collapse
Affiliation(s)
- Elodie Ducros
- INSERM, UMRS 872, CRC, Université Pierre et Marie Curie-Paris 6, Université Paris Descartes, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Dendana M, Messaoudi S, Hizem S, Jazia KB, Almawi WY, Gris JC, Mahjoub T. Endothelial protein C receptor 1651C/G polymorphism and soluble endothelial protein C receptor levels in women with idiopathic recurrent miscarriage. Blood Coagul Fibrinolysis 2012; 23:30-4. [PMID: 22036807 DOI: 10.1097/mbc.0b013e328349cae5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
High levels of soluble endothelial protein C receptor (EPCR) induce coagulation dysfunction by inhibiting protein C activation, and activated protein C (APC) activity. We tested whether EPCR 1651C/G promoter variant and changes in plasma soluble EPCR levels are risk factors for idiopathic recurrent spontaneous miscarriage (RSM). A case-control study involving 283 RSM cases and 380 age and BMI-matched control women. EPCR 1651C/G genotyping was performed by PCR-RFLP method. Plasma-soluble EPCR levels were measured with ELISA. The 1651G allele frequency and C/G genotype were significantly higher in RSM cases than controls; none of the cases or control participants was a 1651G/G homozygote. Lower soluble EPCR levels were seen in RSM cases compared to controls, and higher soluble EPCR levels were seen in 1651C/G compared to 1651C/C carriers in cases and controls. Lower soluble EPCR levels were seen in cases, both in 1651C/C (P = 0.0046) and 1651C/G (P = 0.0032) genotype carriers. Multivariate analysis demonstrated strong association of EPCR 1651C/G [P = 0.011; adjusted odds ratio (aOR) (95% confidence interval [CI] = 3.13 (1.31-7.60)], but not soluble EPCR plasma levels [P = 0.067; aOR (95% CI) = 1.01 (1.00-1.10)], with increased RSM risk. In addition, smoking was independently associated with increased RSM risk [P = 0.002; aOR (95% CI) = 2.86 (1.48-5.52)]. EPCR 1651C/G polymorphism and elevated soluble EPCR levels but low soluble EPCR levels increase the risk of idiopathic RSM. Replication studies on other racial groups, and other EPCR gene variants, are warranted.
Collapse
Affiliation(s)
- Maryam Dendana
- Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | | | | | | | | | | | | |
Collapse
|
41
|
Cooper DKC, Ekser B, Burlak C, Ezzelarab M, Hara H, Paris L, Tector AJ, Phelps C, Azimzadeh AM, Ayares D, Robson SC, Pierson RN. Clinical lung xenotransplantation--what donor genetic modifications may be necessary? Xenotransplantation 2012; 19:144-58. [PMID: 22702466 PMCID: PMC3775598 DOI: 10.1111/j.1399-3089.2012.00708.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Barriers to successful lung xenotransplantation appear to be even greater than for other organs. This difficulty may be related to several macro anatomic factors, such as the uniquely fragile lung parenchyma and associated blood supply that results in heightened vulnerability of graft function to segmental or lobar airway flooding caused by loss of vascular integrity (also applicable to allotransplants). There are also micro-anatomic considerations, such as the presence of large numbers of resident inflammatory cells, such as pulmonary intravascular macrophages and natural killer (NK) T cells, and the high levels of von Willebrand factor (vWF) associated with the microvasculature. We have considered what developments would be necessary to allow successful clinical lung xenotransplantation. We suggest this will only be achieved by multiple genetic modifications of the organ-source pig, in particular to render the vasculature resistant to thrombosis. The major problems that require to be overcome are multiple and include (i) the innate immune response (antibody, complement, donor pulmonary and recipient macrophages, monocytes, neutrophils, and NK cells), (ii) the adaptive immune response (T and B cells), (iii) coagulation dysregulation, and (iv) an inflammatory response (e.g., TNF-α, IL-6, HMGB1, C-reactive protein). We propose that the genetic manipulation required to provide normal thromboregulation alone may include the introduction of genes for human thrombomodulin/endothelial protein C-receptor, and/or tissue factor pathway inhibitor, and/or CD39/CD73; the problem of pig vWF may also need to be addressed. It would appear that exploration of every available therapeutic path will be required if lung xenotransplantation is to be successful. To initiate a clinical trial of lung xenotransplantation, even as a bridge to allotransplantation (with a realistic possibility of survival long enough for a human lung allograft to be obtained), significant advances and much experimental work will be required. Nevertheless, with the steadily increasing developments in techniques of genetic engineering of pigs, we are optimistic that the goal of successful clinical lung xenotransplantation can be achieved within the foreseeable future. The optimistic view would be that if experimental pig lung xenotransplantation could be successfully managed, it is likely that clinical application of this and all other forms of xenotransplantation would become more feasible.
Collapse
Affiliation(s)
- David K C Cooper
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Xue M, Dervish S, Harrison LC, Fulcher G, Jackson CJ. Activated protein C inhibits pancreatic islet inflammation, stimulates T regulatory cells, and prevents diabetes in non-obese diabetic (NOD) mice. J Biol Chem 2012; 287:16356-64. [PMID: 22447930 DOI: 10.1074/jbc.m111.325951] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Activated protein C (aPC) is a natural anticoagulant with strong cyto-protective and anti-inflammatory properties. aPC inhibits pancreatic inflammation and preserves functional islets after intraportal transplantation in mice. Whether aPC prevents the onset or development of type 1 diabetes (T1D) is unknown. In this study, when human recombinant aPC was delivered intraperitoneally, twice weekly for 10 weeks (from week 6 to 15) to non-obese diabetic (NOD) mice, a model for T1D, the incidence of diabetes was reduced from 70% (saline control) to 7.6% by 26 weeks of age. Islets of aPC-treated mice exhibited markedly increased expression of insulin, aPC/protein C, endothelial protein C receptor, and matrix metalloproteinase (MMP)-2 when examined by immunostaining. The insulitis score in aPC-treated mice was 50% less than that in control mice. T regulatory cells (Tregs) in the spleen, pancreatic islets, and pancreatic lymph nodes were increased 37, 53, and 59%, respectively, in NOD mice following aPC treatment. These Tregs had potent suppressor function and, after adoptive transfer, delayed diabetes onset in NOD.severe combined immunodeficiency mice. The culture of NOD mouse spleen cells with aPC reduced the secretion of inflammatory cytokines interleukin (IL)-1β and interferon-γ but increased IL-2 and transforming growth factor-β1, two cytokines required for Treg differentiation. In summary, our results indicate that aPC prevents T1D in the NOD mouse. The aPC mechanism of action is complex, involving induction of Treg differentiation, inhibition of inflammation, and possibly direct cyto-protective effects on β cells.
Collapse
Affiliation(s)
- Meilang Xue
- Sutton Arthritis Research Laboratories, University of Sydney at Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia.
| | | | | | | | | |
Collapse
|
43
|
Weiler H. Multiple receptor-mediated functions of activated protein C. Hamostaseologie 2012; 31:185-95. [PMID: 21826371 DOI: 10.5482/ha-1166] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 06/17/2011] [Indexed: 01/08/2023] Open
Abstract
The central effector protease of the protein C pathway, activated protein C (APC), interacts with the endothelial cell protein C receptor, with protease activated receptors (PAR), the apolipoprotein E2 receptor, and integrins to exert multiple effects on haemostasis and immune cell function. Such receptor interactions modify the activation of PC and determine the biological response to endogenous and therapeutically administered APC. This review summarizes the current knowledge about interactions of APC with cell surface-associated receptors, novel substrates such as histones and tissue factor pathway inhibitor, and their implications for the biologic function of APC in the control of coagulation and inflammation.
Collapse
Affiliation(s)
- H Weiler
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee WI 53226, USA.
| |
Collapse
|
44
|
Conway EM. Thrombomodulin and its role in inflammation. Semin Immunopathol 2012; 34:107-25. [PMID: 21805323 DOI: 10.1007/s00281-011-0282-8] [Citation(s) in RCA: 220] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 07/20/2011] [Indexed: 12/30/2022]
Abstract
The goal is to provide an extensive review of the physiologic role of thrombomodulin (TM) in maintaining vascular homeostasis, with a focus on its anti-inflammatory properties. Data were collected from published research. TM is a transmembrane glycoprotein expressed on the surface of all vascular endothelial cells. Expression of TM is tightly regulated to maintain homeostasis and to ensure a rapid and localized hemostatic and inflammatory response to injury. By virtue of its strategic location, its multidomain structure and complex interactions with thrombin, protein C (PC), thrombin activatable fibrinolysis inhibitor (TAFI), complement components, the Lewis Y antigen, and the cytokine HMGB1, TM exhibits a range of physiologically important anti-inflammatory, anti-coagulant, and anti-fibrinolytic properties. TM is an essential cofactor that impacts on multiple biologic processes. Alterations in expression of TM and its partner proteins may be manifest by inflammatory and thrombotic disorders. Administration of soluble forms of TM holds promise as effective therapies for inflammatory diseases, and infections and malignancies that are complicated by disseminated intravascular coagulation.
Collapse
Affiliation(s)
- Edward M Conway
- Division of Hematology-Oncology, Department of Medicine, Centre for Blood Research (CBR), University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
45
|
Affiliation(s)
- Kenneth G Mann
- Department of Biochemistry, Colchester Research Facility Room 235, University of Vermont, Colchester, VT 05446, USA.
| |
Collapse
|
46
|
Kador K, Mamedov T, Schneider M, Subramanian A. Sequential co-immobilization of thrombomodulin and endothelial protein C receptor on polyurethane: activation of protein C. Acta Biomater 2011; 7:2508-17. [PMID: 21414427 DOI: 10.1016/j.actbio.2011.03.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 03/03/2011] [Accepted: 03/11/2011] [Indexed: 10/18/2022]
Abstract
In an effort to control the surface-mediated activation of thrombin and clot formation, proteins and molecules which mimic the anticoagulant properties of the vascular endothelial lining were immobilized on material surfaces. When immobilized on biomaterial surfaces, thrombomodulin (TM), an endothelial glycoprotein that binds thrombin and activates protein C (PC), was shown to generate activated PC (APC) and delay clot formation. However, TM-mediated activation of PC on biomaterial surfaces was shown to be limited by the transport of PC to the surface, with maximum activation obtained at a surface density of ∼40 fmole TM cm(-2). This work investigates surface immobilized with TM and endothelial protein C receptor (EPCR), a natural cofactor to TM which increases the rate of activation of PC on the native endothelium. A sequential and ordered immobilization of TM and EPCR on polyurethane at an enzymatically relevant distance (<10 nm) resulted in higher amounts of APC compared with surfaces with immobilized TM or with TM and EPCR immobilized randomly and at TM surface densities (1400 fmole cm(-2)) which were previously shown to be transport limited. Ordered TM and EPCR samples also showed increased time to clot formation in experiments with platelet-poor plasma, as measured by thromboelastography. Surfaces immobilized with TM and its natural cofactor EPCR at an enzymatically relevant distance are able to overcome transport limitations, increasing anticoagulant activation and time to clot formation.
Collapse
|
47
|
|
48
|
Identification of new carbohydrate and membrane protein antigens in cardiac xenotransplantation. Transplantation 2011; 91:287-92. [PMID: 21119562 PMCID: PMC10022691 DOI: 10.1097/tp.0b013e318203c27d] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND α1,3-Galactosyltransferase gene knockout (GTKO) pigs reduced the significance of antibody to galactose alpha 1,3-galactose (Gal) antigens but did not eliminate delayed xenograft rejection (DXR). We hypothesize that DXR of GTKO organs results from an antibody response to a limited number of non-Gal endothelial cell (EC) membrane antigens. In this study, we screened a retrovirus expression library to identify EC membrane antigens detected after cardiac xenotransplantation. METHODS Expression libraries were made from GT:CD46 and GTKO porcine aortic ECs. Viral stocks were used to infect human embryonic kidney cells (HEK) that were selected by flow cytometry for IgG binding from sensitized cardiac heterotopic xenograft recipients. After three to seven rounds of selection, individual clones were assessed for non-Gal IgG binding. The porcine complementary DNA was recovered by polymerase chain reaction amplification, sequenced, and identified by homology comparisons. RESULTS A total of 199 and 317 clones were analyzed from GT:CD46 and GTKO porcine aortic EC complementary DNA libraries, respectively. Sequence analysis identified porcine CD9, CD46, CD59, and the EC protein C receptor. We also identified porcine annexin A2 and a glycosyltransferase with homology to the human β1,4 N-acetylgalactosaminyl transferase 2 gene. CONCLUSION The identified proteins include key EC functions and suggest that non-Gal antibody responses may compromise EC functions and thereby contribute to DXR. Recovery of the porcine β1,4 N-acetylgalactosaminyl transferase 2 suggests that an antibody response to a SD-like carbohydrate may represent a new carbohydrate moiety involved in xenotransplantation. The identification of these porcine gene products may lead to further donor modification to enhance resistance to DXR and further reduce the level of xenograft antigenicity.
Collapse
|
49
|
Champagne E. γδ T cell receptor ligands and modes of antigen recognition. Arch Immunol Ther Exp (Warsz) 2011; 59:117-37. [PMID: 21298486 DOI: 10.1007/s00005-011-0118-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Accepted: 12/02/2010] [Indexed: 01/03/2023]
Abstract
T lymphocytes expressing the γδ-type of T cell receptors (TCRs) for antigens contribute to all aspects of immune responses, including defenses against viruses, bacteria, parasites and tumors, allergy and autoimmunity. Multiple subsets have been individualized in humans as well as in mice and they appear to recognize in a TCR-dependent manner antigens as diverse as small non-peptidic molecules, soluble or membrane-anchored polypeptides and molecules related to MHC antigens on cell surfaces, implying diverse modes of antigen recognition. We review here the γδ TCR ligands which have been identified along the years and their characteristics, with emphasis on a few systems which have been extensively studied such as human γδ T cells responding to phosphoantigens or murine γδ T cells activated by allogeneic MHC antigens. We discuss a speculative model of antigen recognition involving simultaneous TCR recognition of MHC-like and non-MHC ligands which could fit with most available data and shares many similarities with the classical model of MHC-restricted antigen recognition for peptides or lipids by T cells subsets with αβ-type TCRs.
Collapse
Affiliation(s)
- Eric Champagne
- INSERM U1043/CNRS U5282; Université de Toulouse, UPS, Centre de Physiopathologie de Toulouse Purpan, Toulouse, France.
| |
Collapse
|
50
|
Sen P, Sahoo S, Pendurthi UR, Rao LVM. Zinc modulates the interaction of protein C and activated protein C with endothelial cell protein C receptor. J Biol Chem 2010; 285:20410-20. [PMID: 20413590 DOI: 10.1074/jbc.m110.111575] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Zinc is an essential trace element for human nutrition and is critical to the structure, stability, and function of many proteins. Zinc ions were shown to enhance activation of the intrinsic pathway of coagulation but down-regulate the extrinsic pathway of coagulation. The protein C pathway plays a key role in blood coagulation and inflammation. At present there is no information on whether zinc modulates the protein C pathway. In the present study we found that Zn(2+) enhanced the binding of protein C/activated protein C (APC) to endothelial cell protein C receptor (EPCR) on endothelial cells. Binding kinetics revealed that Zn(2+) increased the binding affinities of protein C/APC to EPCR. Equilibrium dialysis with (65)Zn(2+) revealed that Zn(2+) bound to the Gla domain as well as sites outside of the Gla domain of protein C/APC. Intrinsic fluorescence measurements suggested that Zn(2+) binding induces conformational changes in protein C/APC. Zn(2+) binding to APC inhibited the amidolytic activity of APC, but the inhibition was reversed by Ca(2+). Zn(2+) increased the rate of APC generation on endothelial cells in the presence of physiological concentrations of Ca(2+) but did not further enhance increased APC generation obtained in the presence of physiological concentrations of Mg(2+) with Ca(2+). Zn(2+) had no effect on the anticoagulant activity of APC. Zn(2+) enhanced APC-mediated activation of protease activated receptor 1 and p44/42 MAPK. Overall, our data show that Zn(2+) binds to protein C/APC, which results in conformational changes in protein C/APC that favor their binding to EPCR.
Collapse
Affiliation(s)
- Prosenjit Sen
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, Texas 75708, USA
| | | | | | | |
Collapse
|