1
|
Jeong Y, Yao Y, Yim EKF. Current understanding of intimal hyperplasia and effect of compliance in synthetic small diameter vascular grafts. Biomater Sci 2020; 8:4383-4395. [PMID: 32643723 PMCID: PMC7452756 DOI: 10.1039/d0bm00226g] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Despite much effort, synthetic small diameter vascular grafts still face limited success due to vascular wall thickening known as intimal hyperplasia (IH). Compliance mismatch between graft and native vessels has been proposed to be one of a key mechanical factors of synthetic vascular grafts that could contribute to the formation of IH. While many methods have been developed to determine compliance both in vivo and in vitro, the effects of compliance mismatch still remain uncertain. This review aims to explain the biomechanical factors that are responsible for the formation and development of IH and their relationship with compliance mismatch. Furthermore, this review will address the current methods used to measure compliance both in vitro and in vivo. Lastly, current limitations in understanding the connection between the compliance of vascular grafts and the role it plays in the development and progression of IH will be discussed.
Collapse
Affiliation(s)
- YeJin Jeong
- Department of Chemical engineering, University of Waterloo, 200 University Ave W, Waterloo, ON N2L 3G1, Canada.
| | | | | |
Collapse
|
2
|
Abdallah RT, Keum JS, El-Shewy HM, Lee MH, Wang B, Gooz M, Luttrell DK, Luttrell LM, Jaffa AA. Plasma kallikrein promotes epidermal growth factor receptor transactivation and signaling in vascular smooth muscle through direct activation of protease-activated receptors. J Biol Chem 2010; 285:35206-15. [PMID: 20826789 DOI: 10.1074/jbc.m110.171769] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The kallikrein-kinin system, along with the interlocking renin-angiotensin system, is a key regulator of vascular contractility and injury response. The principal effectors of the kallikrein-kinin system are plasma and tissue kallikreins, proteases that cleave high molecular weight kininogen to produce bradykinin. Most of the cellular actions of kallikrein (KK) are thought to be mediated by bradykinin, which acts via G protein-coupled B1 and B2 bradykinin receptors on VSMCs and endothelial cells. Here, we find that primary aortic vascular smooth muscle but not endothelial cells possess the ability to activate plasma prekallikrein. Surprisingly, exposing VSMCs to prekallikrein leads to activation of the ERK1/2 mitogen-activated protein kinase cascade via a mechanism that requires kallikrein activity but does not involve bradykinin receptors. In transfected HEK293 cells, we find that plasma kallikrein directly activates G protein-coupled protease-activated receptors (PARs) 1 and 2, which possess consensus kallikrein cleavage sites, but not PAR4. In vascular smooth muscles, KK stimulates ADAM (a disintegrin and metalloprotease) 17 activity via a PAR1/2 receptor-dependent mechanism, leading sequentially to release of the endogenous ADAM17 substrates, amphiregulin and tumor necrosis factor-α, metalloprotease-dependent transactivation of epidermal growth factor receptors, and metalloprotease and epidermal growth factor receptor-dependent ERK1/2 activation. These results suggest a novel mechanism of bradykinin-independent kallikrein action that may contribute to the regulation of vascular responses in pathophysiologic states, such as diabetes mellitus.
Collapse
Affiliation(s)
- Rany T Abdallah
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Weber AA, Schrör K. The significance of platelet-derived growth factors for proliferation of vascular smooth muscle cells. Platelets 2010. [DOI: 10.1080/09537109909169169] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
4
|
Radjabi AR, Sawada K, Jagadeeswaran S, Eichbichler A, Kenny HA, Montag A, Bruno K, Lengyel E. Thrombin induces tumor invasion through the induction and association of matrix metalloproteinase-9 and beta1-integrin on the cell surface. J Biol Chem 2008; 283:2822-34. [PMID: 18048360 PMCID: PMC2805198 DOI: 10.1074/jbc.m704855200] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The procoagulatory serine protease, thrombin, is known to induce invasion and metastasis in various cancers, but the mechanisms by which it promotes tumorigenesis are poorly understood. Because the 92-kDa gelatinase (MMP-9) is a known mediator of tumor cell invasion, we sought to determine whether and how thrombin regulates MMP-9. The thrombin receptor, PAR-1, and MMP-9 are expressed in osteosarcomas, as determined by immunohistochemistry. Stimulation of U2-OS osteosarcoma cells with thrombin and a thrombin receptor-activating peptide induced pro-MMP-9 secretion as well as cell surface-associated pro-MMP-9 expression and proteolytic activity. This was paralleled by an increase in MMP-9 mRNA and MMP-9 promoter activity. Thrombin-induced invasion of U2-OS cells through Matrigel was mediated by the phosphatidylinositol 3-kinase signaling pathway and could be inhibited with an MMP-9 antibody. The stimulation of MMP-9 by thrombin was paralleled by an increase in beta1-integrin mRNA and beta1-integrin expression on the cell surface, which was also mediated by phosphatidylinositol 3-kinase and was required for invasion. Thrombin activation induced and co-localized both beta1-integrin and pro-MMP-9 on the cell membrane, as evidenced by co-immunoprecipitation, confocal microscopy, and a protein binding assay. The thrombin-mediated association of these two proteins, as well as thrombin-mediated invasion of U2-OS cells, could be blocked with a cyclic peptide and with an antibody preventing binding of the MMP-9 hemopexin domain to beta1-integrin. These results suggest that thrombin induces expression and association of beta1-integrin with MMP-9 and that the cell surface localization of the protease by the integrin promotes tumor cell invasion.
Collapse
Affiliation(s)
- A. Reza Radjabi
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology, Center for Integrative Science, University of Chicago, Chicago, Illinois 60637
| | - Kenjiro Sawada
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology, Center for Integrative Science, University of Chicago, Chicago, Illinois 60637
| | - Sujatha Jagadeeswaran
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology, Center for Integrative Science, University of Chicago, Chicago, Illinois 60637
| | - Alfred Eichbichler
- Department of Orthopedic Surgery, Bogenhausen Medical Center, Bavaria, 81925 Munich, Germany
| | - Hilary A. Kenny
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology, Center for Integrative Science, University of Chicago, Chicago, Illinois 60637
| | - Anthony Montag
- Department of Pathology University of Chicago, Chicago, Illinois 60637
| | - Katharina Bruno
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois 60637
| | - Ernst Lengyel
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology, Center for Integrative Science, University of Chicago, Chicago, Illinois 60637
- Department of Committee on Cancer Biology, University of Chicago, Chicago, Illinois 60637
| |
Collapse
|
5
|
Vesey DA, Cheung CW, Kruger WA, Poronnik P, Gobe G, Johnson DW. Thrombin stimulates proinflammatory and proliferative responses in primary cultures of human proximal tubule cells. Kidney Int 2005; 67:1315-29. [PMID: 15780084 DOI: 10.1111/j.1523-1755.2005.00209.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Fibrin deposition is frequently observed within the tubulointerstitium in various forms of chronic renal disease. This suggests the presence of active components of the coagulation pathway, which may contribute to the progressive deterioration in renal function. The aim of this study was to investigate the proinflammatory and fibroproliferative effects of the coagulation protease thrombin on human proximal tubular cells (PTC) in culture. METHODS Primary cultures of PTC were established from normal kidney tissue and grown under serum-free conditions with or without thrombin or the protease-activated receptor (PAR) activating peptides TFLLRN-NH(2), SLIGKV-NH(2), and SFLLRN-NH(2) (100 to 400 micromol/L). DNA synthesis (thymidine incorporation), intracellular Ca(2+) mobilization (fura-2 fluorimetry), fibronectin secretion [enzyme-linked immunosorbent assay (ELISA), immunoblotting], monocyte chemoattractant protein-1 (MCP-1) secretion (ELISA), and transforming growth factor-beta1 (TGF-beta1) secretion (ELISA) were measured. Reverse transcription-polymerase chain reaction (RT-PCR) was used to assess PAR mRNA expression in these cells. RESULTS Thrombin enhanced DNA synthesis, fibronectin secretion, MCP-1 secretion, and TGF-beta1 secretion in a concentration-dependent manner. Cell injury [lactate dehydrogenase (LDH) release] and cellular protein levels were unaffected. RT-PCR showed that cultures of PTC expressed mRNA transcripts for the thrombin receptors PAR-1 and PAR-3, but not PAR-4. Thrombin and each of the PAR activating peptides enhanced intracellular calcium mobilization. However, the other effects of thrombin were only fully reproduced by the PAR-2-specific peptide, SLIGKV-NH(2), only partially by SFLLRN-NH(2), (a PAR-1 peptide that can activate PAR-2), and not at all by the PAR-1-specific peptide, TFLLRN-NH(2). Thrombin-induced DNA synthesis, fibronectin, and MCP-1 secretion were unaffected by a TGF-beta neutralizing antibody, the matrix metalloproteinase (MMP) inhibitor, GM6001 and the epidermal growth factor (EGF) receptor kinase inhibitor AG1478. CONCLUSION Thrombin initiates both proinflammatory and fibroproliferative responses in human PTC. These responses which are dependent on its protease activity appear not to be mediated by PAR-1 activation, the autocrine action of thrombin-induced TGF-beta1 secretion, MMP activation, or EGF receptor transactivation. The proinflammatory and fibroproliferative actions of thrombin on human PTC may help explain the extent of tubulointerstitial fibrosis observed in kidney diseases where fibrin deposition is evident.
Collapse
Affiliation(s)
- David A Vesey
- Department of Renal Medicine, University of Queensland, Princess Alexandra Hospital, Brisbane, Queenlsand, Australia.
| | | | | | | | | | | |
Collapse
|
6
|
Song SJ, Pagel CN, Campbell TM, Pike RN, Mackie EJ. The role of protease-activated receptor-1 in bone healing. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:857-68. [PMID: 15743797 PMCID: PMC1602347 DOI: 10.1016/s0002-9440(10)62306-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Protease-activated receptor (PAR)-1, a G-protein-coupled receptor activated by thrombin, mediates thrombin-induced proliferation of osteoblasts. The current study was undertaken to define the role of PAR-1 in bone repair. Holes were drilled transversely through the diaphysis of both tibiae of PAR-1-null and wild-type mice. Three days later, fewer cells had invaded the drill site from adjacent bone marrow in PAR-1-null mice than in wild-type mice, and a lower percentage of cells were labeled with [(3)H]thymidine in PAR-1-null drill sites. More osteoclasts were also observed in the drill site of PAR-1-null mice than in wild-type mice 7 days after drilling. New mineralized bone area was less in the drill site and on the adjacent periosteal surface in PAR-1-null mice than in wild-type mice at day 9. From day 14, no obvious differences could be seen between PAR-1-null and wild-type tibiae. In vitro thrombin caused a dose-dependent increase in proliferation of bone marrow stromal cells isolated from wild-type mice but not PAR-1-null mice. Thrombin stimulated survival of bone marrow stromal cells from both wild-type and PAR-1-null mice, but it did not affect bone marrow stromal cell migration in either wild-type or PAR-1-null cells. The results indicate that PAR-1 plays an early role in bone repair.
Collapse
Affiliation(s)
- Shu Jun Song
- School of Veterinary Science, University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | | | | | |
Collapse
|
7
|
Kanthou C, Benzakour O. Cellular effects and signalling pathways activated by the anti-coagulant factor, protein S, in vascular cells protein S cellular effects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2001; 476:155-66. [PMID: 10949663 DOI: 10.1007/978-1-4615-4221-6_13] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The anticoagulant factor protein S is a secreted vitamin K-dependent gamma-carboxylated protein that is mainly synthesised in the liver but is also made by endothelial cells and megakaryocytes in culture. In previous studies we have shown that protein S acts as a mitogen for cultured human vascular smooth muscle cells. The synthesis and secretion of protein S by endothelial cells suggests that in addition to its role in the coagulation cascade, protein S may be an important autocrine factor implicated in the pathophysiology of the vascular system. The effects of protein S on hVSMC proliferation, migration and survival are discussed. The activation of the components of the MAP kinase pathway, ERK1/2, JNK/SAPK and p38 is also summarised. Binding and chemical cross-linking experiments provided evidence for the existence of a cell surface protein S receptor(s). By virtue of its many cellular effects, it is suggested here that the anticoagulant factor protein S plays an important role in the pathophysiology of the vasculature.
Collapse
Affiliation(s)
- C Kanthou
- Tumour Microcirculation Group, Gray Laboratory Cancer Research Trust, Mount Vernon Hospital, Northwood, Middlesex, UK
| | | |
Collapse
|
8
|
Abstract
Intimal hyperplasia is the process by which the cell population increases within the innermost layer of the arterial wall, such as occurs physiologically during closure of the ductus arteriosus and during involution of the uterus. It also occurs pathologically in pulmonary hypertension, atherosclerosis, after angioplasty, in transplanted organs, and in vein grafts. The underlying causes of intimal hyperplasia are migration and proliferation of vascular smooth muscle cells provoked by injury, inflammation, and stretch. This review discusses, at a molecular level, both the final common pathways leading to smooth muscle migration and proliferation and their (patho)-physiological triggers. It emphasizes the key roles played by growth factors and extracellular matrix-degrading metalloproteinases, which act in concert to remodel the extracellular matrix and permit cell migration and proliferation.
Collapse
Affiliation(s)
- A C Newby
- Bristol Heart Institute, University of Bristol, Bristol Royal Infirmary, Bristol BS2 8HW, UK.
| | | |
Collapse
|
9
|
Edmead C, Kanthou C, Benzakour O. Thrombin activates transcription factors sp1, NF-kappaB, and CREB: importance of the use of phosphatase inhibitors during nuclear protein extraction for the assessment of transcription factor DNA-binding activities. Anal Biochem 1999; 275:180-6. [PMID: 10552902 DOI: 10.1006/abio.1999.4313] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Thrombin, a serine protease, is an important effector of many cellular processes and has been shown to up-regulate the expression of several genes. The mechanisms underlying thrombin-mediated regulation of gene transcription remain poorly understood. The original aim of this work was to study the effects of thrombin on the activation of transcription factors, Sp1, NF-kappaB, and CREB by means of electrophoretic mobility-shift assays (EMSA). However, an inconsistent pattern of results was observed. We raised the possibility that some EMSA results may have been erroneous by the fact that during the nuclear protein extraction and EMSA procedure, transcription factors are dephosphorylated by cellular phosphatases and hence their DNA-binding capacities are modified. Therefore, we have altered the original nuclear extraction protocol by including a mixture of phosphatase inhibitors during protein extraction and subsequent EMSA steps. We show here that this simple measure led to significant changes in both basal and thrombin-induced levels of activation of Sp1 and CREB, but not of NF-kappaB. In light of the data presented here, it would be important to reexamine the conclusions of many reports in which EMSA was used to assess the basal and agonist-induced levels of transcription factor DNA-binding activities.
Collapse
Affiliation(s)
- C Edmead
- Molecular Cell Biology Laboratory, Thrombosis Research Institute, Manresa Road, London, SW3 6LR, United Kingdom
| | | | | |
Collapse
|
10
|
Cucina A, Borrelli V, Di Carlo A, Pagliei S, Corvino V, Santoro-D'Angelo L, Cavallaro A, Sterpetti AV. Thrombin induces production of growth factors from aortic smooth muscle cells. J Surg Res 1999; 82:61-6. [PMID: 10068527 DOI: 10.1006/jsre.1998.5514] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND Myointimal hyperplasia is a common complication of arterial recontructive surgery. The serine protease thrombin has a major role in vessel wall healing and eventual myointimal hyperplasia formation. The aim of this study was to determine the effect of thrombin on the production of PDGF AA and bFGF by arterial smooth muscle cells. MATERIALS AND METHODS Bovine smooth muscle cells were stimulated with thrombin in a serum-free culture. The release of PDGF AA and bFGF was assessed by ELISA. The effect of thrombin on the proliferation of confluent monolayers of bovine smooth muscle cells was determined by tritiated thymidine uptake. RESULTS Smooth muscle cells stimulated with thrombin released more PDGF AA (P < 0.001) and bFGF (P < 0.001) than the control. Addition of anti-PDGF AA and anti-bFGF antibodies to the medium of smooth muscle cell cultures neutralized the mitogenic effect of thrombin (P < 0.001). CONCLUSIONS The findings of our study suggest that thrombin may lead to myointimal hyperplasia formation through induction of PDGF and bFGF production by smooth muscle cells.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal
- Aorta, Thoracic/cytology
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Arteries/injuries
- Arteries/pathology
- Arteries/surgery
- Cattle
- Cell Division/drug effects
- Cells, Cultured
- Culture Media, Conditioned
- DNA/biosynthesis
- Fibroblast Growth Factor 2/biosynthesis
- Fibroblast Growth Factor 2/immunology
- Humans
- Hyperplasia
- Kinetics
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Platelet-Derived Growth Factor/biosynthesis
- Platelet-Derived Growth Factor/immunology
- Thrombin/pharmacology
Collapse
Affiliation(s)
- A Cucina
- Dipartimento di Istologia & Embriologia Medica, University of Rome, La Sapienza, Italy
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Taylor DS, Cheng X, Pawlowski JE, Wallace AR, Ferrer P, Molloy CJ. Epiregulin is a potent vascular smooth muscle cell-derived mitogen induced by angiotensin II, endothelin-1, and thrombin. Proc Natl Acad Sci U S A 1999; 96:1633-8. [PMID: 9990076 PMCID: PMC15542 DOI: 10.1073/pnas.96.4.1633] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Vasoactive GTP-binding protein-coupled receptor agonists such as angiotensin II (AII), endothelin-1 (ET-1), and alpha-thrombin (alpha-Thr) have been reported to indirectly stimulate vascular smooth muscle cell (VSMC) proliferation by regulating the expression of one or more autocrine growth factors. Using ion-exchange, gel-filtration, and reverse-phase chromatographic purification methods, we isolated a major mitogenic protein present in AII-stimulated rat aortic smooth muscle (RASM) cell conditioned medium. Twenty N-terminal amino acids of the purified peptide were identified, and they had 75% amino acid sequence identity with mouse epiregulin, an epidermal growth factor (EGF)-related growth factor. We cloned the cDNA for rat epiregulin to determine its pattern of expression in G-protein-coupled receptor agonist-stimulated cells and confirm its activity as a mitogen. After treatment of RASM cells with AII, ET-1, or alpha-Thr for 1 h, induction of two epiregulin transcripts was observed, including a 4.8-kb transcript and a novel transcript of approximately 1.2 kb. Recombinant rat epiregulin was strongly mitogenic for RASM cells, stimulating DNA synthesis to levels similar to those induced by serum or platelet-derived growth factor and approximately 3-fold above that observed with saturating concentrations of EGF. In addition, epiregulin caused rapid EGF receptor activation in RASM cells. However, relative levels of EGF receptor tyrosine phosphorylation stimulated by epiregulin were less than those induced by EGF or betacellulin. Taken together, these results indicate that epiregulin is a potent VSMC-secreted mitogen, induced in common by AII, ET-1, and alpha-Thr, that may contribute to VSMC proliferation and vascular remodeling stimulated by vasoactive agonists.
Collapse
MESH Headings
- 3T3 Cells
- Amino Acid Sequence
- Angiotensin II/pharmacology
- Animals
- Aorta
- Base Sequence
- COS Cells
- Cells, Cultured
- Cloning, Molecular
- Culture Media, Conditioned
- DNA/biosynthesis
- Endothelin-1/pharmacology
- Endothelin-1/physiology
- Epidermal Growth Factor/chemistry
- Epidermal Growth Factor/genetics
- Epidermal Growth Factor/pharmacology
- Epiregulin
- ErbB Receptors/drug effects
- ErbB Receptors/physiology
- Humans
- Mice
- Molecular Sequence Data
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Rats
- Recombinant Proteins/biosynthesis
- Recombinant Proteins/chemistry
- Recombinant Proteins/pharmacology
- Sequence Alignment
- Sequence Homology, Amino Acid
- Thrombin/pharmacology
- Thrombin/physiology
- Transfection
Collapse
Affiliation(s)
- D S Taylor
- Cardiovascular Drug Discovery, Bristol-Myers Squibb Pharmaceutical Research Institute, Route 206 and Province Line Road, Princeton, NJ 08543, USA
| | | | | | | | | | | |
Collapse
|
12
|
Abstract
The serine protease, thrombin, is both a potent agonist for platelet aggregation and a mitogen inducing the proliferation of other cell types. Many cellular responses to thrombin are mediated by a G-protein–coupled thrombin receptor (protease-activated receptor-1, PAR-1). This represents the prototype of a new family of proteolytically cleaved receptors that includes PAR-2 and the recently identified PAR-3. Like PAR-1, PAR-3 is a potential thrombin receptor. Their similar gene structure, mechanism of activation, and colocalization to 5q13 raises the question of a common evolutionary origin and of their belonging to a clustered gene family. Construction of a physical map of the 5q13 region by pulsed-field gel electrophoresis (PFGE) has allowed us to identify six potential CpG islands and to establish a linkage of the PAR genes. Southern blot analysis showed that they were in a cluster on a 560-kb Asc I fragment, in the order PAR-2, PAR-1, and PAR-3. PAR-1 and PAR-2 genes were contained within the identical 240-kb Not I fragment, thus confirming a tight linkage between them. The localization of other CpG islands suggested that more PAR-family genes may be present.
Collapse
|
13
|
Déry O, Corvera CU, Steinhoff M, Bunnett NW. Proteinase-activated receptors: novel mechanisms of signaling by serine proteases. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 274:C1429-52. [PMID: 9696685 DOI: 10.1152/ajpcell.1998.274.6.c1429] [Citation(s) in RCA: 596] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Although serine proteases are usually considered to act principally as degradative enzymes, certain proteases are signaling molecules that specifically regulate cells by cleaving and triggering members of a new family of proteinase-activated receptors (PARs). There are three members of this family, PAR-1 and PAR-3, which are receptors for thrombin, and PAR-2, a receptor for trypsin and mast cell tryptase. Proteases cleave within the extracellular NH2-terminus of their receptors to expose a new NH2-terminus. Specific residues within this tethered ligand domain interact with extracellular domains of the cleaved receptor, resulting in activation. In common with many G protein-coupled receptors, PARs couple to multiple G proteins and thereby activate many parallel mechanisms of signal transduction. PARs are expressed in multiple tissues by a wide variety of cells, where they are involved in several pathophysiological processes, including growth and development, mitogenesis, and inflammation. Because the cleaved receptor is physically coupled to its agonist, efficient mechanisms exist to terminate signaling and prevent uncontrolled stimulation. These include cleavage of the tethered ligand, receptor phosphorylation and uncoupling from G proteins, and endocytosis and lysosomal degradation of activated receptors.
Collapse
Affiliation(s)
- O Déry
- Department of Surgery, University of California, San Francisco 94143-0660, USA
| | | | | | | |
Collapse
|
14
|
Kanthou C, Kakkar VV, Benzakour O. Cellular and Molecular Effects of Thrombin in the Vascular System. Angiogenesis 1998. [DOI: 10.1007/978-1-4757-9185-3_27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
15
|
Chang MC, Lan WH, Chan CP, Lin CP, Hsieh CC, Jeng JH. Serine protease activity is essential for thrombin-induced protein synthesis in cultured human dental pulp cells: modulation roles of prostaglandin E2. J Oral Pathol Med 1998; 27:23-9. [PMID: 9466731 DOI: 10.1111/j.1600-0714.1998.tb02086.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Irritations and injuries to the dental pulp usually lead to different degrees of pulpal inflammation. To investigate the roles of thrombin and prostaglandins in the healing and inflammatory processes of dental pulp as well as their effects on pulpal protein synthesis, human dental pulp cell cultures were established and their protein production was measured with or without the presence of exogenous thrombin and prostaglandins. At concentrations of 1-25 U/ml, alpha-thrombin increased the protein synthesis to 1.4-2.3 fold over the vehicle control. On the contrary, 0.1 microg/ml of prostaglandin E1 (PGE1) suppressed protein synthesis by 60%. Prostaglandin E2 (PGE2) also inhibited protein synthesis with an IC50 of 0.4 microg/ml. The stimulatory effects of thrombin (10 U/ml) can be inhibited by antithrombin III (2 U/ml) (a natural thrombin inhibitor) with heparin (2 U/ml), PPACK (D-Phe-Pro-ArgCH2Cl) (20-50 microg/ml) (a serine protease inhibitor), and PGE2 (0.5-1.0 microg/ml). Moreover, TRAP (20-40 microg/ml), a thrombin receptor agonist peptide, also exerted a stimulatory effect (1.21-1.37 fold). In conclusion, thrombin-induced protein synthesis by pulp cells is dependent on proteolytic activity, but not on binding to receptors. Both PGE1 and PGE2 exert suppressive effects on protein synthesis, indicating that interactions between thrombin and prostaglandins are important in regulating the inflammation, repair and regeneration of pulp tissue following injury.
Collapse
Affiliation(s)
- M C Chang
- Team of Biomedical Science, Chang-Gung Institute of Nursing, Taoyuan, Taiwan
| | | | | | | | | | | |
Collapse
|
16
|
Kanse SM, Benzakour O, Kanthou C, Kost C, Lijnen HR, Preissner KT. Induction of vascular SMC proliferation by urokinase indicates a novel mechanism of action in vasoproliferative disorders. Arterioscler Thromb Vasc Biol 1997; 17:2848-54. [PMID: 9409265 DOI: 10.1161/01.atv.17.11.2848] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The urokinase-type plasminogen activator (UPA) and its receptor are expressed in the vasculature and are involved in cell migration and remodeling of the extracellular matrix in the neointima. Vessels with atherosclerosis or neointimal hyperplasia, when compared with normal vessels, contain high UPA activity as well as increased levels of UPA receptor. In this study, we have identified the stimulation of vascular smooth muscle cell proliferation as a novel activity for UPA in the vessel wall. High-molecular-weight-UPA (12-200 nmol/L range) stimulated DNA synthesis and cell proliferation, which was half that induced by fetal calf serum or by platelet-derived growth factor-BB. UPA did not induce growth of endothelial cells, and tissue-type plasminogen activator showed no activity on either cell type. Induction of proliferation required the complete UPA molecule but was independent of the proteolytic activity of UPA, whereas neither the amino-terminal fragment nor the catalytic domain by itself was mitogenic. UPA also stimulated c-fos/c-myc mRNA expression and mitogen-activated protein kinase activity in smooth muscle cells. Blocking monoclonal antibodies against the UPA receptor and the enzymatic removal of receptors were ineffective in inhibiting the mitogenic effect of UPA, suggesting a UPA receptor-independent mechanism. Thus, we provide evidence for a novel function of UPA on vascular smooth muscle cell proliferation that, together with its previously documented involvement in regulating pericellular proteolysis-related events and cell migration, provides additional evidence for a role in the pathogenesis of atherosclerosis/restenosis.
Collapse
MESH Headings
- Aorta/cytology
- Calcium-Calmodulin-Dependent Protein Kinases/metabolism
- Cell Division/drug effects
- Cells, Cultured
- DNA Replication/drug effects
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Gene Expression Regulation/drug effects
- Genes, fos/drug effects
- Genes, myc/drug effects
- Growth Substances/pharmacology
- Humans
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Receptors, Cell Surface/physiology
- Receptors, Urokinase Plasminogen Activator
- Saphenous Vein/cytology
- Umbilical Veins/cytology
- Urokinase-Type Plasminogen Activator/pharmacology
Collapse
Affiliation(s)
- S M Kanse
- Max-Planck-Institute, Kerckhoff-Klinik, Bad Nauheim, Germany.
| | | | | | | | | | | |
Collapse
|
17
|
Duhamel-Clérin E, Orvain C, Lanza F, Cazenave JP, Klein-Soyer C. Thrombin receptor-mediated increase of two matrix metalloproteinases, MMP-1 and MMP-3, in human endothelial cells. Arterioscler Thromb Vasc Biol 1997; 17:1931-8. [PMID: 9351356 DOI: 10.1161/01.atv.17.10.1931] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Matrix metalloproteinases (MMPs) are responsible for the degradation of extracellular matrix components and are secreted by a variety of cells including human endothelial cells. Because alpha-thrombin is known to interact with matrix components and has been shown to activate latent MMP-2 in human umbilical vein endothelial cells, we investigated whether human alpha-thrombin could also regulate other MMPs secreted by the human saphenous vein or mammary artery endothelial cells (EC). After treatment of EC with increasing concentrations of thrombin for different periods of time, a significantly higher gelatinolytic activity of both MMP-1 and MMP-3 was observed in addition to MMP-2 activation. The effect of thrombin was time and dose-dependent, reaching a maximum at 24 hours. After treatment with 5 NIH U/ml thrombin for 24 hours, Western blotting revealed 9.5- and 4.4-fold increases over control values for MMP-3 and MMP-1, respectively. The synthetic thrombin receptor agonist peptide SFLLRNPNDKYEPF fully reproduced the action of thrombin, whereas chemical inactivation of the catalytic site of thrombin abolished its effect on MMP-1 and MMP-3. Thrombin and SFLLRNPNDKYEPF both induced MMP-3 mRNA synthesis but had no significant influence on constitutive MMP-1 mRNA levels. These results demonstrate that thrombin not only activates latent MMP-2 but also modulates MMP-1 and MMP-3 production in EC, this latter effect being mediated by the G-protein-coupled thrombin receptor. Hence, our present data provide evidence to support the suspected role of thrombin in tissue remodeling and angiogenesis.
Collapse
Affiliation(s)
- E Duhamel-Clérin
- INSERM U. 311, Establissement de Transfusion Sanguine de Strasbourg, France
| | | | | | | | | |
Collapse
|
18
|
Bretschneider E, Wittpoth M, Weber AA, Glusa E, Schrör K. Thrombin but not thrombin receptor activating peptide is mitogenic for coronary artery smooth muscle cells. Thromb Res 1997; 87:493-7. [PMID: 9306623 DOI: 10.1016/s0049-3848(97)00164-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- E Bretschneider
- Zentrum für Vaskuläre Biologie und Medizin Erfurt der Friedrich-Schiller-Universität Jena, Germany
| | | | | | | | | |
Collapse
|
19
|
Bretschneider E, Wittpoth M, Weber AA, Glusa E, Schrör K. Activation of NFkappaB is essential but not sufficient to stimulate mitogenesis of vascular smooth muscle cells. Biochem Biophys Res Commun 1997; 235:365-8. [PMID: 9199198 DOI: 10.1006/bbrc.1997.6788] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
This study investigates the role of the transcription factor NFkappaB in thrombin- and thrombin receptor activating peptide (TRAP, SFLLRNPNDKYEPYF)-induced mitogenesis of cultured bovine coronary artery smooth muscle cells (SMC). Stimulation of resting cells by thrombin (10 nM) or TRAP (10-100 microM) resulted in a comparable time-dependent activation of NFkappaB as detected by Western blotting and electrophoretic mobility shift assay (EMSA) of nuclear extracts. The NFkappaB activation was antagonized by N-acetyl-L-cysteine (20 mM) and pentoxifylline (0.5 mM). Thrombin caused a 3-4-fold increase in [3H]thymidine incorporation within 24 h which was prevented by inhibitors of NFkappaB activation. In contrast, TRAP did not cause any mitogenic response. These results demonstrate that activation of NFkappaB is an essential but not a sufficient signal for SMC mitogenesis.
Collapse
Affiliation(s)
- E Bretschneider
- Zentrum für Vaskuläre Biologie und Medizin Erfurt der Friedrich-Schiller-Universität Jena, Germany
| | | | | | | | | |
Collapse
|