1
|
Stefanenko M, Fedoriuk M, Mamenko M, Semenikhina M, Nowling TK, Lipschutz JH, Maximyuk O, Staruschenko A, Palygin O. PAR1-mediated Non-periodical Synchronized Calcium Oscillations in Human Mesangial Cells. FUNCTION 2024; 5:zqae030. [PMID: 38984988 PMCID: PMC11384906 DOI: 10.1093/function/zqae030] [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: 02/05/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 07/11/2024] Open
Abstract
Mesangial cells offer structural support to the glomerular tuft and regulate glomerular capillary flow through their contractile capabilities. These cells undergo phenotypic changes, such as proliferation and mesangial expansion, resulting in abnormal glomerular tuft formation and reduced capillary loops. Such adaptation to the changing environment is commonly associated with various glomerular diseases, including diabetic nephropathy and glomerulonephritis. Thrombin-induced mesangial remodeling was found in diabetic patients, and expression of the corresponding protease-activated receptors (PARs) in the renal mesangium was reported. However, the functional PAR-mediated signaling in mesangial cells was not examined. This study investigated protease-activated mechanisms regulating mesangial cell calcium waves that may play an essential role in the mesangial proliferation or constriction of the arteriolar cells. Our results indicate that coagulation proteases such as thrombin induce synchronized oscillations in cytoplasmic Ca2+ concentration of mesangial cells. The oscillations required PAR1 G-protein coupled receptors-related activation, but not a PAR4, and were further mediated presumably through store-operated calcium entry and transient receptor potential canonical 3 (TRPC3) channel activity. Understanding thrombin signaling pathways and their relation to mesangial cells, contractile or synthetic (proliferative) phenotype may play a role in the development of chronic kidney disease and requires further investigation.
Collapse
Affiliation(s)
- Mariia Stefanenko
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, Kyiv 01024, Ukraine
| | - Mykhailo Fedoriuk
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Mykola Mamenko
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Marharyta Semenikhina
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Tamara K Nowling
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Joshua H Lipschutz
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Medicine, Ralph H. Johnson VAMC, Charleston, SC 29401, USA
| | - Oleksandr Maximyuk
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, Kyiv 01024, Ukraine
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33602, USA
- James A. Haley Veterans’ Hospital, Tampa, FL 33612, USA
| | - Oleg Palygin
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
2
|
Mandl–Weber S, Haslinger B, Sitter T. Thrombin Upregulates Production of Plasminogen Activator Inhibitor Type 1 in Human Peritoneal Mesothelial Cells. Perit Dial Int 2020. [DOI: 10.1177/089686089901900407] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
ObjectiveTo determine the influence of thrombin, which is generated intraperitoneally during peritoneal dialysis, on the synthesis of fibrinolytic system components in human peritoneal mesothelial cells (HMC).MethodsConfluently grown HMC, isolated from the omental tissue, were used in the experiments. Conditioned media were obtained by incubating cells with serum-free M199 containing the appropriate concentration of the test compound. Tissue type plasminogen activator (tPA) and plasminogen activator inhibitor type 1 (PAI-1) antigen concentrations were measured by ELISA. Northern blot analysis was conducted for mRNA expression experiments. To test thrombin specificity, we used the thrombin inhibitor hirudin. The protein kinase C (PKC) inhibitor Ro 31-8220 was inserted to examine whether the effect of thrombin depends on PKC activity.ResultsThrombin increased PAI-1 antigen in the conditioned media of HMC in a time- and concentration-dependent manner. After 24 hours incubation, PAI-1 levels increased from 350 ± 30 ng/105cells in control conditions to 620 ± 30 ng/105cells in HMC exposed to 5 U/mL thrombin ( n = 8, p < 0.05). In contrast, there was no effect of thrombin on tPA antigen levels. An increase of PAI-1 mRNA expression was also observed by Northern blot hybridization. Hirudin (10 U/mL) inhibited the thrombin-induced increase in PAI-1 synthesis. In addition, a complete inhibition of the stimulating effect of thrombin on PAI-1 synthesis was obtained by blocking PKC activity with Ro 31-8220 (3 μmol/L).ConclusionsThrombin increases PAI-1 synthesis in HMC via a PKC-dependent mechanism. Thereby the synthesis of tPA is not affected. Thus, thrombin may not only promote fibrin formation in the peritoneal cavity, but may also inhibit fibrin degradation by release of free PAI-1 from HMC.
Collapse
Affiliation(s)
- Sonja Mandl–Weber
- Medizinische Klinik, Klinikum Innenstadt, Ludwig Maximilians Universität, Munich, Germany
| | - Bettina Haslinger
- Medizinische Klinik, Klinikum Innenstadt, Ludwig Maximilians Universität, Munich, Germany
| | - Thomas Sitter
- Medizinische Klinik, Klinikum Innenstadt, Ludwig Maximilians Universität, Munich, Germany
| |
Collapse
|
3
|
Ceciliani F, Lecchi C. The Immune Functions of α 1 Acid Glycoprotein. Curr Protein Pept Sci 2019; 20:505-524. [PMID: 30950347 DOI: 10.2174/1389203720666190405101138] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 11/20/2018] [Accepted: 11/27/2018] [Indexed: 12/20/2022]
Abstract
α1-acid glycoprotein (orosomucoid, AGP) is an Acute Phase Protein produced by liver and peripheral tissues in response to systemic reaction to inflammation. AGP functions have been studied mostly in human, cattle and fish, although the protein has been also found in many mammalian species and birds. AGP fulfils at least two set of functions, which are apparently different from each other but in fact intimately linked. On one hand, AGP is an immunomodulatory protein. On the other hand, AGP is one of the most important binding proteins in plasma and, beside modulating pharmacokinetics and pharmacodynamics of many drugs, it is also able to bind and transport several endogen ligands related to inflammation. The focus of this review is the immunomodulatory activity of AGP. This protein regulates every single event related to inflammation, including binding of pathogens and modulating white blood cells activity throughout the entire leukocyte attacking sequence. The regulation of AGP activity is complex: the inflammation induces not only an increase in AGP serum concentration, but also a qualitative change in its carbohydrate moiety, generating a multitude of glycoforms, each of them with different, and sometimes opposite and contradictory, activities. We also present the most recent findings about the relationship between AGP and adipose tissue: AGP interacts with leptin receptor and, given its immunomodulatory function, it may be included among the potential players in the field of immunometabolism.
Collapse
Affiliation(s)
- Fabrizio Ceciliani
- Department of Veterinary Medicine, Universita degli Studi di Milano, Milano, Italy
| | - Cristina Lecchi
- Department of Veterinary Medicine, Universita degli Studi di Milano, Milano, Italy
| |
Collapse
|
4
|
Huetsch J, Shimoda LA. Na(+)/H(+) exchange and hypoxic pulmonary hypertension. Pulm Circ 2015; 5:228-43. [PMID: 26064449 DOI: 10.1086/680213] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 07/22/2014] [Indexed: 12/24/2022] Open
Abstract
Intracellular pH (pHi) homeostasis is key to the functioning of vascular smooth muscle cells, including pulmonary artery smooth muscle cells (PASMCs). Sodium-hydrogen exchange (NHE) is an important contributor to pHi control in PASMCs. In this review, we examine the role of NHE in PASMC function, in both physiologic and pathologic conditions. In particular, we focus on the contribution of NHE to the PASMC response to hypoxia, considering both acute hypoxic pulmonary vasoconstriction and the development of pulmonary vascular remodeling and pulmonary hypertension in response to chronic hypoxia. Hypoxic pulmonary hypertension remains a disease with limited therapeutic options. Thus, this review explores past efforts at disrupting NHE signaling and discusses the therapeutic potential that such efforts may have in the field of pulmonary hypertension.
Collapse
Affiliation(s)
- John Huetsch
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21224, USA
| | - Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21224, USA
| |
Collapse
|
5
|
Su CM, Hsu CJ, Tsai CH, Huang CY, Wang SW, Tang CH. Resistin Promotes Angiogenesis in Endothelial Progenitor Cells Through Inhibition of MicroRNA206: Potential Implications for Rheumatoid Arthritis. Stem Cells 2015; 33:2243-55. [PMID: 25828083 DOI: 10.1002/stem.2024] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 03/11/2015] [Indexed: 12/15/2022]
Abstract
Endothelial progenitor cells (EPCs) promote angiogenesis and are therefore key contributors to a wide variety of angiogenesis-related autoimmune diseases such as rheumatoid arthritis (RA). However, the signaling mechanisms through which these progenitor cells influence RA pathogenesis remain unknown. The aim of this study was to examine whether resistin plays a role in the pathogenesis of and angiogenesis associated with RA by circulating EPCs. We found that levels of resistin in synovial fluid and tissue from patients with RA and from mice with collagen-induced arthritis were overexpressed and promoted the homing of EPCs into the synovium, thereby inducing angiogenesis. EPCs isolated from healthy donors were used to investigate the signal transduction pathway underlying EPC migration and tube formation after treatment with resistin. We found that resistin directly induced a significant increase in expression of vascular endothelial growth factor (VEGF) in EPCs. We also found that the expression of microRNA-206 (miR-206) was negatively correlated with the expression of resistin during EPC-mediated angiogenesis. Notably, the increased expression of VEGF was associated with decreased binding of miR-206 to the VEGF-A 3' untranslated region through protein kinase C delta-dependent AMP-activated protein kinase signaling pathway. Moreover, blockade of resistin reduced EPC homing into synovial fluid and angiogenesis in vivo. Taken together, our study is the first to demonstrate that resistin promotes EPCs homing into the synovium during RA angiogenesis via a signal transduction pathway that involves VEGF expression in primary EPCs. These findings provide support for resistin as a therapeutic target for the patients with RA. Stem Cells 2015;33:2243-2255.
Collapse
Affiliation(s)
- Chen-Ming Su
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chin-Jung Hsu
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University, Taichung, Taiwan
| | - Chun-Hao Tsai
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University, Taichung, Taiwan
| | - Chun-Yin Huang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University Beigang Hospital, Yun-Lin County, Taiwan
| | - Shih-Wei Wang
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
6
|
Lin CY, Hung SY, Chen HT, Tsou HK, Fong YC, Wang SW, Tang CH. Brain-derived neurotrophic factor increases vascular endothelial growth factor expression and enhances angiogenesis in human chondrosarcoma cells. Biochem Pharmacol 2014; 91:522-33. [PMID: 25150213 DOI: 10.1016/j.bcp.2014.08.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 08/11/2014] [Accepted: 08/11/2014] [Indexed: 01/07/2023]
Abstract
Chondrosarcomas are a type of primary malignant bone cancer, with a potent capacity for local invasion and distant metastasis. Brain-derived neurotrophic factor (BDNF) is commonly upregulated during neurogenesis. The aim of the present study was to examine the mechanism involved in BDNF-mediated vascular endothelial growth factor (VEGF) expression and angiogenesis in human chondrosarcoma cells. Here, we knocked down BDNF expression in chondrosarcoma cells and assessed their capacity to control VEGF expression and angiogenesis in vitro and in vivo. We found knockdown of BDNF decreased VEGF expression and abolished chondrosarcoma conditional medium-mediated angiogenesis in vitro as well as angiogenesis effects in vivo in the chick chorioallantoic membrane and Matrigel plug nude mouse models. In addition, in the xenograft tumor angiogenesis model, the knockdown of BDNF significantly reduced tumor growth and tumor-associated angiogenesis. BDNF increased VEGF expression and angiogenesis through the TrkB receptor, PLCγ, PKCα, and the HIF-1α signaling pathway. Finally, we analyzed samples from chondrosarcoma patients by immunohistochemical staining. The expression of BDNF and VEGF protein in 56 chondrosarcoma patients was significantly higher than in normal cartilage. In addition, the high level of BDNF expression correlated strongly with VEGF expression and tumor stage. Taken together, our results indicate that BDNF increases VEGF expression and enhances angiogenesis through a signal transduction pathway that involves the TrkB receptor, PLCγ, PKCα, and the HIF-1α. Therefore, BDNF may represent a novel target for anti-angiogenic therapy for human chondrosarcoma.
Collapse
Affiliation(s)
- Chih-Yang Lin
- Graduate Institute of Basic Medical Science, China Medical University, No. 91, Hsueh-Shih Road, Taichung, Taiwan
| | - Shih-Ya Hung
- Epigenome Research Center, China Medical University Hospital, Taichung, Taiwan; Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Hsien-Te Chen
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan; Department of Materials Science and Engineering, Feng Chia University, Taichung, Taiwan
| | - Hsi-Kai Tsou
- Department of Materials Science and Engineering, Feng Chia University, Taichung, Taiwan; Department of Neurosurgery, Taichung Veterans General Hospital, Taichung, Taiwan; Department of Early Childhood Care and Education, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli County, Taiwan
| | - Yi-Chin Fong
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Shih-Wei Wang
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, No. 91, Hsueh-Shih Road, Taichung, Taiwan; Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan; Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan.
| |
Collapse
|
7
|
Berberine Reduces the Metastasis of Chondrosarcoma by Modulating the α v β 3 Integrin and the PKC δ , c-Src, and AP-1 Signaling Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:423164. [PMID: 24027594 PMCID: PMC3763569 DOI: 10.1155/2013/423164] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 07/16/2013] [Indexed: 02/06/2023]
Abstract
Chondrosarcoma is a primary malignant bone cancer, with a potent capacity to invade locally and cause distant metastasis, especially to the lungs. Patients diagnosed with chondrosarcoma have poor prognosis. Berberine, an active component of the Ranunculaceae and Papaveraceae families of plant, has been proven to induce tumor apoptosis and to prevent the metastasis of cancer cells. However, the effects of berberine in human chondrosarcoma are largely unknown. In this study, we found that berberine did not induce cell apoptosis in human primary chondrocytes and chondrosarcoma cells. However, at noncytotoxic concentrations, berberine reduced the migration and invasion of chondrosarcoma cancer cells. Integrins are the major adhesive molecules in mammalian cells and have been associated with the metastasis of cancer cells. We also found that incubation of chondrosarcoma cells with berberine reduced mRNA transcription for, and cell surface expression of, the αvβ3 integrin, with additional inhibitory effects on PKCδ, c-Src, and NF-κB activation. Thus, berberine may be a novel antimetastasis agent for the treatment of metastatic chondrosarcoma.
Collapse
|
8
|
Lin TH, Liu HH, Tsai TH, Chen CC, Hsieh TF, Lee SS, Lee YJ, Chen WC, Tang CH. CCL2 increases αvβ3 integrin expression and subsequently promotes prostate cancer migration. Biochim Biophys Acta Gen Subj 2013; 1830:4917-27. [PMID: 23845726 DOI: 10.1016/j.bbagen.2013.06.033] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 05/20/2013] [Accepted: 06/28/2013] [Indexed: 01/14/2023]
Abstract
BACKGROUND Chemokine ligand 2 (CCL2), also known as monocyte chemoattractant protein-1 (MCP-1), belongs to the CC chemokine family which is associated with the disease status and outcomes of cancers. Prostate cancer is the most commonly diagnosed malignancy in men and shows a predilection for metastasis to the bone. However, the effect of CCL2 on human prostate cancer cells is largely unknown. The aim of this study was to examine the role of CCL2 in integrin expression and migratory activity in prostate cancers. METHODS Prostate cancer migration was examined using Transwell, wound healing, and invasion assay. The PKCδ and c-Src phosphorylations were examined by using western blotting. The qPCR was used to examine the mRNA expression of integrins. A transient transfection protocol was used to examine AP-1 activity. RESULTS Stimulation of prostate cancer cell lines (PC3, DU145, and LNCaP) induced migration and expression of integrin αvβ3. Treatment of cells with αvβ3 antibody or siRNA abolished CCL2-increased cell migration. CCL2-increased migration and integrin expression were diminished by CCR2 but not by CCR4 inhibitors, suggesting that the CCR2 receptor is involved in CCL2-promoted prostate cancer migration. CCL2 activated a signal transduction pathway that includes PKCδ, c-Src, and AP-1. Reagents that inhibit specific components of this pathway each diminished the ability of CCL2 to effect cell migration and integrin expression. CONCLUSIONS Interaction between CCL2 and CCR2 enhances migration of prostate cancer cells through an increase in αvβ3 integrin production. GENERAL SIGNIFICANCE CCL2 is a critical factor of prostate cancer metastasis.
Collapse
Affiliation(s)
- Tien-Huang Lin
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Liu JF, Hou SM, Tsai CH, Huang CY, Hsu CJ, Tang CH. CCN4 induces vascular cell adhesion molecule-1 expression in human synovial fibroblasts and promotes monocyte adhesion. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:966-75. [PMID: 23313051 DOI: 10.1016/j.bbamcr.2012.12.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 12/13/2012] [Accepted: 12/26/2012] [Indexed: 01/05/2023]
Abstract
CCN4 is a cysteine-rich protein that belongs to the Cyr61, CTGF, Nov family of matricellular proteins. Here, we investigated the intracellular signaling pathways involved in CCN4-induced vascular cell adhesion molecule-1 expression in human osteoarthritis synovial fibroblasts. Stimulation of OASFs with CCN4 induced VCAM-1 expression. CCN4-induced VCAM-1 expression was attenuated by αvβ5 or α6β1 integrin antibody, Syk inhibitor, PKCδ inhibitor (rottlerin), JNK inhibitor (SP600125), and AP-1 inhibitors (curcumin and tanshinone). Stimulation of cells with CCN4 increased Syk, PKCδ, and JNK activation. Treatment of OASFs with CCN4 also increased c-Jun phosphorylation, AP-1-luciferase activity, and c-Jun binding to the AP-1 element in the VCAM-1 promoter. Moreover, up-regulation of VCAM-1 increased the adhesion of monocytes to OASF monolayers, and this adhesion was attenuated by transfection with a VCAM-1 siRNA. Our results suggest that CCN4 increases VCAM-1 expression in human OASFs via the Syk, PKCδ, JNK, c-Jun, and AP-1 signaling pathways. The CCN4-induced VCAM-1 expression promoted monocyte adhesion to human OASFs.
Collapse
Affiliation(s)
- Ju-Fang Liu
- Central Laboratory, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
10
|
Yu HS, Lin TH, Tang CH. Bradykinin enhances cell migration in human prostate cancer cells through B2 receptor/PKCδ/c-Src dependent signaling pathway. Prostate 2013; 73:89-100. [PMID: 22653778 DOI: 10.1002/pros.22544] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 05/14/2012] [Indexed: 12/14/2022]
Abstract
BACKGROUND Prostate cancer is the most commonly diagnosed malignancy in men and shows a predilection for metastasis to the bone. Bradykinin (BK) is an inflammatory mediator, and shows elevated levels in regions of severe injury and inflammatory diseases. The aim of this study was to investigate whether Bradykinin is associated with migration of prostate cancer cells. METHODS Cancer cells migration activity was examined using the Transwell assay. The c-Src and PKCδ phosphorylation was examined by using Western blot method. The qPCR was used to examine the mRNA expression of metalloproteinase. A transient transfection protocol was used to examine NF-κB activity. RESULTS We found that bradykinin increased the chemomigration and the expression of MMP-9 of human prostate cancer cells. Bradykinin-mediated chemomigration and metalloproteinase expression was attenuated by PKCδ inhibitor (rottlerin), PKCδ siRNA, c-Src inhibitor (PP2) and c-Src mutant. Activations of PKCδ, c-Src and NF-κB pathways after bradykinin treatment was demonstrated, and bradykinin-induced expression of metalloproteinase and chemomigration activity was inhibited by the specific inhibitor and mutant of PKCδ, c-Src, and NF-κB cascades. CONCLUSIONS This study showed for the first time that the bradykinin mediates migration of human prostate cancer cells. One of the mechanisms underlying bradykinin directed migration was transcriptional up-regulation of MMP-9 and activation of B2 receptor, PKCδ, c-Src, and NF-κB pathways.
Collapse
Affiliation(s)
- Hsin-Shan Yu
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | | | | |
Collapse
|
11
|
Huang CY, Chen SY, Tsai HC, Hsu HC, Tang CH. Thrombin induces epidermal growth factor receptor transactivation and CCL2 expression in human osteoblasts. ACTA ACUST UNITED AC 2012; 64:3344-54. [DOI: 10.1002/art.34557] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
12
|
Zimmerman MC, Takapoo M, Jagadeesha DK, Stanic B, Banfi B, Bhalla RC, Miller FJ. Activation of NADPH oxidase 1 increases intracellular calcium and migration of smooth muscle cells. Hypertension 2011; 58:446-53. [PMID: 21810651 DOI: 10.1161/hypertensionaha.111.177006] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Redox-dependent migration and proliferation of vascular smooth muscle cells (SMCs) are central events in the development of vascular proliferative diseases; however, the underlying intracellular signaling mechanisms are not fully understood. We tested the hypothesis that activation of Nox1 NADPH oxidase modulates intracellular calcium ([Ca(2+)](i)) levels. Using cultured SMCs from wild-type and Nox1 null mice, we confirmed that thrombin-dependent generation of reactive oxygen species requires Nox1. Thrombin rapidly increased [Ca(2+)](i), as measured by fura-2 fluorescence ratio imaging, in wild-type but not Nox1 null SMCs. The increase in [Ca(2+)](i) in wild-type SMCs was inhibited by antisense to Nox1 and restored by expression of Nox1 in Nox1 null SMCs. Investigation into potential mechanisms by which Nox1 modulates [Ca(2+)](i) showed that thrombin-induced inositol triphosphate generation and thapsigargin-induced intracellular calcium mobilization were similar in wild-type and Nox1 null SMCs. To examine the effects of Nox1 on Ca(2+) entry, cells were either bathed in Ca(2+)-free medium or exposed to dihydropyridines to block L-type Ca(2+) channel activity. Treatment with nifedipine or removal of extracellular Ca(2+) reduced the thrombin-mediated increase of [Ca(2+)](i) in wild-type SMCs, whereas the response in Nox1 null SMCs was unchanged. Sodium vanadate, an inhibitor of protein tyrosine phosphatases, restored the thrombin-induced increase of [Ca(2+)](i) in Nox1 null SMCs. Migration of SMCs was impaired with deficiency of Nox1 and restored with expression of Nox1 or the addition of sodium vanadate. In summary, we conclude that Nox1 NADPH oxidase modulates Ca(2+) mobilization in SMCs, in part through regulation of Ca(2+) influx, to thereby promote cell migration.
Collapse
Affiliation(s)
- Matthew C Zimmerman
- Jr, University of Iowa, 285 Newton Rd, Room 2269 CBRB, Iowa City, IA 52242, USA
| | | | | | | | | | | | | |
Collapse
|
13
|
Tang CH, Hsu CJ, Fong YC. The CCL5/CCR5 axis promotes interleukin-6 production in human synovial fibroblasts. ACTA ACUST UNITED AC 2010; 62:3615-24. [DOI: 10.1002/art.27755] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
14
|
Yang SF, Chen MK, Hsieh YS, Chung TT, Hsieh YH, Lin CW, Su JL, Tsai MH, Tang CH. Prostaglandin E2/EP1 signaling pathway enhances intercellular adhesion molecule 1 (ICAM-1) expression and cell motility in oral cancer cells. J Biol Chem 2010; 285:29808-16. [PMID: 20647315 DOI: 10.1074/jbc.m110.108183] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Oral squamous cell carcinoma has a striking tendency to migrate and metastasize. Cyclooxygenase (COX)-2, the inducible isoform of prostaglandin (PG) synthase, has been implicated in tumor metastasis. However, the effects of COX-2 on human oral cancer cells are largely unknown. We found that overexpression of COX-2 or exogenous PGE(2) increased migration and intercellular adhesion molecule 1 (ICAM)-1 expression in human oral cancer cells. Using pharmacological inhibitors, activators, and genetic inhibition of EP receptors, we discovered that the EP1 receptor, but not other PGE receptors, is involved in PGE(2)-mediated cell migration and ICAM-1 expression. PGE(2)-mediated migration and ICAM-1 up-regulation were attenuated by inhibitors of protein kinase C (PKC)δ, and c-Src. Activation of the PKCδ, c-Src, and AP-1 signaling pathway occurred after PGE(2) treatment. PGE(2)-induced expression of ICAM-1 and migration activity were inhibited by a specific inhibitor, siRNA, and mutants of PKCδ, c-Src, and AP-1. In addition, migration-prone sublines demonstrated that cells with increased migration ability had higher expression of COX-2 and ICAM-1. Taken together, these results indicate that the PGE(2) and EP1 interaction enhanced migration of oral cancer cells through an increase in ICAM-1 production.
Collapse
Affiliation(s)
- Shun-Fa Yang
- From the Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Chen HT, Tsou HK, Tsai CH, Kuo CC, Chiang YK, Chang CH, Fong YC, Tang CH. Thrombin enhanced migration and MMPs expression of human chondrosarcoma cells involves PAR receptor signaling pathway. J Cell Physiol 2010; 223:737-45. [PMID: 20175118 DOI: 10.1002/jcp.22083] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Thrombin is a multifunctional protease that can activate hemostasis and coagulation through the cleavage of fibrinogen to form fibrin clots. Thrombin also plays a crucial role in migration and metastasis of human cancer cells. However, the effect of thrombin on migration activity in human chondrosarcoma cells is mostly unknown. Here, we found that thrombin increased the migration and expression of matrix metalloproteinase (MMP)-2 and MMP-13 in human chondrosarcoma cells (JJ012 and SW1353 cells). By using pharmacological inhibitors or activators or genetic inhibition by the protease-activated receptor (PAR), we found that the PAR1 and PAR4 receptor but not PAR3 receptor are involved in thrombin-mediated cell migration and MMPs expression. Thrombin-mediated migration and MMPs up-regulation was attenuated by phospholipase C (PLC), protein kinase C, and c-Src inhibitor. Activations of PLCbeta, PKCalpha, c-Src, and NF-kappaB pathways after thrombin treatment was demonstrated, and thrombin-induced MMPs expression and migration activity was inhibited by the specific inhibitors and mutants of PLC, PKC, c-Src, and NF-kappaB cascades. Taken together, our results indicated that thrombin enhances the migration of chondrosarcoma cells by increasing MMP-2 and MMP-13 expression through the PAR/PLC/PKCalpha/c-Src/NF-kappaB signal transduction pathway.
Collapse
Affiliation(s)
- Hsien-Te Chen
- Department of Orthopaedic, China Medical University Hospital, Taichung, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
16
|
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]
|
17
|
Cyclooxygenase-2 enhances alpha2beta1 integrin expression and cell migration via EP1 dependent signaling pathway in human chondrosarcoma cells. Mol Cancer 2010; 9:43. [PMID: 20178602 PMCID: PMC2837621 DOI: 10.1186/1476-4598-9-43] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Accepted: 02/23/2010] [Indexed: 12/13/2022] Open
Abstract
Background Cyclooxygenase (COX)-2, the inducible isoform of prostaglandin (PG) synthase, has been implicated in tumor metastasis. Interaction of COX-2 with its specific EP receptors on the surface of cancer cells has been reported to induce cancer invasion. However, the effects of COX-2 on migration activity in human chondrosarcoma cells are mostly unknown. In this study, we examined whether COX-2 and EP interaction are involved in metastasis of human chondrosarcoma. Results We found that over-expression of COX-2 or exogenous PGE2 increased the migration of human chondrosarcoma cells. We also found that human chondrosarcoma tissues and chondrosarcoma cell lines had significant expression of the COX-2 which was higher than that in normal cartilage. By using pharmacological inhibitors or activators or genetic inhibition by the EP receptors, we discovered that the EP1 receptor but not other PGE receptors is involved in PGE2-mediated cell migration and α2β1 integrin expression. Furthermore, we found that human chondrosarcoma tissues expressed a higher level of EP1 receptor than normal cartilage. PGE2-mediated migration and integrin up-regulation were attenuated by phospholipase C (PLC), protein kinase C (PKC) and c-Src inhibitor. Activation of the PLCβ, PKCα, c-Src and NF-κB signaling pathway after PGE2 treatment was demonstrated, and PGE2-induced expression of integrin and migration activity were inhibited by the specific inhibitor, siRNA and mutants of PLC, PKC, c-Src and NF-κB cascades. Conclusions Our results indicated that PGE2 enhances the migration of chondrosarcoma cells by increasing α2β1 integrin expression through the EP1/PLC/PKCα/c-Src/NF-κB signal transduction pathway.
Collapse
|
18
|
Tang CH, Hsu CJ, Yang WH, Fong YC. Lipoteichoic acid enhances IL-6 production in human synovial fibroblasts via TLR2 receptor, PKCdelta and c-Src dependent pathways. Biochem Pharmacol 2010; 79:1648-57. [PMID: 20109438 DOI: 10.1016/j.bcp.2010.01.025] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Revised: 01/20/2010] [Accepted: 01/20/2010] [Indexed: 02/02/2023]
Abstract
Patients with rheumatoid arthritis (RA) are at increased risk of developing infections and appear to be particularly susceptible to septic arthritis. Lipoteichoic acid (LTA), a cell wall component of Gram-positive bacteria is an amphiphilic, negatively charged glycolipid. However, the effects of LTA on human synovial fibroblasts are largely unknown. We investigated the signaling pathway involved in IL-6 production stimulated by LTA in rheumatoid arthritis synovial fibroblasts (RASF). LTA caused concentration- and time-dependent increases in IL-6 production. LTA-mediated IL-6 production was attenuated by Toll-like receptor 2 (TLR2) monoclonal antibody or siRNA. Pretreatment with PKCdelta inhibitor (rottlerin), c-Src inhibitor (PP2), AP-1 inhibitor (tanshinone IIA) and NF-kappaB inhibitor (PDTC and TPCK) also inhibited the potentiating action of LTA. However, focal adhesion kinase (FAK) mutant and siRNA did not affect LTA-mediated IL-6 production. Stimulation of cells with LTA increased the PKCdelta and c-Src phosphorylation and kinase activity. LTA increased the accumulation of p-c-Jun and p-p65 in the nucleus, as well as AP-1 and NF-kappaB luciferase activity. LTA-mediated increase of AP-1 and NF-kappaB luciferase activity was inhibited by rottlerin and PP2 or TLR2 and PKCdelta siRNA or c-Src mutant. Our results suggest that LTA-increased IL-6 production in human synovial fibroblasts via the TLR2 receptor, PKCdelta, c-Src, AP-1 and NF-kappaB signaling pathways.
Collapse
Affiliation(s)
- Chih-Hsin Tang
- Department of Pharmacology, China Medical University, Taichung, Taiwan.
| | | | | | | |
Collapse
|
19
|
Chuang JY, Yang WH, Chen HT, Huang CY, Tan TW, Lin YT, Hsu CJ, Fong YC, Tang CH. CCL5/CCR5 axis promotes the motility of human oral cancer cells. J Cell Physiol 2009; 220:418-26. [DOI: 10.1002/jcp.21783] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
20
|
Wang Z, Kong L, Kang J, Morgan JH, Shillcutt SD, Robinson JS, Nakayama DK. Thrombin stimulates mitogenesis in pig cerebrovascular smooth muscle cells involving activation of pro-matrix metalloproteinase-2. Neurosci Lett 2009; 451:199-203. [DOI: 10.1016/j.neulet.2009.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Revised: 12/23/2008] [Accepted: 01/02/2009] [Indexed: 11/28/2022]
|
21
|
Bradykinin-induced IL-6 expression through bradykinin B2 receptor, phospholipase C, protein kinase Cδ and NF-κB pathway in human synovial fibroblasts. Mol Immunol 2008; 45:3693-702. [DOI: 10.1016/j.molimm.2008.06.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Revised: 06/06/2008] [Accepted: 06/08/2008] [Indexed: 11/21/2022]
|
22
|
Chiu YC, Fong YC, Lai CH, Hung CH, Hsu HC, Lee TS, Yang RS, Fu WM, Tang CH. Thrombin-induced IL-6 production in human synovial fibroblasts is mediated by PAR1, phospholipase C, protein kinase C alpha, c-Src, NF-kappa B and p300 pathway. Mol Immunol 2007; 45:1587-99. [PMID: 18062909 DOI: 10.1016/j.molimm.2007.10.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2007] [Revised: 09/30/2007] [Accepted: 10/04/2007] [Indexed: 11/15/2022]
Abstract
Thrombin is a key factor in the stimulation of fibrin deposition, angiogenesis and proinflammatory processes. Abnormalities in these processes are primary features of rheumatoid arthritis (RA) in synovial tissues. We investigated the signaling pathway involved in IL-6 production caused by thrombin in synovial fibroblasts. Thrombin caused concentration- and time-dependent increases in IL-6 production. By using pharmacological inhibitors or activators or genetic inhibition by the protease activated receptor (PAR), siRNA revealed that the PAR1 receptor but not other PAR receptors is involved in thrombin-mediated up-regulation of IL-6. Thrombin-mediated IL-6 production was attenuated by thrombin inhibitor (PPACK), phospholipase C inhibitor (U73122), protein kinase C alpha inhibitor (Ro320432), Src inhibitor (PP2), NF-kappaB inhibitor (PDTC), I kappa B protease inhibitor (TPCK), or NF-kappaB inhibitor peptide. Stimulation of synovial fibroblasts with thrombin activated I kappa B kinase alpha/beta (IKK alpha/beta), I kappa B alpha phosphorylation, I kappa B alpha degradation, p65 phosphorylation at Ser(276), p65 and p50 translocation from the cytosol to the nucleus, and kappaB-luciferase activity. Thrombin-mediated an increase of IKK alpha/beta activity, kappaB-luciferase activity and p65 and p50 binding to the NF-kappaB element was inhibited by PPACK, U73122, Ro320432 and PP2. The binding of p65 and p50 to the NF-kappaB elements, as well as the recruitment of p300 and the enhancement of p50 acetylation on the IL-6 promoter was enhanced by thrombin. Our results suggest that thrombin increased IL-6 production in synovial fibroblasts via the PAR1 receptor/PI-PLC/PKC alpha/c-Src/NF-kappaB and p300 signaling pathway.
Collapse
Affiliation(s)
- Yung-Cheng Chiu
- Department of Orthopaedics, Taichung Veterans General Hospital, Taichung, Taiwan; Department of Pharmacology, College of Medicine, China Medical University, Taichung, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Lyle AN, Griendling KK. Modulation of vascular smooth muscle signaling by reactive oxygen species. Physiology (Bethesda) 2006; 21:269-80. [PMID: 16868316 DOI: 10.1152/physiol.00004.2006] [Citation(s) in RCA: 178] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Modulation of signaling in vascular cells by reactive oxygen species (ROS) affects many aspects of cellular function, including growth, migration, and contraction. NADPH oxidases, important sources of ROS, regulate many growth-specific and migration-related signaling pathways. Identifying the precise intracellular targets of ROS enhances understanding of their role in cardiovascular physiology and pathophysiology.
Collapse
Affiliation(s)
- Alicia N Lyle
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, Georgia, USA
| | | |
Collapse
|
24
|
Lin CH, Cheng HW, Hsu MJ, Chen MC, Lin CC, Chen BC. c-Src Mediates Thrombin-Induced NF-κB Activation and IL-8/CXCL8 Expression in Lung Epithelial Cells. THE JOURNAL OF IMMUNOLOGY 2006; 177:3427-38. [PMID: 16920985 DOI: 10.4049/jimmunol.177.5.3427] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In this study, we examined the regulation of NF-kappaB activation and IL-8/CXCL8 expression by thrombin in human lung epithelial cells (EC). Thrombin caused a concentration-dependent increase in IL-8/CXCL8 release in a human lung EC line (A549) and primary normal human bronchial EC. In A549 cells, thrombin, SFLLRN-NH2 (a protease-activated receptor 1 (PAR1) agonist peptide), and GYPGQV-NH2 (a PAR4 agonist peptide), but not TFRGAP-NH2 (a PAR3 agonist peptide), induced an increase in IL-8/CXCL8-luciferase (Luc) activity. The thrombin-induced IL-8/CXCL8 release was attenuated by D-phenylalanyl-L-prolyl-L-arginine chloromethyl ketone (a thrombin inhibitor), U73122 (a phosphoinositide-phospholipase C inhibitor), Ro-32-0432 (a protein kinsase C alpha (PKC alpha) inhibitor), an NF-kappaB inhibitor peptide, and Bay 117082 (an IkappaB phosphorylation inhibitor). Thrombin-induced increase in IL-8/CXCL8-Luc activity was inhibited by the dominant-negative mutant of c-Src and the cells transfected with the kappaB site mutation of the IL-8/CXCL8 construct. Thrombin caused time-dependent increases in phosphorylation of c-Src at tyrosine 416 and c-Src activity. Thrombin-elicited c-Src activity was inhibited by Ro-32-0432. Stimulation of cells with thrombin activated IkappaB kinase alphabeta (IKK alphabeta), IkappaB alpha phosphorylation, IkappaB alpha degradation, p50 and p65 translocation from the cytosol to the nucleus, NF-kappaB-specific DNA-protein complex formation, and kappaB-Luc activity. Pretreatment of A549 cells with Ro-32-4032 and the dominant-negative mutant of c-Src DN inhibited thrombin-induced IKK alphabeta activity, kappaB-Luc activity, and NF-kappaB-specific DNA-protein complex formation. Further studies revealed that thrombin induced PKC alpha, c-Src, and IKK alphabeta complex formation. These results show for the first time that thrombin, acting through PAR1 and PAR4, activates the phosphoinositide-phospholipase C/PKC alpha/c-Src/IKK alphabeta signaling pathway to induce NF-kappaB activation, which in turn induces IL-8/CXCL8 expression and release in human lung EC.
Collapse
Affiliation(s)
- Chien-Huang Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taiwan
| | | | | | | | | | | |
Collapse
|
25
|
Madamanchi NR, Hu ZY, Li F, Horaist C, Moon SK, Patterson C, Runge MS, Ruef J, Fritz PH, Aaron J. A noncoding RNA regulates human protease-activated receptor-1 gene during embryogenesis. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1576:237-45. [PMID: 12084570 DOI: 10.1016/s0167-4781(02)00308-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Activation of the human protease-activated receptor-1 (PAR-1) by thrombin leads to myriad functions essential for maintaining vascular integrity. Upregulation of PAR-1 expression is considered important in atherosclerosis, angiogenesis and tumor metastasis. In vitro analysis of the human PAR-1 promoter function revealed a positive regulatory element between -4.2 and -3.2 kb of the transcription start site. This element was examined in transgenic mice containing either 4.1 or 2.9 kb of the 5' flanking sequence driving a LacZ reporter gene. Only the 4.1 kb PAR-1 transgene was expressed in vivo and only during embryonic development. The transgene expression was observed only in developing arteries and not in veins. Further examination of this putative regulatory sequence identified a novel noncoding RNA (ncR-uPAR:noncoding RNA upstream of the PAR-1) gene at -3.4 kb. The ncR-uPAR upregulated PAR-1-core promoter-driven luciferase activity and mRNA expression in vitro in a Pol II-dependent manner. This noncoding RNA appears to act in trans, albeit locally at the adjacent PAR-1 promoter. These data suggest that an untranslated RNA plays a role in PAR-1 gene expression during embryonic growth.
Collapse
MESH Headings
- Animals
- Arteries/cytology
- Arteries/embryology
- Base Sequence
- Embryo, Mammalian/anatomy & histology
- Embryo, Mammalian/physiology
- Female
- Gene Expression Regulation, Developmental
- Genes, Reporter
- Humans
- Male
- Mice
- Mice, Transgenic
- Molecular Sequence Data
- Promoter Regions, Genetic
- RNA, Untranslated/genetics
- RNA, Untranslated/metabolism
- Receptor, PAR-1
- Receptors, Thrombin/genetics
- Receptors, Thrombin/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
Collapse
Affiliation(s)
- Nageswara R Madamanchi
- Carolina Cardiovascular Biology Center, Department of Medicine, University of North Carolina, Chapel Hill, NC 27599-7126, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Boulom V, Lee HW, Zhao L, Eghbali-Webb M. Stimulation of DNA synthesis, activation of mitogen-activated protein kinase ERK2 and nuclear accumulation of c-fos in human aortic smooth muscle cells by ketamine. Cell Prolif 2002; 35:155-65. [PMID: 12027951 PMCID: PMC6495842 DOI: 10.1046/j.1365-2184.2002.00233.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Proliferation of vascular smooth muscle cells is known to be regulated by autocrine and paracrine stimuli, including extracellular matrix, reactive oxygen species, lipids, and biomechanical forces. The effect of many pharmacological agents to which smooth muscle cells may be exposed, however, is widely unexplored. Ketamine, an intravenous anaesthetic and a phencyclidine derivative, regulates diverse intracellular signalling pathways in smooth muscle cells, several of which are known to affect cell proliferation. The effect of ketamine on proliferative response of smooth muscle cells, however, is not determined. We tested the hypothesis that ketamine may regulate proliferation of smooth muscle cells, and investigated the effects of pharmacological doses of ketamine on their proliferative capacity by measuring DNA synthesis and activation of mitogen-activated protein (MAP) kinase signalling pathway in human aortic smooth muscle cells. DNA synthesis, as determined by incorporation of 3H-thymidine into DNA, was enhanced by 73% (P < 0.0001) and 130% (P < 0.0001) with 10 and 100 microm ketamine, respectively. Ketamine-induced DNA synthesis was dependent on de novo protein synthesis, as it was abolished by an inhibitor of protein synthesis, cycloheximide. A synthetic inhibitor of MAP kinase pathway, PD98059, decreased 50% (P < 0.0001) of ketamine-induced DNA synthesis, suggesting that the activation of MAP kinase pathway was partially responsible for ketamine-induced effects. Consistently, in-gel kinase assay and in vitro kinase assay of cell lysates showed ketamine-induced MAP kinase activation and expression of ERK2 (extracellular signal-regulated kinase) in smooth muscle cells. This effect of ketamine was not dependent on de novo protein synthesis. Immunofluorescent light microscopy showed ketamine-induced nuclear accumulation of c-fos, a downstream effect of MAP kinase activation, in smooth muscle cells. In conclusion, these data support the hypothesis of the study and demonstrate that ketamine, by stimulating DNA synthesis in human aortic smooth muscle cells, may have an impact on proliferative capacity of these cells. The present results also demonstrate that ketamine induces the activation of MAP kinase pathway and nuclear accumulation of transcription factor c-fos in smooth muscle cells. They further demonstrate that the activation of MAP kinases is partially responsible for ketamine-induced DNA synthesis in human aortic smooth muscle cells. Together, these findings suggest that ketamine may play a role as a pharmacological regulator of mechanisms involved in proliferation of smooth muscle cells.
Collapse
Affiliation(s)
- V Boulom
- Department of Anaesthesiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | |
Collapse
|
27
|
Stepien O, Zhang Y, Zhu D, Marche P. Dual mechanism of action of amlodipine in human vascular smooth muscle cells. J Hypertens 2002; 20:95-102. [PMID: 11791031 DOI: 10.1097/00004872-200201000-00014] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES It has been recently shown that calcium channel blockers (CCBs) could also control smooth muscle cell (SMC) growth/reactivity through mechanisms that were unrelated to their CCB property. Here, we investigated the effects of amlodipine and isradipine on Ca2+ movements and p42/p44 mitogen-activated protein kinase (ERK 1/2) activities, which are two early signalling events triggered by growth factors such as thrombin and basic fibroblast growth factor (bFGF). METHODS In cultured human SMCs isolated from internal mammary arteries, Ca2+ movements and ERK 1/2 activation were studied by measurement of the intracellular Ca2+ concentration in Fura 2-labelled SMCs and by Western blots, respectively. RESULTS In thrombin- and thapsigargin-stimulated SMCs, amlodipine and not isradipine dose-dependently reduced Ca2+ mobilization (i.e. Ca2+ release from internal stores); these dihydropyridines did not affect either Ca2+ influx or ERK 1/2 activation. In bFGF-stimulated SMCs, amlodipine and isradipine reduced both Ca2+ influx and ERK 1/2 activation without affecting Ca2+ mobilization. ERK 1/2 activation could also be directly stimulated by the l-type channel agonist Bay K 8644, demonstrating the involvement of voltage-gated Ca2+ influx in this process. Most of the observed effects described were obtained with approximately 10 nmol/l amlodipine/isradipine (i.e. concentrations close to the peak plasma level in treated patients). CONCLUSIONS In human SMCs, amlodipine can (i) specifically alter Ca2+ mobilization, likely by interacting with the sarcoplasmic reticulum and (ii) inhibit voltage-dependent Ca2+ influx and the resulting ERK 1/2 activation. It is likely that amlodipine exerts its growth-inhibitory potency by interfering with multiple branches of mitogenic signalling pathways.
Collapse
Affiliation(s)
- Olivier Stepien
- Department of Pharmacology, CNRS UMR 8604 and University René Descartes, Paris, France
| | | | | | | |
Collapse
|
28
|
Boncela J, Papiewska I, Fijalkowska I, Walkowiak B, Cierniewski CS. Acute phase protein alpha 1-acid glycoprotein interacts with plasminogen activator inhibitor type 1 and stabilizes its inhibitory activity. J Biol Chem 2001; 276:35305-11. [PMID: 11418606 DOI: 10.1074/jbc.m104028200] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
alpha(1)-Acid glycoprotein, one of the major acute phase proteins, was found to interact with plasminogen activator inhibitor type 1 (PAI-1) and to stabilize its inhibitory activity toward plasminogen activators. This conclusion is based on the following observations: (a) alpha(1)-acid glycoprotein was identified to bind PAI-1 by a yeast two-hybrid system. Three of 10 positive clones identified by this method to interact with PAI-1 contained almost the entire sequence of alpha(1)-acid glycoprotein; (b) this protein formed complexes with PAI-1 that could be immunoprecipitated from both the incubation mixtures and blood plasma by specific antibodies to either PAI-1 or alpha(1)-acid glycoprotein. Such complexes could be also detected by a solid phase binding assay; and (c) the real-time bimolecular interactions monitored by surface plasmon resonance indicated that the complex of alpha(1)-acid glycoprotein with PAI-1 is less stable than that formed by vitronectin with PAI-1, but in both cases, the apparent K(D) values were in the range of strong interactions (4.51 + 1.33 and 0.58 + 0.07 nm, respectively). The on rate for binding of PAI-1 to alpha(1)-glycoprotein or vitronectin differed by 2-fold, indicating much faster complex formation by vitronectin than by alpha(1)-acid glycoprotein. On the other hand, dissociation of PAI-1 bound to vitronectin was much slower than that from the alpha(1)-acid glycoprotein, as indicated by 4-fold lower k(off) values. Furthermore, the PAI-1 activity toward urokinase-type plasminogen activator and tissue-type plasminogen activator was significantly prolonged in the presence of alpha(1)-acid glycoprotein. These observations suggest that the complex of PAI-1 with alpha(1)-acid glycoprotein can play a role as an alternative reservoir of the physiologically active form of the inhibitor, particularly during inflammation or other acute phase reactions.
Collapse
Affiliation(s)
- J Boncela
- Center for Microbiology and Virology, Polish Academy of Sciences 93-232 Lodz, Poland
| | | | | | | | | |
Collapse
|
29
|
Abstract
Vascular smooth muscle cells (VSMC) exhibit several growth responses to agonists that regulate their function including proliferation (hyperplasia with an increase in cell number), hypertrophy (an increase in cell size without change in DNA content), endoreduplication (an increase in DNA content and usually size), and apoptosis. Both autocrine growth mechanisms (in which the individual cell synthesizes and/or secretes a substance that stimulates that same cell type to undergo a growth response) and paracrine growth mechanisms (in which the individual cells responding to the growth factor synthesize and/or secrete a substance that stimulates neighboring cells of another cell type) are important in VSMC growth. In this review I discuss the autocrine and paracrine growth factors important for VSMC growth in culture and in vessels. Four mechanisms by which individual agonists signal are described: direct effects of agonists on their receptors, transactivation of tyrosine kinase-coupled receptors, generation of reactive oxygen species, and induction/secretion of other growth and survival factors. Additional growth effects mediated by changes in cell matrix are discussed. The temporal and spatial coordination of these events are shown to modulate the environment in which other growth factors initiate cell cycle events. Finally, the heterogeneous nature of VSMC developmental origin provides another level of complexity in VSMC growth mechanisms.
Collapse
Affiliation(s)
- B C Berk
- Center for Cardiovascular Research, University of Rochester, School of Medicine and Dentistry, Rochester, New York 14642, USA.
| |
Collapse
|
30
|
Sugo S, Minamino N, Shoji H, Isumi Y, Nakao K, Kangawa K, Matsuo H. Regulation of endothelin-1 production in cultured rat vascular smooth muscle cells. J Cardiovasc Pharmacol 2001; 37:25-40. [PMID: 11152371 DOI: 10.1097/00005344-200101000-00004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Endothelin-1 (ET-1) is secreted from all rat vascular smooth muscle cells (VSMCs) examined, in addition to endothelial cells (ECs). An average secretion rate of ET-1 from rat VSMCs was determined to be 10% that excreted from ECs. We examined the effects of 22 substances on ET-1 secretion from VSMCs and compared them with those from ECs. Transforming growth factor-beta1 (TGF-beta), acidic and basic fibroblast growth factors, epidermal growth factor, angiotensin II, and adrenaline stimulated ET-1 secretion from VSMCs, whereas forskolin, thrombin, and platelet-derived growth factor-BB reduced it. Only TGF-beta and phorbol ester elicited consistent effects on ET-1 secretion from VSMCs and ECs. Regulation of ET-1 and adrenomedullin secretion from VSMCs was distinctly different. These data suggest that ET- 1 production in VSMCs is regulated by a mechanism separate from that in ECs and from adrenomedullin production in VSMCs. Chromatographic analysis showed immunoreactive ET-1 secreted from VSMCs was mainly composed of big ET- 1, whereas approximately 90% of that from ECs was ET-1. By TGF-beta stimulation of VSMCs, the ratio of big ET-1 to ET-1 was further increased. Because big ET-1 is converted into ET-1 only on the surface of the ECs in the culture system, big ET-1 secreted from the VSMCs may function as a mediator transmitting a signal from VSMCs to ECs in vivo.
Collapse
MESH Headings
- Adrenomedullin
- Animals
- Cattle
- Cells, Cultured
- Dose-Response Relationship, Drug
- Endothelin-1/drug effects
- Endothelin-1/metabolism
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Gene Expression/drug effects
- Gene Expression/physiology
- Humans
- Mice
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Peptides/drug effects
- Peptides/metabolism
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Time Factors
- Transforming Growth Factor beta/pharmacology
Collapse
Affiliation(s)
- S Sugo
- National Cardiovascular Center Research Institute, Osaka, Japan
| | | | | | | | | | | | | |
Collapse
|
31
|
Stepien O, Marche P. Amlodipine inhibits thapsigargin-sensitive CA(2+) stores in thrombin-stimulated vascular smooth muscle cells. Am J Physiol Heart Circ Physiol 2000; 279:H1220-7. [PMID: 10993788 DOI: 10.1152/ajpheart.2000.279.3.h1220] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ca(2+) channel blockers, such as amlodipine, inhibit vascular smooth muscle cell (VSMC) growth through interactions with targets other than L-type Ca(2+) channels. The effects of amlodipine on Ca(2+) movements in thrombin- and thapsigargin-stimulated VSMCs were therefore investigated by determining the variations of intracellular free Ca(2+) concentration in fura 2-loaded cultured VSMCs. Results indicated that 10-1,000 nM amlodipine inhibited 1) thrombin-induced Ca(2+) mobilization from a thapsigargin-sensitive pool and 2) thapsigargin-induced Ca(2+) responses, including Ca(2+) mobilization from internal stores and store-operated Ca(2+) entry. These effects of amlodipine do not involve L-type Ca(2+) channels and could not be reproduced with 100 nM isradipine, diltiazem, or verapamil. The inhibition by amlodipine of Ca(2+) mobilization appears therefore to be a specific property of the drug, in addition to its Ca(2+) channel-blocking property. It is suggested that amlodipine acts in this capacity by interacting with Ca(2+)-ATPases of the sarcoplasmic reticulum, thus modulating the enzyme activity. This mechanism might participate in the inhibitory effect of amlodipine on VSMC growth.
Collapse
MESH Headings
- Amlodipine/pharmacology
- Animals
- Calcium/metabolism
- Calcium Channel Blockers/pharmacology
- Calcium Channels, L-Type/drug effects
- Calcium Channels, L-Type/metabolism
- Calcium-Transporting ATPases/antagonists & inhibitors
- Cells, Cultured
- Diltiazem/pharmacology
- Dose-Response Relationship, Drug
- Enzyme Inhibitors/pharmacology
- Intracellular Fluid/metabolism
- Isradipine/pharmacology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Potassium Chloride/pharmacology
- Rats
- Rats, Inbred WKY
- Sarcoplasmic Reticulum/metabolism
- Signal Transduction/drug effects
- Thapsigargin/pharmacology
- Thrombin/metabolism
- Thrombin/pharmacology
- Verapamil/pharmacology
Collapse
Affiliation(s)
- O Stepien
- Département de Pharmacologie, Université René Descartes, 75015 Paris, France
| | | |
Collapse
|
32
|
Ludwicka-Bradley A, Tourkina E, Suzuki S, Tyson E, Bonner M, Fenton JW, Hoffman S, Silver RM. Thrombin upregulates interleukin-8 in lung fibroblasts via cleavage of proteolytically activated receptor-I and protein kinase C-gamma activation. Am J Respir Cell Mol Biol 2000; 22:235-43. [PMID: 10657945 DOI: 10.1165/ajrcmb.22.2.3642] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Acute and chronic interstitial lung diseases are accompanied by evidence of inflammation and vascular injury. Thrombin activity in bronchoalveolar lavage fluid from such conditions is often increased, as well as interleukin (IL)-8. We observed that conditioned medium from lung fibroblasts exposed to thrombin has chemotactic activity for polymorphonuclear cells, and that this activity can be abolished by antibody to IL-8. We report that thrombin stimulates expression of IL-8 in human lung fibroblasts on both the messenger RNA and protein levels in a time- and dose-dependent manner. Stimulation of IL-8 expression by thrombin is inhibited by specific thrombin inhibitors. Synthetic thrombin receptor agonist peptide-14 mimics thrombin's stimulation of IL-8 expression in a dose-dependent manner consistent with the idea that upregulation of IL-8 by thrombin in human lung fibroblasts requires cleavage of proteolytically activated receptor-I. We demonstrate further that thrombin-induced IL-8 synthesis is regulated by protein kinase (PK) C. PKC-gamma may be involved in the upregulation of lung fibroblast IL-8 by thrombin because stimulation of lung fibroblasts with thrombin caused significant upregulation of PKC-gamma and because PKC-gamma antisense oligonucleotides inhibited the accumulation of PKC-gamma protein and IL-8 protein. Our data suggest that the PKC-gamma isoform increase observed after thrombin stimulation is required for thrombin-induced IL-8 formation by human lung fibroblasts.
Collapse
Affiliation(s)
- A Ludwicka-Bradley
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Mizuno O, Hojo Y, Ikeda U, Katsuki T, Fukazawa H, Kurosaki K, Fujikawa H, Shimada K. Assessment of coagulation and platelet activation in coronary sinus blood induced by transcatheter coronary intervention for narrowing of the left anterior descending coronary artery. Am J Cardiol 2000; 85:154-60. [PMID: 10955369 DOI: 10.1016/s0002-9149(99)00651-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Influences of recently developed methods for coronary intervention on hemostasis in the coronary circulation are unclear. The objective of this study was to investigate changes in coagulation and platelet activation in the coronary circulation induced by percutaneous transluminal coronary angioplasty (PTCA). We studied 35 patients with coronary heart disease who underwent elective PTCA to isolated stenotic narrowing of left coronary arteries. Seven patients received only PTCA, 12 underwent percutaneous transluminal rotational atherectomy (PTRA), and 16 underwent stent implantation. Blood samples were drawn from the coronary sinus immediately before and after as well as 4 and 24 hours after PTCA. Plasma levels of tissue factor (TF), thrombin-antithrombin III complex, plasminogen activator inhibitor (PAI)-1, tissue plasminogen activator (t-PA), beta-thromboglobulin, and platelet factor 4 were measured by enzyme-linked immunosorbent assay. In all patients, TF levels in the coronary sinus blood showed significant increases 4 and 24 hours after PTCA and thrombin-antithrombin III complex levels showed significant increases 24 hours after PTCA. PAI-1 showed significant increases 24 hours after PTCA and t-PA showed significant increases 4 and 24 hours after PTCA. Changes in levels of these markers by PTCA were similar among the 3 groups. In PTRA, levels of beta-thromboglobulin and platelet factor 4, markers of platelet activation, increased immediately after the procedure and returned to baseline levels after 4 hours. PTCA induced increases in blood coagulation and fibrinolysis in the coronary circulation. PTRA caused a marked but transient activation of platelets. These changes may contribute to acute complications during the procedure.
Collapse
Affiliation(s)
- O Mizuno
- Department of Cardiology, Jichi Medical School, Tochigi, Japan
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Sherer TB, Neff PS, Parks JK, Tuttle JB. Mitochondrial impact on nerve growth factor production in vascular smooth muscle-derived cells. BIOCHIMICA ET BIOPHYSICA ACTA 1999; 1473:305-20. [PMID: 10594368 DOI: 10.1016/s0304-4165(99)00194-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ht30</=Ht10>/=Ht5). Cells with reduced mitochondrial activity also showed abnormal responses to the stimulation of NGF output. Thrombin and phorbol ester elevated NGF production from Ht100, Ht30 and Ht10 cells, but not from Ht5 cells. Ht30 cells, despite secreting less NGF basally than Ht100 cells, reached a similar or greater NGF output upon stimulation. Mitogens increased NGF output and NGF mRNA levels with the largest effect on NGF protein in Ht30 cells. Free radical production and the ability of cells to respond to NGF-inducing agents were related. These data suggest that chronic impairment of mitochondrial function associates with disturbances in cellular production of a signaling protein.
Collapse
Affiliation(s)
- T B Sherer
- Center for the Study of Neurodegenerative Disease, Health Science Center, University of Virginia, MR4 Box 5148, Charlottesville, VA 22908, USA
| | | | | | | |
Collapse
|
35
|
Sato T, Yoshinouchi T, Sugimoto T, Sakamoto T, Fujieda H, Murao S, Sato H, Ohe T. Prognostic value of serum hepatocyte growth factor in patients with acute coronary syndromes. JAPANESE CIRCULATION JOURNAL 1999; 63:583-8. [PMID: 10478806 DOI: 10.1253/jcj.63.583] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The present study examined whether or not hepatocyte growth factor (HGF), an endothelium-specific growth factor that stimulates regeneration of the endothelium, is increased or has a prognostic significance in patients with acute coronary syndromes. HGF was measured in 106 patients with coronary artery disease (20 stable effort angina, 12 unstable angina without adverse events, 24 unstable angina with adverse events and 50 acute myocardial infarction) on admission and 21 normal volunteers. The measurements in all patients were recorded before administration of heparin, and in acute myocardial infarction patients they were recorded from days 2 to 6 after heparin discontinuation on day 1. HGF levels (ng/ml) were 0.30+/-0.06 for the controls, 0.31+/-0.08 for stable effort angina patients, 0.31+/-0.08 for unstable angina patients without adverse events, 0.40+/-0.20 for unstable angina patients with adverse events and in acute myocardial infarction patients they were 0.45+/-0.18 on day 0, 0.57+/-0.45 on day 2, 0.50+/-0.35 on day 3, 0.48+/-0.32 on day 4, 0.44+/-0.20 on day 5, and 0.38+/-0.14 on day 6. HGF plays a crucial role in the restoration of injured endothelial cells and is a predictor of adverse events in patients with acute coronary syndromes.
Collapse
Affiliation(s)
- T Sato
- Department of Cardiovascular Medicine, Matsuyama Shimin Hospital, Matsuyama City, Ehime Pref., Japan.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Sower LE, Payne DA, Meyers R, Carney DH. Thrombin peptide, TP508, induces differential gene expression in fibroblasts through a nonproteolytic activation pathway. Exp Cell Res 1999; 247:422-31. [PMID: 10066370 DOI: 10.1006/excr.1998.4372] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Prior studies have shown that synthetic peptides representing the domain of thrombin responsible for high-affinity binding to fibroblasts stimulate chemotactic and cell proliferative signals through a nonproteolytic mechanism. One of these peptides, TP508, has recently been shown to be chemotactic for neutrophils, to enhance collagen accumulation in wounds, to enhance revascularization of wounds, and to accelerate the healing of incisional and open wounds in normal animals and in animals with impaired healing. To determine whether TP508 activates the proteolytically activated receptor for thrombin (PAR1), or the signals that are activated by PAR1, we treated human fibroblasts with TP508 and the PAR1-activating peptide, SFLLRNP, and analyzed the effects of these peptides on gene expression using differential display reverse transcriptase polymerase chain reaction. TP508 induces expression of a number of specific message fragments with short tyrosine kinase-like domains that are not induced by SFLLRNP. Sequencing full-length clones prepared by Marathon extension of TP508-induced fragments revealed that among the induced transcripts, there was a sequence with 88% homology to human annexin V. Northern analysis with authentic annexin V cDNA confirms that TP508, but not SFLLRNP, induces expression of annexin V in human fibroblasts. These results demonstrate that TP508 activates a cellular response separate from that activated through PAR1 and supports the hypothesis that TP508 acts through a separate nonproteolytically activated thrombin receptor that may be responsible for high-affinity thrombin binding and for nonproteolytic signals that are required for thrombin stimulation of cell proliferation.
Collapse
Affiliation(s)
- L E Sower
- The Department of Human Biological Chemistry and Genetics, The University of Texas Medical Branch, Galveston, Texas, 77555-0645, USA
| | | | | | | |
Collapse
|
37
|
Ikeda T, Iwamoto T, Wakabayashi S, Shigekawa M. Regulation of the Na+/H+ exchanger in fibroblasts overexpressing the Na+/Ca2+ exchanger. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 274:C1537-44. [PMID: 9696696 DOI: 10.1152/ajpcell.1998.274.6.c1537] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
To assess the role of Ca2+ in regulation of the Na+/H+ exchanger (NHE1),we used CCL-39 fibroblasts overexpressing the Na+/Ca2+ exchanger (NCX1). Expression of NCX1 markedly inhibited the transient cytoplasmic Ca2+ rise and long-lasting cytoplasmic alkalinization (60-80% inhibition) induced by alpha-thrombin. In contrast, coexpression of NCX1 did not inhibit this alkalinization in cells expressing the NHE1 mutant with the calmodulin (CaM)-binding domain deleted (amino acids 637-656), suggesting that the effect of NCX1 transfection involves Ca2+-CaM binding. Expression of NCX1 only slightly inhibited platelet-derived growth factor BB-induced alkalinization and did not affect hyperosmolarity- or phorbol 12-myristate 13-acetate-induced alkalinization. Downregulation of protein kinase C (PKC) inhibited thrombin-induced alkalinization partially in control cells and abolished it completely in NCX1-transfected cells, suggesting that the thrombin effect is mediated exclusively via Ca2+ and PKC. On the other hand, deletion mutant study revealed that PKC-dependent regulation occurs through a small cytoplasmic segment (amino aids 566-595). These data suggest that a mechanism involving direct Ca2+-CaM binding lasts for a relatively long period after agonist stimulation, despite apparent short-lived Ca2+ mobilization, and further support our previous conclusion that Ca2+- and PKC-dependent mechanisms are mediated through distinct segments of the NHE1 cytoplasmic domain.
Collapse
Affiliation(s)
- T Ikeda
- Department of Molecular Physiology, National Cardiovascular Research Institute, Osaka, Japan
| | | | | | | |
Collapse
|
38
|
Abstract
Strokes are heterogeneous not only with respect to their presentation, but more importantly in terms of the underlying pathology. There are now a number of choices available for the treatment of ischemic stroke, and the causative mechanisms responsible for each individual stroke must be considered in choosing an appropriate form of treatment. This article explores the underlying pathophysiological mechanisms responsible for the major categories of stroke and also examines the reasons why strokes worsen or evolve.
Collapse
Affiliation(s)
- R C Hinton
- Department of Neurology, University of Texas Southwestern Medical School, Dallas, USA
| |
Collapse
|
39
|
Varela O, Martínez-González J, Badimon L. The response of smooth muscle cells to alpha-thrombin depends on its arterial origin: comparison among different species. Eur J Clin Invest 1998; 28:313-23. [PMID: 9615911 DOI: 10.1046/j.1365-2362.1998.00280.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Thrombin plays a pivotal role in the pathogenesis of arterial thrombosis and exerts a wide spectrum of effects on the cells of vessel walls. METHODS In this paper we focus on the direct role of thrombin as a mitogen for smooth muscle cells (SMCs) derived from different vessels from the vascular tree (coronary artery, aorta, carotid artery and pulmonary artery) of different species (human and pig). All cell populations examined responded mitogenically to alpha-thrombin, however the extent of this response was dependent on both vascular origin and the species from which vessels were derived. RESULTS Thrombin (1-100 nmol L-1) induced DNA synthesis ranging from 1.5- to 4-fold baseline depending on cell type. Porcine coronary SMCs showed the highest response to thrombin (100 nmol L-1) in terms of protein and DNA synthesis (4.0 +/- 0.2-fold) and cell division (53.4 +/- 8.8%) among the tested cells. In these cells recombinant (r)-hirudin, a specific thrombin inhibitor, exhibited maximal effectiveness to block the mitogenic effect of thrombin. Human SMC response ([3H]-thymidine incorporation) to either sera or thrombin was lower than that of porcine cells. In contrast, c-fos mRNA levels induced by thrombin in human SMCs were higher than those induced in porcine cells. In human cells, thrombin elicited an overexpression of c-fos and a lower rate of [3H]-thymidine incorporation than in porcine cells. Insulin-like growth factor I but not insulin showed additive mitogenic effects with thrombin in human coronary SMCs. The response of these cells to thrombin from different sources was a function of thrombin specific activity. CONCLUSION These results suggest that the cell system chosen to check thrombin mitogenicity not only determines thrombin quantitative effects but also may affect the effectiveness of an inhibitor to block its biological activity.
Collapse
Affiliation(s)
- O Varela
- Cardiovascular Research Center, CSIC-HSCSP-UAB, Barcelona, Spain
| | | | | |
Collapse
|
40
|
Jerius H, Beall A, Woodrum D, Epstein A, Brophy C. Thrombin-induced vasospasm: Cellular signaling mechanisms. Surgery 1998. [DOI: 10.1016/s0039-6060(98)70227-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
41
|
Cizmeci-Smith G, Carey DJ. Thrombin stimulates syndecan-1 promotor activity and expression of a form of syndecan-1 that binds antithrombin III in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 1997; 17:2609-16. [PMID: 9409233 DOI: 10.1161/01.atv.17.11.2609] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Vascular smooth muscle (VSM) cells express transmembrane proteoglycans of the syndecan gene family. We reported previously that the expression of syndecans by VSM cells is regulated by mitogens such as serum, platelet-derived growth factor, and basic fibroblast growth factor and that syndecan expression is induced after balloon injury in vivo. We now show that thrombin is a potent inducer of syndecan-1 expression in VSM cells. Transient transfection experiments with a rat syndecan-1 promoter construct demonstrated that thrombin stimulates transcription of the syndecan-1 gene. Syndecan expression in response to thrombin was not inhibited by downregulation of protein kinase C. Thrombin-induced syndecan-1 expression was dependent on tyrosine kinase activity. Calcium was necessary for syndecan-1 expression, but increasing the intracellular calcium levels was not sufficient to induce syndecan-1 expression. Analysis of antithrombin III (AT III) binding activity revealed that thrombin caused an increase in the synthesis of syndecan-1 molecules that exhibited high-affinity AT III binding. These results suggest that VSM cells could play an important role in controlling local thrombus formation subsequent to vascular injury, via a feedback mechanism that involves thrombin-induced stimulation of an inhibitor of thrombin activity.
Collapse
|
42
|
Seewald S, Nickenig G, Ko Y, Vetter H, Sachinidis A. Low density lipoprotein enhances the thrombin-induced growth of vascular smooth muscle cells. Cardiovasc Res 1997; 36:92-100. [PMID: 9415277 DOI: 10.1016/s0008-6363(97)00139-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE In the present study we investigated whether low density lipoprotein is able to enhance the growth promoting effects of thrombin in vascular smooth muscle cells. METHODS DNA synthesis was examined by measurement of the [3H]thymidine incorporation into the cell DNA. Cell count was measured with a Neubauer cell box. Thrombin receptor mRNA was determined by Northern blotting. Ca2+ was measured by the fura 2-method. RESULTS Thrombin (5 nmol/l), thrombin receptor activating protein (3 mumol/l) and low density lipoprotein (33 nmol/l) induce a 652 +/- 80%, 593 +/- 80% and a 316 +/- 60% increase in [3H]thymidine incorporation into DNA (mean +/- SD, n = 3), respectively. A coincubation of thrombin or thrombin receptor activating protein with low density lipoprotein led to a 1245 +/- 160% or 1200 +/- 40% increase of DNA synthesis (mean +/- SD, n = 3). Thus, coincubation of low density lipoprotein and thrombin causes a synergistic rather than an additive mitogenic effect on smooth muscle cells. Thrombin and low density lipoprotein induced a 22 +/- 8.4% and a 29% +/- 6% increase in cell number, respectively. Simultaneous treatment of vascular smooth muscle cells with thrombin and low density lipoprotein caused a 63 +/- 14% increase in cell number (mean +/- SD, n = 3). To further elucidate the underlying mechanism, we studied the effect of low density lipoprotein on the expression of thrombin receptor mRNA. Low density lipoprotein caused a 2.5-fold increase of thrombin receptor mRNA within 24 h, as assessed by Northern analysis. Preincubation of cells for 24 h with 33 nmol/l low density lipoprotein resulted in an elevation of the thrombin-induced increase in cytosolic free Ca2+ concentration from 538 +/- 54 to 923 +/- 75 nmol/l (mean +/- SD, n = 4). CONCLUSION In summary, low density lipoprotein may enhance the mitogenic effect of thrombin probably by an up-regulation of thrombin receptor gene expression in vascular smooth muscle cells or by an elevation of the thrombin-induced increase in cytosolic free Ca2+ concentration.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic
- Blotting, Northern
- Calcium/analysis
- Calcium/metabolism
- Cell Division/drug effects
- Cells, Cultured
- DNA/biosynthesis
- Intracellular Fluid/metabolism
- Lipoproteins, LDL/pharmacology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- RNA/analysis
- Rats
- Rats, Inbred WKY
- Statistics, Nonparametric
- Thrombin/pharmacology
Collapse
Affiliation(s)
- S Seewald
- Medizinische Universitäts-Poliklinik, Bonn, Germany
| | | | | | | | | |
Collapse
|
43
|
The Suicide Substrate Reaction Between Plasminogen Activator Inhibitor 1 and Thrombin Is Regulated by the Cofactors Vitronectin and Heparin. Blood 1997. [DOI: 10.1182/blood.v90.5.1874] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractThe interaction of thrombin with plasminogen activator inhibitor 1 (PAI-1) is shown to result in the simultaneous formation of both cleaved PAI-1 and a sodium dodecyl sulfate-stable thrombin-PAI-1 complex. The kinetics of this reaction can be described by a “suicide substrate” mechanism that includes a branched reaction pathway, which terminates in either the stable inhibitor-enzyme complex or the cleaved inhibitor plus free enzyme. Because of the branched pathway, approximately three moles of PAI-1 are needed to completely inhibit one mole of thrombin. Heparin and vitronectin enhance the rate of inhibition from 9.8 × 102 L mol−1 s−1 to 6.2 × 104 L mol−1 s−1 and 2.1 × 105 L mol−1 s−1, respectively, under optimal conditions. In addition to enhancing the rate of inhibition, both cofactors increase the apparent stoichiometry of the PAI-1–thrombin interaction, with cofactor concentration dependencies similar to the inhibition reaction. Thus, at 37°C approximately six cleavage reactions occur per inhibition reaction. Therefore, thrombin will efficiently inactivate PAI-1 in the presence of either vitronectin or heparin, unless a sufficient excess of the inhibitor is present. These results show that physiological cofactors are able to switch a protease-serpin inhibition reaction to a substrate reaction, depending on the local concentrations of each of the components.
Collapse
|
44
|
Van Belle E, Maillard L, Tio FO, Isner JM. Accelerated endothelialization by local delivery of recombinant human vascular endothelial growth factor reduces in-stent intimal formation. Biochem Biophys Res Commun 1997; 235:311-6. [PMID: 9199188 DOI: 10.1006/bbrc.1997.6772] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Endothelium plays an important role in vascular smooth muscle cell proliferation and thrombus deposition. We thus hypothesized that local delivery of recombinant human vascular endothelial growth factor (rhVEGF) might reduce in-stent intimal formation. Balloon injury followed by Palmaz-Schatz stent implantation was performed in the external iliac artery of 30 New Zealand rabbits. Animals were then randomized to: 1) no local delivery (Control group, n=10); 2) local delivery via a channel balloon catheter of 100 microg rhVEGF165 (VEGF group, n=10); or 3) local delivery of the vehicle solution (Vehicle group, n=10). Animals were sacrificed 28 days later and morphometric analysis was performed. Maximal intimal area was reduced from 1.47+/-0.12 mm2 and 1.44+/-0.10 mm2 in the Control and Vehicle groups, respectively, to 0.87+/-0.06 mm2 in the VEGF group (p<.001). Accelerated endothelialization by local delivery of an endothelial-specific growth factor, rhVEGF, significantly reduces in-stent intimal formation and could constitute an attractive alternative to direct antiproliferative strategies.
Collapse
Affiliation(s)
- E Van Belle
- Department of Medicine (Cardiology), St Elizabeth's Medical Center, Tufts University School of Medicine, Boston, Massachusetts 02135, USA
| | | | | | | |
Collapse
|
45
|
Phan VN, Kusuhara M, Lucchesi PA, Berk BC. A 90-kD Na(+)-H+ exchanger kinase has increased activity in spontaneously hypertensive rat vascular smooth muscle cells. Hypertension 1997; 29:1265-72. [PMID: 9180627 DOI: 10.1161/01.hyp.29.6.1265] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Increased activity of the Na(+)-H+ exchanger (NHE-1 isoform) has been observed in cells and tissues from hypertensive humans and animals, including the spontaneously hypertensive rat (SHR). No mutation in NHE-1 DNA sequence or alteration in NHE-1 mRNA and protein expression has been demonstrated in hypertension, indicating that alterations in proteins that regulate NHE-1 activity are responsible for increased activity. The recent finding that NHE-1 phosphorylation in SHR vascular smooth muscle cells (VSMCs) was greater than in Wistar-Kyoto rat (WKY) VSMCs suggested that NHE-1 kinases may represent an abnormal regulatory pathway present in hypertension. To define NHE-1 kinases altered in the hypertensive phenotype. We measured NHE-1 kinase activity by an in-gel-kinase assay using a recombinant glutathione S-transferase NHE-1 fusion protein as a substrate. At least 7 NHE-1 kinases (42 to 90 kD) were present in VSMCs. We studied a 90-kD kinase because it was the major NHE-1 kinase and exhibited differences between SHR and WKY. Comparison of 90-kD kinase activity revealed that SHR VSMCs had increased activity in growth-arrested cells and in cells stimulated by angiotensin II (100 nmol/L for 5 minutes). Activation of the 90-kD kinase by angiotensin II was Ca2+ dependent, PKC independent, and partially dependent on the mitogen-activated protein kinase pathway. These findings indicate that increased activity of a 90-kD NHE-1 kinase is a characteristic of SHR VSMCs in culture and suggest that alterations in the 90-kD NHE-1 kinase and/or proteins that regulate its activity may be a pathogenic component in hypertension in the SHR.
Collapse
Affiliation(s)
- V N Phan
- Department of Medicine, University of Washington, Seattle 98195, USA
| | | | | | | |
Collapse
|
46
|
Abendschein DR, Recchia D, Meng YY, Oltrona L, Wickline SA, Eisenberg PR. Inhibition of thrombin attenuates stenosis after arterial injury in minipigs. J Am Coll Cardiol 1996; 28:1849-55. [PMID: 8962575 DOI: 10.1016/s0735-1097(96)00362-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVES We sought to determine whether brief, profound inhibition of thrombin or prothrombin activation by factor Xa limits neointimal formation and stenosis after arterial injury. BACKGROUND Thrombin has been implicated as a mediator of neointimal formation, but adjunctive administration of anticoagulant agents has not proven effective to decrease restenosis in patients undergoing coronary angioplasty. METHODS We infused recombinant desulfatohirudin (r-hirudin, bolus of 2 mg/kg body weight followed by 2 mg/kg per h, n = 9), heparin (100 U/kg per h, n = 6) or recombinant tick anticoagulant peptide (rTAP, 1-mg/kg bolus followed by 3 mg/kg per h, n = 5), a specific inhibitor of factor Xa, intravenously, beginning 15 min before and for up to 3 h after repetitive balloon hyperinflations sufficient to disrupt the internal elastic lamina in a carotid artery of minipigs with hypercholesterolemia induced by feeding them an atherogenic diet. RESULTS Partial thromboplastin time was increased six- to sevenfold over baseline levels at the end of the infusions of the anticoagulant agents. Lumen stenosis measured histologically 4 weeks after balloon-induced carotid injury was 29 +/- 16% (mean +/- SEM) in r-hirudin-treated, 52 +/- 19% in rTAP-treated and 76 +/- 18% in heparin-treated pigs (p < 0.02 for r-hirudin vs. heparin treatment). CONCLUSIONS The marked reduction of stenosis in r-hirudin-treated animals indicates that thrombin plays a major role in neointimal formation after balloon-induced arterial injury. A relatively brief interval of profound, direct inhibition of thrombin may be particularly effective to attenuate restenosis after balloon angioplasty.
Collapse
Affiliation(s)
- D R Abendschein
- Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri 63110, USA
| | | | | | | | | | | |
Collapse
|
47
|
Nori D, Parikh S, Moni J. Management of peripheral vascular disease: innovative approaches using radiation therapy. Int J Radiat Oncol Biol Phys 1996; 36:847-56. [PMID: 8960512 DOI: 10.1016/s0360-3016(96)00412-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Despite the early successes at vascular recanalization with percutaneous transluminal angioplasty, vascular restenosis has emerged as a clinical problem of near epidemic proportions. Numerous pharmacologic and mechanical adjuncts have been tried with little success. Over the last few years, there have major advances in our understanding of the biology of the restenotic process. The process is now recognized as a proliferation disorder, and therapies akin to those used in the treatment of malignant diseases are being explored. Endovascular brachytherapy has shown strong potential in controlling this pathologic proliferative process in the laboratory and in early clinical studies. In this review we discuss some of the basic issues involved in vascular restenosis and the current status of endovascular brachytherapy.
Collapse
Affiliation(s)
- D Nori
- Department of Radiation Oncology, New York Hospital Medical Center of Queens, Flushing, NY 11355, USA
| | | | | |
Collapse
|
48
|
Yasutake M, Haworth RS, King A, Avkiran M. Thrombin activates the sarcolemmal Na(+)-H+ exchanger. Evidence for a receptor-mediated mechanism involving protein kinase C. Circ Res 1996; 79:705-15. [PMID: 8831494 DOI: 10.1161/01.res.79.4.705] [Citation(s) in RCA: 78] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Thrombin can activate the plasma membrane Na(+)-H+ exchanger in a variety of noncardiac cells. We have studied (1) the effect of thrombin on the activity of the sarcolemmal Na(+)-H+ exchanger in freshly isolated quiescent ventricular myocytes from the adult rat heart and (2) the signaling mechanism(s) underlying any effect. Reverse-transcription polymerase chain reaction analysis revealed thrombin receptor mRNA expression in a myocyte-enriched cell preparation. As an index of Na(+)-H+ exchanger activity, acid efflux rates (JHS) were determined in single myocytes (n = 4 to 11 per group) loaded with the pH-sensitive fluoroprobe carboxy-seminaphthorhodafluor-1 after two consecutive intracellular acid pulses (induced by transient exposure to 20 mmol/L NH4Cl) in bicarbonate-free medium. At a pHi of 6.9, JH did not change significantly during the second pulse relative to the first in control cells. However, when the second pulse occurred in the presence of 0.2, 1, or 5 U/mL thrombin, JH increased by 30%, 62% (P < .05), and 87% (P < .05), respectively. A hexameric thrombin receptor-activating peptide (SFLLRN) mimicked the effect of thrombin and increased JH by 73% (P < .05) at 25 mumol/L. In contrast, an inactive control peptide (FLLRN) was without effect at 25 mumol/L. In cells pretreated with 100 nmol/L GF109203X or 5 mumol/L chelerythrine (protein kinase C inhibitors), neither 5 U/mL thrombin nor 25 mumol/L SFLLRN produced a significant increase in JH. In the presence of 10 mumol/L HOE-694 (a Na(+)-H+ exchanger inhibitor), pHi did not recover after an acid load, even during exposure to 5 U/mL thrombin or 25 mumol/L SFLLRN, confirming that the Na(+)-H+ exchanger was the primary acid efflux mechanism under the conditions used. Neither 5 U/mL thrombin nor 25 mumol/L SFLLRN affected resting pHi and Ca2+ or background acid loading. We conclude that (1) adult rat ventricular myocytes express a functional thrombin receptor, whose stimulation results in increased activity of the sarcolemmal Na(+)-H+ exchanger, and (2) this effect appears to occur through a protein kinase C-mediated mechanism.
Collapse
Affiliation(s)
- M Yasutake
- Rayne Institute, St. Thomas' Hospital, London, UK
| | | | | | | |
Collapse
|
49
|
Wilensky RL, Gradus-Pizlo I, Sandusky G, March KL. Vascular repair mechanisms after directional atherectomy or percutaneous transluminal coronary angioplasty in atherosclerotic rabbit iliac arteries. Am Heart J 1996; 132:13-22. [PMID: 8701854 DOI: 10.1016/s0002-8703(96)90385-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Although directional atherectomy (DA) reduces the plaque burden, successful revascularization is not associated with a reduced restenosis rate when compared with percutaneous transluminal coronary angioplasty (PTCA). The purpose of this study was to compare and contrast the vascular response to DA-induced and PTCA-induced injury. Six to 8 weeks after induction of atherosclerosis, PTCA (n = 34) was performed in one iliac artery and DA in the other (n = 30). Arteries were obtained at 6 time points: 1, 3, 5, 7, 14, and 28 days. Eleven arteries that did not undergo an intervention acted as controls. Radiograms obtained before and after intervention and at euthanization were compared. Morphometric, histologic, and immunohistochemical analyses were performed. Both PTCA and DA resulted in an immediate increase in luminal diameter that subsequently decreased over the ensuing month. PTCA caused deep dissection (7 of 8 arteries), often extending to the adventitia, whereas stand alone DA resulted in deep cleft formation (4 of 5). Of the 30 arteries that underwent DA, 4 exhibited an increase in luminal diameter in the absence of tissue retrieval. Thrombus was observed in both the dissection planes and the clefts within the first 7 days, and cellular ingrowth was appreciated at 5 to 7 days. By 7 days the artery was repaired, and the histologic appearance of the arteries that had undergone PTCA could not be differentiated from the arteries that had undergone DA. Increased intimal and medial collagen and elastin was noted at 14 and 28 days. An increase in the area bordered by the external elastic lamina was observed in both groups. Although successful DA results in tissue removal and the production of a deep tissue cleft and PTCA causes a dissection, both produce a condition in which the arterial injury exposes the arterial media to blood, causing thrombus formation and inflammation with subsequent cellular accumulation into the thrombotic framework.
Collapse
Affiliation(s)
- R L Wilensky
- Krannert Institute of Cardiology, Department of Medicine, Indiana School of Medicine, USA
| | | | | | | |
Collapse
|
50
|
Hubbell JA. Hydrogel systems for barriers and local drug delivery in the control of wound healing. J Control Release 1996. [DOI: 10.1016/0168-3659(95)00162-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|