51
|
Curiel TJ, Cheng P, Mottram P, Alvarez X, Moons L, Evdemon-Hogan M, Wei S, Zou L, Kryczek I, Hoyle G, Lackner A, Carmeliet P, Zou W. Dendritic Cell Subsets Differentially Regulate Angiogenesis in Human Ovarian Cancer. Cancer Res 2004; 64:5535-8. [PMID: 15313886 DOI: 10.1158/0008-5472.can-04-1272] [Citation(s) in RCA: 210] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Angiogenesis is essential for both primary and metastatic tumor growth. Tumor blood vessel formation is complex and regulated by many factors. Ovarian carcinomas have a poor prognosis, often associated with multifocal intraperitoneal dissemination accompanied by intense neovascularization. To examine tumor angiogenesis in the tumor microenvironment, we studied malignant ascites of patients with untreated ovarian carcinoma. We observed high numbers of plasmacytoid dendritic cells (PDCs) and significant stromal-derived factor (CXCL-12/SDF)-1 in their malignant ascites, attracting PDCs into the tumor environment. We now show that tumor-associated PDCs induced angiogenesis in vivo through production of tumor necrosis factor alpha and interleukin 8. By contrast, myeloid dendritic cells (MDCs) were absent from malignant ascites. MDCs derived in vitro suppressed angiogenesis in vivo through production of interleukin 12. Thus, the tumor may attract PDCs to augment angiogenesis while excluding MDCs to prevent angiogenesis inhibition, demonstrating a novel mechanism for modulating tumor neovascularization. Because dendritic cells (DCs) have long been known to affect tumor immunity, our data also implicate DCs in regulation of tumor neoangiogenesis, suggesting a novel role of DCs in tumor pathology.
Collapse
Affiliation(s)
- Tyler J Curiel
- Tulane University Health Science Center, New Orleans, Louisiana 70112, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
52
|
Stafforini DM, McIntyre TM, Zimmerman GA, Prescott SM. Platelet-activating factor, a pleiotrophic mediator of physiological and pathological processes. Crit Rev Clin Lab Sci 2004; 40:643-72. [PMID: 14708958 DOI: 10.1080/714037693] [Citation(s) in RCA: 155] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Platelet-activating factor (PAF) is a potent proinflammatory phospholipid with diverse pathological and physiological effects. This bioactive phospholipid mediates processes as diverse as wound healing, physiological inflammation, apoptosis, angiogenesis, reproduction and long-term potentiation. Recent progress has demonstrated the participation of MAP kinase signaling pathways as modulators of the two critical enzymes, phospholipase A2 and acetyltransferase, involved in the remodeling pathway of PAF biosynthesis. The unregulated production of structural analogs of PAF by non-specific oxidative reactions has expanded this superfamily of signaling molecules to include "PAF-like" lipids whose mode of action is identical to that of authentic PAF. The action of members of this family is mediated by the PAF receptor, a G protein-coupled membrane-spanning molecule that can engage multiple signaling pathways in various cell types. Inappropriate activation of this signaling pathway is associated with many diseases in which inflammation is thought to be one of the underlying features. Inactivation of all members of the PAF superfamily occurs by a unique class of enzymes, the PAF acetylhydrolases, that have been characterized at the molecular level and that terminate signals initiated by both regulated and unregulated PAF production.
Collapse
Affiliation(s)
- Diana M Stafforini
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112-5550, USA.
| | | | | | | |
Collapse
|
53
|
Chen JX, Chen Y, DeBusk L, Lin W, Lin PC. Dual functional roles of Tie-2/angiopoietin in TNF-alpha-mediated angiogenesis. Am J Physiol Heart Circ Physiol 2004; 287:H187-95. [PMID: 15210451 DOI: 10.1152/ajpheart.01058.2003] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Inflammation and angiogenesis are associated with pathological disorders. TNF-alpha is a major inflammatory cytokine that also regulates angiogenesis. TNF-alpha has been shown to regulate Tie-2 and angiopoietin (Ang) expression, but the functional significance is less clear. In this study, we showed that TNF-alpha induced a weak angiogenic response in a mouse cornea assay. Systemic overexpression of Ang-1 or Ang-2 dramatically increased corneal angiogenesis induced by TNF-alpha. In the absence of TNF-alpha, neither Ang-1 nor Ang-2 promoted corneal angiogenesis. Low doses (0-25 ng/ml) of TNF-alpha increased vascular branch formation of cultured endothelial cells. Overexpression of Ang-1 or Ang-2 enhanced the effects of TNF-alpha. These data suggest that Tie-2 signaling synergistically amplifies and participates in TNF-alpha-mediated angiogenesis. In addition, high doses (>/=50 ng/ml) of TNF-alpha induced apoptosis in endothelial cells, but addition of Ang-1 or Ang-2 significantly reduced cell death. Enhanced endothelial cell survival was correlated with Akt phosphorylation. Collectively, our data reveal dual functional roles of Tie-2: low doses enhance TNF-alpha-induced angiogenesis, and high doses attenuate TNF-alpha-induced cell death. The study provides evidence supporting a role for Tie-2 in inflammatory angiogenesis.
Collapse
Affiliation(s)
- Jian-Xiong Chen
- Department of Radiation Oncology, Vanderbilt University Medical Center, 2220 Pierce Ave., Preston Research Bldg., Rm. 315, Nashville, TN 37232, USA
| | | | | | | | | |
Collapse
|
54
|
Russo S, Bussolati B, Deambrosis I, Mariano F, Camussi G. Platelet-activating factor mediates CD40-dependent angiogenesis and endothelial-smooth muscle cell interaction. THE JOURNAL OF IMMUNOLOGY 2004; 171:5489-97. [PMID: 14607955 DOI: 10.4049/jimmunol.171.10.5489] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The aim of the present study was to investigate whether stimulation of CD40 expressed by endothelial or smooth muscle cells triggers the synthesis of platelet-activating factor (PAF), an inflammatory mediator with angiogenic properties, and whether PAF contributes to CD40-induced neoangiogenesis. The results obtained indicate that the interaction of CD40 with soluble CD154 or with CD154 expressed on the membrane of leukocytes (CD154-transfected J558 cells) or of activated platelets, stimulated the synthesis of PAF by endothelial cells but not by smooth cells. The synthesis of PAF triggered by activated platelets was inhibited by a soluble CD40-murine Ig fusion protein that prevents the interaction between membrane CD40 and CD154. Studies with specific inhibitors and evaluation of protein phosphorylation indicated the involvement in PAF synthesis of two intracellular signaling pathways leading to cytosolic phospholipase A(2) activation: a phospholipase Cgamma-protein kinase C-Raf-p42/p44-mitogen-activated protein kinase (MAPK) and a MAPK kinase-3/6-dependent activation of p38 MAPK. PAF synthesized by endothelial cells after CD40 stimulation was instrumental in the in vitro migration and vessel-like organization of endothelial cells, and in the interaction between endothelial cells and smooth muscle cells, as inferred by the inhibitory effect of two different PAF receptor antagonists, WEB2170 and CV3988. In vivo, blockade of PAF receptors prevented the angiogenic effect triggered by CD40 stimulation in a murine model of s.c. Matrigel implantation. In conclusion, these observations indicate that PAF synthesis induced by stimulation of endothelial CD40 contributes to the formation and organization of new vessels. This may be relevant in the vascular remodeling associated with tumor and inflammatory neoangiogenesis.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Azepines/administration & dosage
- CD40 Antigens/immunology
- CD40 Antigens/metabolism
- CD40 Antigens/physiology
- CD40 Ligand/pharmacology
- Cell Communication/immunology
- Cell Movement/immunology
- Cells, Cultured
- Collagen/administration & dosage
- Drug Combinations
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Female
- Humans
- Injections, Subcutaneous
- Intracellular Fluid/immunology
- Intracellular Fluid/metabolism
- Laminin/administration & dosage
- Mice
- Mice, Inbred C57BL
- Models, Immunological
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/immunology
- Muscle, Smooth, Vascular/metabolism
- Neovascularization, Physiologic/drug effects
- Neovascularization, Physiologic/immunology
- Platelet Activating Factor/metabolism
- Platelet Activating Factor/physiology
- Platelet Membrane Glycoproteins/antagonists & inhibitors
- Platelet Membrane Glycoproteins/metabolism
- Proteoglycans/administration & dosage
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, G-Protein-Coupled/metabolism
- Triazoles/administration & dosage
Collapse
Affiliation(s)
- Simona Russo
- Dipartimento di Medicina Interna e di Scienze Cliniche e Biologiche, Università di Torino and Centro Ricerca Medicina Sperimentale, Ospedale San Giovanni Battista, Torino, Italy
| | | | | | | | | |
Collapse
|
55
|
Axelrad TW, Deo DD, Ottino P, Van Kirk J, Bazan NG, Bazan HEP, Hunt JD. Platelet‐activating factor (PAF) induces activation of matrix metalloproteinase 2 activity and vascular endothelial cell invasion and migration. FASEB J 2004; 18:568-70. [PMID: 14715700 DOI: 10.1096/fj.03-0479fje] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Tumor-induced angiogenic responses lead to complex phenotypic changes in vascular endothelial cells, which must coordinate the expression of both proteases and protease inhibitors prior to the proliferation and invasion of surrounding stroma. Matrix metalloproteinase 2 (MMP2), which degrades Type IV collagen, is produced as proMMP2. proMMP2 is activated in part through its interactions with membrane Type 1 MMP (MT1-MMP) and tissue inhibitor of matrix metalloproteinase 2 (TIMP2). In this study, we demonstrate that platelet-activating factor (PAF) is a potent inducer of human umbilical vein endothelial cell (HUVEC) migration and invasion, which is attenuated by PAF receptor antagonists, and that PAF receptor antagonists inhibit the migration and invasion of HUVEC mediated by medium conditioned by a prostatic carcinoma cell line. We confirm that PAF receptor antagonists inhibit proliferation of HUVEC grown in rich growth medium. We show that PAF increases mRNA levels for MT1-MMP and TIMP2, followed by increased temporal conversion of latent proMMP2 to MMP2. Finally, we demonstrate that the ratio of MT1-MMP to TIMP2 in membrane preparations from PAF-stimulated HUVEC is 1.6:1, approximating the hypothesized ideal ratio of 2:1 necessary for the conversion of proMMP2 to MMP2. Our data support the involvement of PAF in vascular endothelial cell migration and invasion.
Collapse
Affiliation(s)
- T William Axelrad
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, USA
| | | | | | | | | | | | | |
Collapse
|
56
|
Deo DD, Bazan NG, Hunt JD. Activation of platelet-activating factor receptor-coupled G alpha q leads to stimulation of Src and focal adhesion kinase via two separate pathways in human umbilical vein endothelial cells. J Biol Chem 2003; 279:3497-508. [PMID: 14617636 DOI: 10.1074/jbc.m304497200] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Platelet-activating factor (PAF), a phospholipid second messenger, has diverse physiological functions, including responses in differentiated endothelial cells to external stimuli. We used human umbilical vein endothelial cells (HUVECs) as a model system. We show that PAF activated pertussis toxin-insensitive G alpha(q) protein upon binding to its seven transmembrane receptor. Elevated cAMP levels were observed via activation of adenylate cyclase, which activated protein kinase A (PKA) and was attenuated by a PAF receptor antagonist, blocking downstream activity. Phosphorylation of Src by PAF required G alpha(q) protein and adenylate cyclase activation; there was an absolute requirement of PKA for PAF-induced Src phosphorylation. Immediate (1 min) PAF-induced STAT-3 phosphorylation required the activation of G alpha(q) protein, adenylate cyclase, and PKA, and was independent of these intermediates at delayed (30 min) and prolonged (60 min) PAF exposure. PAF activated PLC beta 3 through its G alpha(q) protein-coupled receptor, whereas activation of phospholipase C gamma 1 (PLC gamma 1) by PAF was independent of G proteins but required the involvement of Src at prolonged PAF exposure (60 min). We demonstrate for the first time in vascular endothelial cells: (i) the involvement of signaling intermediates in the PAF-PAF receptor system in the induction of TIMP2 and MT1-MMP expression, resulting in the coordinated proteolytic activation of MMP2, and (ii) a receptor-mediated signal transduction cascade for the tyrosine phosphorylation of FAK by PAF. PAF exposure induced binding of p130(Cas), Src, SHC, and paxillin to FAK. Clearly, PAF-mediated signaling in differentiated endothelial cells is critical to endothelial cell functions, including cell migration and proteolytic activation of MMP2.
Collapse
Affiliation(s)
- Dayanand D Deo
- Department of Biochemistry and Molecular Biology, Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, USA
| | | | | |
Collapse
|
57
|
DeBusk LM, Chen Y, Nishishita T, Chen J, Thomas JW, Lin PC. Tie2 receptor tyrosine kinase, a major mediator of tumor necrosis factor alpha-induced angiogenesis in rheumatoid arthritis. ARTHRITIS AND RHEUMATISM 2003; 48:2461-71. [PMID: 13130465 DOI: 10.1002/art.11213] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is an inflammatory disease and an angiogenic disease. However, the molecular mechanisms promoting angiogenesis in RA are not clearly identified. Our objective was to study the role of an endothelium-specific receptor tyrosine kinase, Tie2, in angiogenesis of inflammatory arthritis. METHODS Expression of Tie2 and its ligand, angiopoietin 1 (Ang1), in human synovium was examined by immunohistochemistry and Western blot. A novel synovium vascular window model was established to study the role of Tie2 in angiogenesis in vivo. Primary cultured endothelial cells and synoviocytes were used to study tumor necrosis factor alpha (TNF alpha)-induced Tie2 and Ang1 expression. RESULTS Tie2 was implicated in pathologic angiogenesis. We observed that Tie2 and Ang1 were elevated in human RA synovium. Using a novel collagen-induced arthritis synovial window model, we demonstrated that Tie2 signaling regulated arthritis angiogenesis in vivo. We also showed that Tie2 mediated TNF alpha-induced angiogenesis in a mouse cornea assay. In addition, we observed that TNF alpha can regulate Tie2 activation in multiple ways that may involve interactions between endothelial cells and synoviocytes. TNF alpha up-regulates Tie2 in endothelial cells through nuclear factor kappa B, and it up-regulates Ang1 in synoviocytes. These findings suggest paracrine regulation of angiogenesis between endothelial cells and synoviocytes. CONCLUSION This study demonstrates that Tie2 regulates angiogenesis in inflammatory synovium. Tie2 signaling is an important angiogenic mediator that links the proinflammatory cytokine TNF alpha to pathologic angiogenesis.
Collapse
Affiliation(s)
- Laura M DeBusk
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | | | | | | | | | | |
Collapse
|
58
|
Cai D, Xaymardan M, Holm JM, Zheng J, Kizer JR, Edelberg JM. Age-associated impairment in TNF-alpha cardioprotection from myocardial infarction. Am J Physiol Heart Circ Physiol 2003; 285:H463-9. [PMID: 12730063 DOI: 10.1152/ajpheart.00144.2003] [Citation(s) in RCA: 24] [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: 11/22/2022]
Abstract
Age-associated dysfunction in cardiac microvascular endothelial cells with impaired induction of cardioprotective platelet-derived growth factor (PDGF)-dependent pathways suggests that alterations in critical vascular receptor(s) may contribute to the increased severity of cardiovascular pathology in older persons. In vivo murine phage-display peptide library biopanning revealed a senescent decrease in cardiac microvascular binding of phage epitopes homologous to tumor necrosis factor-alpha (TNF-alpha), suggesting that its receptor(s) may be downregulated in older cardiac endothelial cells. Immunostaining demonstrated that TNF-receptor 1 (TNF-R1) density was significantly lower in the subendocardial endothelium of the aging murine heart. Functional studies confirmed the senescent dysregulation of TNF-alpha receptor pathways, demonstrating that TNF-alpha induced PDGF-B expression in cardiac microvascular endothelial cells of 4-mo-old, but not 24-mo-old, rats. Moreover, TNF-alpha mediated cardioprotective pathways were impaired in the aging heart. In young rat hearts, injection of TNF-alpha significantly reduced the extent of myocardial injury after coronary ligation: TNF-alpha, 7.9 +/- 1.9% left ventricular injury (n = 4) versus PBS, 16.2 +/- 7.9% (n = 10; P < 0.05). The addition of PDGF-AB did not augment the cardioprotective action of TNF-alpha. In myocardial infarctions of older hearts, however, TNF-alpha induced significant postcoronary occlusion mortality (TNF-alpha 80% vs. PBS 0%; n = 10 each, P < 0.05) that was reversed by the coadministration of PDGF-AB. Overall, these studies demonstrate that aging-associated alterations in TNF-alpha receptor cardiac microvascular pathways may contribute to the increased cardiovasular pathology of the aging heart. Strategies targeted at restoring TNF-alpha receptor-mediated expression of PDGF-B may improve cardiac microvascular function and provide novel approaches for treatment and possible prevention of cardiovascular disease in older individuals.
Collapse
Affiliation(s)
- Dongqing Cai
- Department of Medicine, Weill Medical College of Cornell University, 520 East 70th Street, A352, New York, NY 10021, USA
| | | | | | | | | | | |
Collapse
|
59
|
Guenzi E, Töpolt K, Lubeseder-Martellato C, Jörg A, Naschberger E, Benelli R, Albini A, Stürzl M. The guanylate binding protein-1 GTPase controls the invasive and angiogenic capability of endothelial cells through inhibition of MMP-1 expression. EMBO J 2003; 22:3772-82. [PMID: 12881412 PMCID: PMC169055 DOI: 10.1093/emboj/cdg382] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Expression of the large GTPase guanylate binding protein-1 (GBP-1) is induced by inflammatory cytokines (ICs) in endothelial cells (ECs), and the helical domain of the molecule mediates the repression of EC proliferation by ICs. Here we show that the expression of GBP-1 and of the matrix metalloproteinase-1 (MMP-1) are inversely related in vitro and in vivo, and that GBP-1 selectively inhibits the expression of MMP-1 in ECs, but not the expression of other proteases. The GTPase activity of GBP-1 was necessary for this effect, which inhibited invasiveness and tube-forming capability of ECs in three-dimensional collagen-I matrices. A GTPase-deficient mutant (D184N-GBP-1) operated as a transdominant inhibitor of wild-type GBP-1 and rescued MMP-1 expression in the presence of ICs. Expression of D184N-GBP-1, as well as paracrine supplementation of MMP-1, restored the tube-forming capability of ECs in the presence of wild-type GBP-1. The latter finding indicated that the inhibition of capillary formation is specifically due to the repression of MMP-1 expression by GBP-1, and is not affected by the anti-proliferative activity of the helical domain of GBP-1. These findings substantiate the role of GBP-1 as a major regulator of the anti-angiogenic response of ECs to ICs.
Collapse
Affiliation(s)
- Eric Guenzi
- Department of Virus-induced Vasculopathy, Institute of Molecular Virology, GSF-National Research Center for Environment and Health, Neuherberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
60
|
Choi IW, Kim YS, Kim DK, Choi JH, Seo KH, Im SY, Kwon KS, Lee MS, Ha TY, Lee HK. Platelet-activating factor-mediated NF-kappaB dependency of a late anaphylactic reaction. J Exp Med 2003; 198:145-51. [PMID: 12835479 PMCID: PMC2196087 DOI: 10.1084/jem.20022129] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Anaphylaxis is a life-threatening systemic allergic reaction with the potential for a recurrent or biphasic pattern. Despite an incidence of biphasic reaction between 5 and 20%, the molecular mechanism for the reaction is unknown. Using a murine model of penicillin V-induced systemic anaphylaxis, we show an autoregulatory cascade of biphasic anaphylactic reactions. Induction of anaphylaxis caused a rapid increase in circulating platelet-activating factor (PAF) levels. In turn, the elevated PAF contributes to the early phase of anaphylaxis as well as the subsequent activation of the nuclear factor (NF)-kappaB, a crucial transcription factor regulating the expression of many proinflammatory cytokines and immunoregulatory molecules. The induction of NF-kappaB activity is accompanied by TNF-alpha production, which, in turn, promotes late phase PAF synthesis. This secondary wave of PAF production leads eventually to the late phase of anaphylactic reactions. Mast cells do not appear to be required for development of the late phase anaphylaxis. Together, this work reveals the first mechanistic basis for biphasic anaphylactic reactions and provides possible therapeutic strategies for human anaphylaxis.
Collapse
Affiliation(s)
- Il-Whan Choi
- Department of Immunology, University National Medical School, Chonju, Chonbuk, 561-182, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Saeed RW, Peng T, Metz CN. Ascorbic acid blocks the growth inhibitory effect of tumor necrosis factor-alpha on endothelial cells. Exp Biol Med (Maywood) 2003; 228:855-65. [PMID: 12876306 DOI: 10.1177/15353702-0322807-12] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Impaired endothelial cell proliferation has been proposed to be an early, critical defect contributing to the development of atherosclerosis. Recent studies show that high plasma tumor necrosis factor (TNF)-alpha levels and low serum ascorbic acid (AA) levels correlate with atherosclerosis severity. Additionally, AA has been reported to have potential beneficial effects in preventing atherosclerosis. Based on these studies, we investigated the role of AA (< or =1mM) on TNF-alpha-mediated vascular endothelial cell growth inhibition in vitro. In accordance with previous reports, we found that TNF-alpha alone inhibited endothelial cell proliferation. Further studies revealed that AA alone enhanced endothelial cell proliferation and that AA blocked endothelial cell growth inhibition induced by TNF-alpha. By contrast, we observed no effect of AA on endothelial cell activation or nuclear entry of nuclear factor-kappaB in response to TNF-alpha. The protective effect of AA on endothelial cell proliferation was not simply the result of its antioxidant activity but did correlate with collagen IV expression by endothelial cells. AA pre-treatment of proliferating endothelial cells promoted retinoblastoma protein (Rb) phosphorylation and decreased p53 levels when compared to untreated cells. Furthermore, the addition of AA to TNF-alpha-treated proliferating endothelial cells blocked both the inhibition of retinoblastoma protein phosphorylation and enhanced p53 expression induced by TNF-alpha. Consistent with these results, we found that AA protects endothelial cells against TNF-alpha-induced apoptosis. These studies highlight the potential therapeutic role of AA in promoting endothelial cell proliferation during inflammatory conditions, such as atherosclerosis and cardiovascular disease.
Collapse
Affiliation(s)
- Rubina W Saeed
- Laboratory of Vascular Biology, Division of Medicinal Biochemistry, North Shore-Long Island Jewish Research Institute, Manhasset, New York 11030, USA
| | | | | |
Collapse
|
62
|
Han SJ, Ko HM, Choi JH, Seo KH, Lee HS, Choi EK, Choi IW, Lee HK, Im SY. Molecular mechanisms for lipopolysaccharide-induced biphasic activation of nuclear factor-kappa B (NF-kappa B). J Biol Chem 2002; 277:44715-21. [PMID: 12235132 DOI: 10.1074/jbc.m202524200] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The nuclear factor-kappaB (NF-kappaB) is an important transcription factor necessary for initiating and sustaining inflammatory and immune reactions. The inducers of NF-kappaB are well characterized, but the molecular mechanisms underlying multiple in vivo NF-kappaB activation processes are poorly understood. The injection of lipopolysaccharide resulted in a biphasic activation of NF-kappaB during the 18-h observation period in various organs of mice. The early and late phases of NF-kappaB activation occurred at 0.5-2 h and 8-12 h, respectively. Platelet-activating factor, which is released in response to lipopolysaccharide injection, was responsible for the activation of the early phase of NF-kappaB. The early NF-kappaB activity led to the induction of proinflammatory cytokines, tumor necrosis factor (TNF), and interleukin (IL)-1beta, which are known to be efficient inducers of NF-kappaB. Using the TNF knockout and IL-1 receptor knockout mice, we found that TNF and IL-1beta had a role in the second phase activation of NF-kappaB. These cytokines did promote the synthesis of platelet-activating factor, which in turn induced the secondary activation of NF-kappaB. These observations describe a novel autoregulatory molecular mechanism for the biphasic activation of NF-kappaB.
Collapse
Affiliation(s)
- Su-Ji Han
- Department of Biological Sciences, College of Natural Sciences, The Institute of Basic Sciences, Chonnam National University, Kwangju 500-757, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Marmé A, Strauss G, Bastert G, Grischke EM, Moldenhauer G. Intraperitoneal bispecific antibody (HEA125xOKT3) therapy inhibits malignant ascites production in advanced ovarian carcinoma. Int J Cancer 2002; 101:183-9. [PMID: 12209996 DOI: 10.1002/ijc.10562] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Bispecific antibody HEA125 x OKT3 was shown to redirect T lymphocytes toward carcinoma cells and to induce tumor cell lysis in vitro. Preclinical studies have demonstrated that tumor-associated lymphocytes (TAL) derived from malignant ascites can be used as effector cells with a high efficacy and without prior stimulation. These data provided the rationale for a clinical trial to investigate whether bsAb HEA125 x OKT3 is also able to induce tumor cell lysis in vivo and can be used for local treatment of malignant ascites arising from ovarian carcinoma. Ten ovarian carcinoma patients presenting with malignant ascites resistant to chemotherapy were enrolled in the study. They received weekly intraperitoneal injections of 1 mg bsAb diluted in 500 ml NaCl solution to allow homogeneous antibody distribution within the peritoneal cavity. All patients responded clinically well to the therapy. Eight patients experienced a complete and 2 patients a partial reduction of ascites production. A decrease or stabilization of tumor marker CA125 was detected in the sera of 8 patients. Only WHO Grade I and II toxicity was observed including mild fever, chills and allergic eczema. Thus, intraperitoneal application of bsAb HEA125 x OKT3 appears to be an easy and cost effective palliative treatment for ovarian carcinoma with recurrent ascites that leads to a substantially increased quality of life for the patients. During therapy TNF-alpha concentrations raised markedly in the ascites fluid whereas VEGF and sFLT-1 ascites levels declined. This indicates that not only T cell-mediated tumor cell lysis but also changes in vascular permeability due to downregulation of VEGF and its receptors might be responsible for the beneficial therapeutic effect.
Collapse
Affiliation(s)
- Alexander Marmé
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | |
Collapse
|
64
|
Fallani A, Grieco B, Barletta E, Mugnai G, Giorgi G, Salvini L, Ruggieri S. Synthesis of platelet-activating factor (PAF) in transformed cell lines of a different origin. Prostaglandins Other Lipid Mediat 2002; 70:209-26. [PMID: 12428690 DOI: 10.1016/s0090-6980(02)00109-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Interest in the possible involvement of the platelet-activating factor (PAF) in tumor growth and invasiveness has been stimulated by the recognition that PAF influences various biological responses relevant to metastatic diffusion, such as angiogenesis, adhesiveness to endothelia and cellular motility. In the present study, we investigated the extent to which PAF is synthesized by a series of human and murine transformed cell lines of a different histotype. Synthesis of PAF was studied by combining the 14C-acetate incorporation into PAF with the quantitative analysis of PAF performed by a procedure based on gas chromatography-mass spectrometry with a negative ion chemical ionization. In the presence of the Ca2+ ionophore A23187, cultures of human melanoma (Hs294T), fibrosarcoma (HT1080) and colon carcinoma (LS180) cell lines synthesized conspicuous amounts of PAF, comparable to those produced by resident peritoneal macrophages. Substantial quantities of PAF were also synthesized by the murine melanoma (F10-M3 cells). PAF synthesis was rather limited in RSV-transformed Balb/c3T3 (B77-3T3) cells and in one of their high metastatic variants (B77-AA6 cells), although it was more abundant in the latter. We also investigated whether certain cytokines, such as TNFalpha and IFNgamma might induce PAF synthesis in our systems of cell lines which we found to express mRNAs encoding receptors for these cytokines. We observed that PAF synthesis was enhanced in human melanoma and colon carcinoma cell lines and in the murine B77-AA6 cells to levels comparable to those obtained with the Ca2+ ionophore. Synthesis of PAF was not inducible by TNFalpha in murine F10-M3 melanoma cells. IFNgamma also stimulated PAF synthesis in human and murine melanoma lines, and in human LS180 colon carcinoma line, but not in the B77-AA6 cells. PAF synthesis was also inducible by exogenous PAF in the human and murine melanoma lines, and in the human LS180 colon carcinoma line, all of which expressed cell surface PAF receptors. PAF synthesis was not inducible by exogenous PAF in the B77-AA6 cells, which do not express PAF receptors.
Collapse
Affiliation(s)
- Anna Fallani
- Department of Experimental Pathology and Oncology, University of Florence, Italy.
| | | | | | | | | | | | | |
Collapse
|
65
|
Deo DD, Axelrad TW, Robert EG, Marcheselli V, Bazan NG, Hunt JD. Phosphorylation of STAT-3 in response to basic fibroblast growth factor occurs through a mechanism involving platelet-activating factor, JAK-2, and Src in human umbilical vein endothelial cells. Evidence for a dual kinase mechanism. J Biol Chem 2002; 277:21237-45. [PMID: 11940567 DOI: 10.1074/jbc.m110955200] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Platelet-activating factor (PAF) is a potent proinflammatory phospholipid with multiple pathological and physiological effects. We have shown that basic fibroblast growth factor (bFGF) supplementation induces rapid proliferation of human umbilical vein endothelial cells (HUVEC), which is reduced upon removal of bFGF or by bFGF immunoneutralization. The PAF receptor antagonist LAU-8080 inhibited bFGF-stimulated HUVEC proliferation, indicating the involvement of PAF in the bFGF-mediated signaling of HUVEC. Although FGF receptor phosphorylation was not affected by LAU-8080, the bFGF-mediated prolonged phosphorylation, and activation of Erk-1 and -2 were attenuated. Phosphorylation of STAT-3 was observed in the presence of PAF or bFGF, which was attenuated by PAFR antagonists. PAF-induced STAT-3 phosphorylation observed in HUVEC pretreated with either Src inhibitor PP1 or JAK-2 inhibitor AG-490 indicated (i) immediate (1 min) phosphorylation of STAT-3 is dependent on Src, (ii) JAK-2-dependent STAT-3 phosphorylation occurs after the delayed (30 min) PAF exposure, and (iii) prolonged (60 min) STAT-3 phosphorylation may be either through Src and/or JAK-2. Attenuation of the STAT-3 phosphorylation by the PAFR antagonists indicated signaling through the PAF receptor. Taken together, these findings suggest the production of PAF is important for bFGF-mediated signaling and that a dual kinase mechanism is involved in the PAF-mediated signal transduction cascade.
Collapse
Affiliation(s)
- Dayanand D Deo
- Department of Biochemistry and Molecular Biology, Stanley S. Scott Cancer Center and Neuroscience Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, USA
| | | | | | | | | | | |
Collapse
|
66
|
Janin A, Deschaumes C, Daneshpouy M, Estaquier J, Micic-Polianski J, Rajagopalan-Levasseur P, Akarid K, Mounier N, Gluckman E, Socié G, Ameisen JC. CD95 engagement induces disseminated endothelial cell apoptosis in vivo: immunopathologic implications. Blood 2002; 99:2940-7. [PMID: 11929785 DOI: 10.1182/blood.v99.8.2940] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Fas (CD95) is a death receptor involved in apoptosis induction on engagement by Fas ligand (CD95L). Although CD95L-mediated apoptosis has been proposed as a pathogenic mechanism in a wide range of diseases, including graft-versus-host disease, systemic CD95 engagement in mice by agonistic CD95-specific antibodies or by soluble multimeric CD95L (smCD95L), though lethal, has been reported to cause apoptosis only in a limited range of cell types, that is, hepatocytes, hepatic sinusoidal endothelial cells, and lymphocytes. Another member of the tumor necrosis factor (TNF)/CD95L family, TNF-alpha, induces disseminated vascular endothelial cell apoptosis, which precedes apoptosis of other cell types and lethal multiorgan failure. Here we show that systemic CD95 engagement in vivo by agonistic CD95-specific antibody or smCD95L causes rapid, extensive, and disseminated endothelial cell apoptosis throughout the body, by a mechanism that does not depend on TNF-alpha. Disseminated endothelial cell apoptosis was also the first detectable lesion in a murine model of acute tissue damage induced by systemic transfer of allogeneic lymphocytes and did not occur when allogeneic lymphocytes were from CD95L-defective mice. Both vascular and additional tissue lesions induced by agonistic CD95-specific antibody, smCD95L, or allogeneic lymphocytes were prevented by treatment with an inhibitor of caspase-8, the upstream caspase coupled to CD95 death signaling. Vascular lesions are likely to play an important role in the pathogenesis of allogeneic immune responses and of other diseases involving circulating CD95L-expressing cells or smCD95L, and the prevention of CD95-mediated death signaling in endothelial cells may have therapeutic implications in these diseases.
Collapse
Affiliation(s)
- Anne Janin
- EMI-U 9922 INSERM/Université Paris 7, IFR 02, Hôpital Bichat-Claude Bernard, AP-HP, 75877 Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Joseph L, Fink LM, Hauer-Jensen M. Cytokines in coagulation and thrombosis: a preclinical and clinical review. Blood Coagul Fibrinolysis 2002; 13:105-16. [PMID: 11914652 DOI: 10.1097/00001721-200203000-00005] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The cytokine network is a complex and dynamic system, involved in numerous biological responses in the human body. This review of the current literature describes the role of cytokines and their interaction with the coagulation system, specifically in the maintenance of the thrombo-hemorrhagic balance in vivo in human subjects and in animals. In general, cytokines are thrombogenic, but they are amenable to therapeutic manipulations and hence are a potentially attractive tool in the clinician's armamentarium. Studies of the effects of cytokines in vivo are difficult because cytokines act in a very finite microenvironment and, although their actions are significant, they are transient. Most of the available clinical data related to interactions between cytokines and the coagulation system focuses on the role of tumor necrosis factor-alpha and interleukin-1 in septicemia and septic shock. However, several other cytokines and related proteins, such as platelet activating factor and plasminogen activator inhibitor, are also known to influence coagulation and thrombosis. These factors interact closely with cytokines, and have been included in this review for a better understanding of their interactions with traditional cytokines. Studies that utilize cell culture systems do not accurately model the in vivo status of this complex system and, hence, this review has excluded such studies. The role of the cytokine network in coronary artery disease, angiogenesis, or neoplasia has been addressed elsewhere by other workers and is not discussed here. By emphasizing important in vivo interactions, the intention of this review is to serve as an impetus to further translational research, both clinical and in the laboratory.
Collapse
Affiliation(s)
- L Joseph
- Department of Pathology, University of Arkansas for Medical Sciences, and Central Arkansas Veterans Healthcare System, Little Rock, Arkansas 72205, USA.
| | | | | |
Collapse
|
68
|
Guenzi E, Töpolt K, Cornali E, Lubeseder-Martellato C, Jörg A, Matzen K, Zietz C, Kremmer E, Nappi F, Schwemmle M, Hohenadl C, Barillari G, Tschachler E, Monini P, Ensoli B, Stürzl M. The helical domain of GBP-1 mediates the inhibition of endothelial cell proliferation by inflammatory cytokines. EMBO J 2001; 20:5568-77. [PMID: 11598000 PMCID: PMC125279 DOI: 10.1093/emboj/20.20.5568] [Citation(s) in RCA: 150] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Inflammatory cytokines (IC) activate endothelial cell adhesiveness for monocytes and inhibit endothelial cell growth. Here we report the identification of the human guanylate binding protein-1 (GBP-1) as the key and specific mediator of the anti-proliferative effect of IC on endothelial cells. GBP-1 expression was induced by IC, downregulated by angiogenic growth factors, and inversely related to cell proliferation both in vitro in microvascular and macrovascular endothelial cells and in vivo in vessel endothelial cells of Kaposi's sarcoma. Experimental modulation of GBP-1 expression demonstrated that GBP-1 mediates selectively the anti-proliferative effect of IC, without affecting endothelial cell adhesiveness for monocytes. GBP-1 anti-proliferative activity did not affect ERK-1/2 activation, occurred in the absence of apoptosis, was found to be independent of the GTPase activity and isoprenylation of the molecule, but was specifically mediated by the C-terminal helical domain of the protein. These results define GBP-1 as an important tool for dissection of the complex activity of IC on endothelial cells, and detection and specific modulation of the IC-activated non-proliferating phenotype of endothelial cells in vascular diseases.
Collapse
MESH Headings
- Apoptosis/drug effects
- Cell Adhesion/drug effects
- Cell Division/drug effects
- Cells, Cultured/drug effects
- DNA, Antisense/genetics
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/chemistry
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/physiology
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- GTP-Binding Proteins/biosynthesis
- GTP-Binding Proteins/chemistry
- GTP-Binding Proteins/genetics
- GTP-Binding Proteins/physiology
- Gene Expression Profiling
- Gene Expression Regulation/drug effects
- HIV Infections/complications
- Humans
- Inflammation Mediators/antagonists & inhibitors
- Inflammation Mediators/pharmacology
- Interferon-gamma/pharmacology
- MAP Kinase Signaling System/drug effects
- Male
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3
- Mitogen-Activated Protein Kinases/metabolism
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Protein Prenylation
- Protein Processing, Post-Translational
- Protein Structure, Tertiary
- RNA, Messenger/biosynthesis
- Recombinant Fusion Proteins/physiology
- Sarcoma, Kaposi/blood supply
- Sarcoma, Kaposi/etiology
- Sarcoma, Kaposi/pathology
- Skin Neoplasms/blood supply
- Skin Neoplasms/etiology
- Skin Neoplasms/pathology
- Structure-Activity Relationship
- U937 Cells/metabolism
- Umbilical Veins
Collapse
Affiliation(s)
| | | | - Emmanuelle Cornali
- Department of Virus-induced Vasculopathy, Institute of Molecular Virology, GSF-National Research Center for Environment and Health, Ingolstädter Landstrasse 1, D-85764 Neuherberg,
Department of Virus Research, Max Planck Institute for Biochemistry, D-82152 Martinsried, Institute of Pathology, Ludwig Maximilians University, D-80337 Munich, Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, D-81377 Munich, Germany, Laboratory of Virology, Retrovirus Division, Istituto Superiore di Sanità, 00161 Rome, Italy, Department of Virology, Institute of Medical Microbiology and Hygiene, University of Freiburg, D-79104 Freiburg, Germany and Department of Dermatology, Division of Immunology, Allergy, and Infectious Diseases, University of Vienna Medical School, 1090 Vienna, Austria Present address: Ingenium Pharmaceuticals AG, Frauenhoferstrasse 13, D-82152 Martinsried, Germany Present address: Austrian Nordic Biotherapeutics, Veterinärplatz 1, 1210 Vienna, Austria Corresponding author e-mail:
E.Guenzi, K.Töpolt, E.Cornali and C.Lubeseder-Martellato contributed equally to this work
| | | | | | | | - Christian Zietz
- Department of Virus-induced Vasculopathy, Institute of Molecular Virology, GSF-National Research Center for Environment and Health, Ingolstädter Landstrasse 1, D-85764 Neuherberg,
Department of Virus Research, Max Planck Institute for Biochemistry, D-82152 Martinsried, Institute of Pathology, Ludwig Maximilians University, D-80337 Munich, Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, D-81377 Munich, Germany, Laboratory of Virology, Retrovirus Division, Istituto Superiore di Sanità, 00161 Rome, Italy, Department of Virology, Institute of Medical Microbiology and Hygiene, University of Freiburg, D-79104 Freiburg, Germany and Department of Dermatology, Division of Immunology, Allergy, and Infectious Diseases, University of Vienna Medical School, 1090 Vienna, Austria Present address: Ingenium Pharmaceuticals AG, Frauenhoferstrasse 13, D-82152 Martinsried, Germany Present address: Austrian Nordic Biotherapeutics, Veterinärplatz 1, 1210 Vienna, Austria Corresponding author e-mail:
E.Guenzi, K.Töpolt, E.Cornali and C.Lubeseder-Martellato contributed equally to this work
| | - Elisabeth Kremmer
- Department of Virus-induced Vasculopathy, Institute of Molecular Virology, GSF-National Research Center for Environment and Health, Ingolstädter Landstrasse 1, D-85764 Neuherberg,
Department of Virus Research, Max Planck Institute for Biochemistry, D-82152 Martinsried, Institute of Pathology, Ludwig Maximilians University, D-80337 Munich, Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, D-81377 Munich, Germany, Laboratory of Virology, Retrovirus Division, Istituto Superiore di Sanità, 00161 Rome, Italy, Department of Virology, Institute of Medical Microbiology and Hygiene, University of Freiburg, D-79104 Freiburg, Germany and Department of Dermatology, Division of Immunology, Allergy, and Infectious Diseases, University of Vienna Medical School, 1090 Vienna, Austria Present address: Ingenium Pharmaceuticals AG, Frauenhoferstrasse 13, D-82152 Martinsried, Germany Present address: Austrian Nordic Biotherapeutics, Veterinärplatz 1, 1210 Vienna, Austria Corresponding author e-mail:
E.Guenzi, K.Töpolt, E.Cornali and C.Lubeseder-Martellato contributed equally to this work
| | - Filomena Nappi
- Department of Virus-induced Vasculopathy, Institute of Molecular Virology, GSF-National Research Center for Environment and Health, Ingolstädter Landstrasse 1, D-85764 Neuherberg,
Department of Virus Research, Max Planck Institute for Biochemistry, D-82152 Martinsried, Institute of Pathology, Ludwig Maximilians University, D-80337 Munich, Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, D-81377 Munich, Germany, Laboratory of Virology, Retrovirus Division, Istituto Superiore di Sanità, 00161 Rome, Italy, Department of Virology, Institute of Medical Microbiology and Hygiene, University of Freiburg, D-79104 Freiburg, Germany and Department of Dermatology, Division of Immunology, Allergy, and Infectious Diseases, University of Vienna Medical School, 1090 Vienna, Austria Present address: Ingenium Pharmaceuticals AG, Frauenhoferstrasse 13, D-82152 Martinsried, Germany Present address: Austrian Nordic Biotherapeutics, Veterinärplatz 1, 1210 Vienna, Austria Corresponding author e-mail:
E.Guenzi, K.Töpolt, E.Cornali and C.Lubeseder-Martellato contributed equally to this work
| | - Martin Schwemmle
- Department of Virus-induced Vasculopathy, Institute of Molecular Virology, GSF-National Research Center for Environment and Health, Ingolstädter Landstrasse 1, D-85764 Neuherberg,
Department of Virus Research, Max Planck Institute for Biochemistry, D-82152 Martinsried, Institute of Pathology, Ludwig Maximilians University, D-80337 Munich, Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, D-81377 Munich, Germany, Laboratory of Virology, Retrovirus Division, Istituto Superiore di Sanità, 00161 Rome, Italy, Department of Virology, Institute of Medical Microbiology and Hygiene, University of Freiburg, D-79104 Freiburg, Germany and Department of Dermatology, Division of Immunology, Allergy, and Infectious Diseases, University of Vienna Medical School, 1090 Vienna, Austria Present address: Ingenium Pharmaceuticals AG, Frauenhoferstrasse 13, D-82152 Martinsried, Germany Present address: Austrian Nordic Biotherapeutics, Veterinärplatz 1, 1210 Vienna, Austria Corresponding author e-mail:
E.Guenzi, K.Töpolt, E.Cornali and C.Lubeseder-Martellato contributed equally to this work
| | - Christine Hohenadl
- Department of Virus-induced Vasculopathy, Institute of Molecular Virology, GSF-National Research Center for Environment and Health, Ingolstädter Landstrasse 1, D-85764 Neuherberg,
Department of Virus Research, Max Planck Institute for Biochemistry, D-82152 Martinsried, Institute of Pathology, Ludwig Maximilians University, D-80337 Munich, Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, D-81377 Munich, Germany, Laboratory of Virology, Retrovirus Division, Istituto Superiore di Sanità, 00161 Rome, Italy, Department of Virology, Institute of Medical Microbiology and Hygiene, University of Freiburg, D-79104 Freiburg, Germany and Department of Dermatology, Division of Immunology, Allergy, and Infectious Diseases, University of Vienna Medical School, 1090 Vienna, Austria Present address: Ingenium Pharmaceuticals AG, Frauenhoferstrasse 13, D-82152 Martinsried, Germany Present address: Austrian Nordic Biotherapeutics, Veterinärplatz 1, 1210 Vienna, Austria Corresponding author e-mail:
E.Guenzi, K.Töpolt, E.Cornali and C.Lubeseder-Martellato contributed equally to this work
| | - Giovanni Barillari
- Department of Virus-induced Vasculopathy, Institute of Molecular Virology, GSF-National Research Center for Environment and Health, Ingolstädter Landstrasse 1, D-85764 Neuherberg,
Department of Virus Research, Max Planck Institute for Biochemistry, D-82152 Martinsried, Institute of Pathology, Ludwig Maximilians University, D-80337 Munich, Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, D-81377 Munich, Germany, Laboratory of Virology, Retrovirus Division, Istituto Superiore di Sanità, 00161 Rome, Italy, Department of Virology, Institute of Medical Microbiology and Hygiene, University of Freiburg, D-79104 Freiburg, Germany and Department of Dermatology, Division of Immunology, Allergy, and Infectious Diseases, University of Vienna Medical School, 1090 Vienna, Austria Present address: Ingenium Pharmaceuticals AG, Frauenhoferstrasse 13, D-82152 Martinsried, Germany Present address: Austrian Nordic Biotherapeutics, Veterinärplatz 1, 1210 Vienna, Austria Corresponding author e-mail:
E.Guenzi, K.Töpolt, E.Cornali and C.Lubeseder-Martellato contributed equally to this work
| | - Erwin Tschachler
- Department of Virus-induced Vasculopathy, Institute of Molecular Virology, GSF-National Research Center for Environment and Health, Ingolstädter Landstrasse 1, D-85764 Neuherberg,
Department of Virus Research, Max Planck Institute for Biochemistry, D-82152 Martinsried, Institute of Pathology, Ludwig Maximilians University, D-80337 Munich, Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, D-81377 Munich, Germany, Laboratory of Virology, Retrovirus Division, Istituto Superiore di Sanità, 00161 Rome, Italy, Department of Virology, Institute of Medical Microbiology and Hygiene, University of Freiburg, D-79104 Freiburg, Germany and Department of Dermatology, Division of Immunology, Allergy, and Infectious Diseases, University of Vienna Medical School, 1090 Vienna, Austria Present address: Ingenium Pharmaceuticals AG, Frauenhoferstrasse 13, D-82152 Martinsried, Germany Present address: Austrian Nordic Biotherapeutics, Veterinärplatz 1, 1210 Vienna, Austria Corresponding author e-mail:
E.Guenzi, K.Töpolt, E.Cornali and C.Lubeseder-Martellato contributed equally to this work
| | - Paolo Monini
- Department of Virus-induced Vasculopathy, Institute of Molecular Virology, GSF-National Research Center for Environment and Health, Ingolstädter Landstrasse 1, D-85764 Neuherberg,
Department of Virus Research, Max Planck Institute for Biochemistry, D-82152 Martinsried, Institute of Pathology, Ludwig Maximilians University, D-80337 Munich, Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, D-81377 Munich, Germany, Laboratory of Virology, Retrovirus Division, Istituto Superiore di Sanità, 00161 Rome, Italy, Department of Virology, Institute of Medical Microbiology and Hygiene, University of Freiburg, D-79104 Freiburg, Germany and Department of Dermatology, Division of Immunology, Allergy, and Infectious Diseases, University of Vienna Medical School, 1090 Vienna, Austria Present address: Ingenium Pharmaceuticals AG, Frauenhoferstrasse 13, D-82152 Martinsried, Germany Present address: Austrian Nordic Biotherapeutics, Veterinärplatz 1, 1210 Vienna, Austria Corresponding author e-mail:
E.Guenzi, K.Töpolt, E.Cornali and C.Lubeseder-Martellato contributed equally to this work
| | - Barbara Ensoli
- Department of Virus-induced Vasculopathy, Institute of Molecular Virology, GSF-National Research Center for Environment and Health, Ingolstädter Landstrasse 1, D-85764 Neuherberg,
Department of Virus Research, Max Planck Institute for Biochemistry, D-82152 Martinsried, Institute of Pathology, Ludwig Maximilians University, D-80337 Munich, Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, D-81377 Munich, Germany, Laboratory of Virology, Retrovirus Division, Istituto Superiore di Sanità, 00161 Rome, Italy, Department of Virology, Institute of Medical Microbiology and Hygiene, University of Freiburg, D-79104 Freiburg, Germany and Department of Dermatology, Division of Immunology, Allergy, and Infectious Diseases, University of Vienna Medical School, 1090 Vienna, Austria Present address: Ingenium Pharmaceuticals AG, Frauenhoferstrasse 13, D-82152 Martinsried, Germany Present address: Austrian Nordic Biotherapeutics, Veterinärplatz 1, 1210 Vienna, Austria Corresponding author e-mail:
E.Guenzi, K.Töpolt, E.Cornali and C.Lubeseder-Martellato contributed equally to this work
| | - Michael Stürzl
- Department of Virus-induced Vasculopathy, Institute of Molecular Virology, GSF-National Research Center for Environment and Health, Ingolstädter Landstrasse 1, D-85764 Neuherberg,
Department of Virus Research, Max Planck Institute for Biochemistry, D-82152 Martinsried, Institute of Pathology, Ludwig Maximilians University, D-80337 Munich, Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, D-81377 Munich, Germany, Laboratory of Virology, Retrovirus Division, Istituto Superiore di Sanità, 00161 Rome, Italy, Department of Virology, Institute of Medical Microbiology and Hygiene, University of Freiburg, D-79104 Freiburg, Germany and Department of Dermatology, Division of Immunology, Allergy, and Infectious Diseases, University of Vienna Medical School, 1090 Vienna, Austria Present address: Ingenium Pharmaceuticals AG, Frauenhoferstrasse 13, D-82152 Martinsried, Germany Present address: Austrian Nordic Biotherapeutics, Veterinärplatz 1, 1210 Vienna, Austria Corresponding author e-mail:
E.Guenzi, K.Töpolt, E.Cornali and C.Lubeseder-Martellato contributed equally to this work
| |
Collapse
|
69
|
Del Sorbo L, Arese M, Giraudo E, Tizzani M, Biancone L, Bussolino F, Camussi G. Tat-induced platelet-activating factor synthesis contributes to the angiogenic effect of HIV-1 Tat. Eur J Immunol 2001; 31:376-83. [PMID: 11180101 DOI: 10.1002/1521-4141(200102)31:2<376::aid-immu376>3.0.co;2-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
This study shows that human umbilical cord vein-derived endothelial cells (HUVEC) stimulated with HIV-1 Tat synthesized platelet-activating factor (PAF), a phospholipid mediator of inflammation that possesses angiogenic properties. The synthesis of PAF by HUVEC stimulated with Tat was dose and time dependent. Moreover, in vitro experiments were performed to evaluate whether migration of HUVEC induced by Tat was dependent on the synthesis of PAF. It was found that the cell motility induced by Tat was inhibited by WEB 2170, a specific PAF receptor antagonist. In vivo, the neoangiogenesis induced by Tat was also inhibited by WEB 2170 in a murine model, in which matrigel subcutaneously injected was used as substratum for angiogenesis. These results suggest that the synthesis of PAF by endothelial cells mediates, at least in part, the angiogenic activity of Tat by promoting the endothelial cell migration.
Collapse
Affiliation(s)
- L Del Sorbo
- Dipartimento di Medicina Interna, Università di Torino, Torino, Italy
| | | | | | | | | | | | | |
Collapse
|
70
|
Bassi F, Marchisella C, Schierano G, Gasser E, Montrucchio G, Valente G, Camussi G, Preti G. Detection of platelet-activating factor in gingival tissue surrounding failed dental implants. J Periodontol 2001; 72:57-64. [PMID: 11210074 DOI: 10.1902/jop.2001.72.1.57] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Dental implant therapy has entered routine clinical practice. However, the failure rate of implants at 5 years, due to biological factors, is still around 7%. The pathogenesis of implant loss involves a complex network of cells and inflammatory mediators. This study evaluated platelet-activating factor (PAF), a potent phospholipid mediator of inflammation, in soft tissue surrounding failed dental implants versus healthy implants. METHODS PAF was estimated on extracted lipids by bioassay on washed rabbit platelets; inflammatory cell populations were assessed semiquantitatively after staining, and microvessel density was evaluated after immunohistochemical staining. RESULTS Biologically active PAF was detected in the lipid extracts of samples excised from gingival tissue of patients with failed implants, but not in samples from patients with osseointegrated implants or from healthy edentulous subjects. The amount of PAF detected in failed implants was significantly higher than in healthy implants, suggesting a local production of this mediator. CONCLUSIONS The presence of PAF was associated with histopathological findings of local inflammation and increased blood vessel density.
Collapse
Affiliation(s)
- F Bassi
- Department of Clinical Physiopathology, University of Turin, Italy
| | | | | | | | | | | | | | | |
Collapse
|
71
|
Bussolati B, Biancone L, Cassoni P, Russo S, Rola-Pleszczynski M, Montrucchio G, Camussi G. PAF produced by human breast cancer cells promotes migration and proliferation of tumor cells and neo-angiogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2000; 157:1713-25. [PMID: 11073830 PMCID: PMC1885724 DOI: 10.1016/s0002-9440(10)64808-0] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Platelet-activating factor (PAF), a phospholipid mediator of inflammation, is present in breast cancer tissue and correlates with microvessel density. In the present study, we investigated the biological significance of PAF synthesized within breast cancer. In vitro, we observed the production of PAF by two estrogen-dependent (MCF7 and T-47D) and an estrogen-independent (MDA-MB231) breast cancer cell lines after stimulation with vascular endothelial growth factor, basic fibroblast growth factor, hepatocyte growth factor, tumor necrosis factor, thrombin but not with estrogen, progesterone, and oxytocin. The sensitivity to agonist stimulation and the amount of PAF synthesized as cell-associated or released varied in different cell lines, being higher in MDA-MB231 cells, which are known to be highly invasive. We further demonstrate, by reverse transcriptase-polymerase chain reaction and cytofluorimetry, that all of the breast cancer cells express the PAF receptor and respond to PAF stimulation in terms of proliferation. Moreover, in MDA-MB231 cells PAF elicited cell motility. In vivo, two structurally different PAF receptor antagonists WEB 2170 and CV 3988 significantly reduced the formation of new vessels in a tumor induced by subcutaneous implantation of MDA-MB231 cells into SCID mice. In conclusion, these results suggest that PAF, produced and released by breast cancer cells, can contribute to tumor development by enhancing cell motility and proliferation and by stimulating the angiogenic response.
Collapse
Affiliation(s)
- B Bussolati
- Department of Internal Medicine, University of Torino, Torino, Italy
| | | | | | | | | | | | | |
Collapse
|
72
|
Montrucchio G, Alloatti G, Camussi G. Role of platelet-activating factor in cardiovascular pathophysiology. Physiol Rev 2000; 80:1669-99. [PMID: 11015622 DOI: 10.1152/physrev.2000.80.4.1669] [Citation(s) in RCA: 268] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Platelet-activating factor (PAF) is a phospholipid mediator that belongs to a family of biologically active, structurally related alkyl phosphoglycerides. PAF acts via a specific receptor that is coupled with a G protein, which activates a phosphatidylinositol-specific phospholipase C. In this review we focus on the aspects that are more relevant for the cell biology of the cardiovascular system. The in vitro studies provided evidence for a role of PAF both as intercellular and intracellular messenger involved in cell-to-cell communication. In the cardiovascular system, PAF may have a role in embryogenesis because it stimulates endothelial cell migration and angiogenesis and may affect cardiac function because it exhibits mechanical and electrophysiological actions on cardiomyocytes. Moreover, PAF may contribute to modulation of blood pressure mainly by affecting the renal vascular circulation. In pathological conditions, PAF has been involved in the hypotension and cardiac dysfunctions occurring in various cardiovascular stress situations such as cardiac anaphylaxis and hemorrhagic, traumatic, and septic shock syndromes. In addition, experimental studies indicate that PAF has a critical role in the development of myocardial ischemia-reperfusion injury. Indeed, PAF cooperates in the recruitment of leukocytes in inflamed tissue by promoting adhesion to the endothelium and extravascular transmigration of leukocytes. The finding that human heart can produce PAF, expresses PAF receptor, and is sensitive to the negative inotropic action of PAF suggests that this mediator may have a role also in human cardiovascular pathophysiology.
Collapse
Affiliation(s)
- G Montrucchio
- Laboratorio di Immunopatologia Renale, Dipartimento di Medicina Interna, Dipartimento di Biologia Animale e dell'Uomo e Istituto Nazionale di Fisica della Materia, Università di Torino, Torino, Italy
| | | | | |
Collapse
|
73
|
Montrucchio G, Lupia E, Battaglia E, Del Sorbo L, Boccellino M, Biancone L, Emanuelli G, Camussi G. Platelet-activating factor enhances vascular endothelial growth factor-induced endothelial cell motility and neoangiogenesis in a murine matrigel model. Arterioscler Thromb Vasc Biol 2000; 20:80-8. [PMID: 10634803 DOI: 10.1161/01.atv.20.1.80] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We previously reported that platelet-activating factor (PAF) enhances the angiogenic activity of certain polypeptide mediators such as tumor necrosis factor and hepatocyte growth factor by promoting endothelial cell motility. The purpose of the present study was to evaluate whether the synthesis of PAF induced by vascular endothelial growth factor (VEGF) might affect endothelial cell motility, microvascular permeability, and angiogenesis. The neoangiogenesis and synthesis of PAF induced by VEGF were studied in vivo in a murine Matrigel model. Dermal permeability was studied in mice by injection of (125)I-albumin. The synthesis of PAF, cell motility, and the increased (125)I-albumin transfer across endothelial monolayers were studied in vitro by using cultures of human umbilical cord vein-derived endothelial cells (HUVECs). The results obtained demonstrate that the neoangiogenesis induced by VEGF in vivo was associated with a local synthesis of PAF and was inhibited by WEB2170 and CV3988, 2 chemically unrelated, specific PAF-receptor antagonists. In contrast, WEB2170 did not inhibit VEGF-enhanced dermal permeability, suggesting that the latter was independent of the synthesis of PAF. In vitro, it was found that VEGF induced the synthesis of PAF by HUVECs in a dose- and time-dependent manner. The cell motility induced by VEGF was inhibited by PAF-receptor antagonists. In contrast, VEGF-induced proliferation of HUVECs and albumin transfer through HUVEC monolayer were unaffected by PAF-receptor antagonists. These results suggest that the synthesis of PAF induced by VEGF enhances endothelial cell migration and contributes to the angiogenic effect of VEGF in the in vivo Matrigel model.
Collapse
Affiliation(s)
- G Montrucchio
- Dipartimento di Fisiopatologia Clinica, Università di Torino, Italy
| | | | | | | | | | | | | | | |
Collapse
|
74
|
Biancone L, Cantaluppi V, Boccellino M, Bussolati B, Del Sorbo L, Conaldi PG, Albini A, Toniolo A, Camussi G. Motility induced by human immunodeficiency virus-1 Tat on Kaposi's sarcoma cells requires platelet-activating factor synthesis. THE AMERICAN JOURNAL OF PATHOLOGY 1999; 155:1731-9. [PMID: 10550329 PMCID: PMC1866979 DOI: 10.1016/s0002-9440(10)65488-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
In the present study, we evaluated whether motility of Kaposi's sarcoma (KS) spindle cells induced by HIV-1 Tat protein is dependent on the synthesis of platelet-activating factor (PAF). The results obtained indicate that Tat induced a dose-dependent synthesis of PAF from KS cells at a concentration as low as 0.1 ng/ml. PAF production started rapidly after Tat stimulation, peaking at 30 minutes and declining thereafter. Tat-induced cell migration was also a rapid event starting at 30 minutes. The motility was abrogated by addition of a panel of chemically unrelated PAF receptor antagonists (WEB 2170, CV 3988, CV 6209, and BN 52021), suggesting that the synthesized PAF mediates the motogenic effect of Tat. This effect was also present on cells plated on a type-I collagen-, fibronectin-, or basement membrane extract-coated surface. Expression of PAF receptor-specific mRNA was detected in KS cells. In addition, examination of the cytoskeletal organization showed that Tat-mediated KS cell redistribution of actin filaments and shape change was also inhibited by a PAF receptor antagonist. Moreover, PAF receptor blockade prevented the up-regulation of beta1 integrin and the down-regulation of alphavbeta3 observed after stimulation of KS cells with Tat. In conclusion, the results of the present study indicate that Tat-induced PAF synthesis plays a critical role in triggering the events involved in motility of KS cells.
Collapse
Affiliation(s)
- L Biancone
- Cattedra di Nefrologia, Dipartimento di Medicina Interna, Università di Torino, Torino, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
75
|
González MV, González-Sancho JM, Caelles C, Munoz A, Jiménez B. Hormone-activated nuclear receptors inhibit the stimulation of the JNK and ERK signalling pathways in endothelial cells. FEBS Lett 1999; 459:272-6. [PMID: 10518034 DOI: 10.1016/s0014-5793(99)01257-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Glucocorticoid hormones, retinoids, and vitamin D3 display anti-angiogenic activity in tumor-bearing animals. However, despite their in vivo effect on the tumor vasculature little is known about their mechanism of action. Here we show that the synthetic glucocorticoid dexamethasone (Dex) and retinoic acid (RA) inhibit the activation of c-Jun N-terminal kinase (JNK) and extracellular-regulated kinase (ERK) signalling pathways by the pro-angiogenic agents tumor necrosis factor and vascular endothelial growth factor in endothelial cells. In contrast, Dex and RA failed to inhibit the activation of the p38 mitogen-activated protein kinase cascade. As a number of pro-angiogenic factors activate AP-1 transcription factor via the JNK and ERK pathways, our results suggest that the antagonism with AP-1 may underlie at least partially the anti-angiogenic effect of glucocorticoids and retinoids.
Collapse
Affiliation(s)
- M V González
- Instituto de Investigaciones Biomédicas 'Alberto Sols', Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Arturo Duperier 4, E-28029, Madrid, Spain
| | | | | | | | | |
Collapse
|
76
|
Dentelli P, Sorbo LD, Rosso A, Molinar A, Garbarino G, Camussi G, Pegoraro L, Brizzi MF. Human IL-3 Stimulates Endothelial Cell Motility and Promotes In Vivo New Vessel Formation. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.4.2151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Abstract
Angiogenesis is a critical process for growth of new capillary blood vessels from preexisting capillaries and postcapillary venules, both in physiological and pathological conditions. Endothelial cell proliferation is a major component of angiogenesis and it is regulated by several growth factors. It has been previously shown that the human hemopoietic growth factor IL-3 (hIL-3), predominantly produced by activated T lymphocytes, stimulates both endothelial cell proliferation and functional activation. In the present study, we report that hIL-3 is able to induce directional migration and tube formation of HUVEC. The in vivo neoangiogenetic effect of hIL-3 was also demonstrated in a murine model in which Matrigel was used for the delivery of the cytokine, suggesting a role of hIL-3 in sustaining neoangiogenesis. Challenge of HUVEC with hIL-3 lead to the synthesis of platelet-activating factor (PAF), which was found to act as secondary mediator for hIL-3-mediated endothelial cell motility but not for endothelial cell proliferation. Consistent with the role of STAT5 proteins in regulating IL-3-mediated mitogenic signals, we herein report that, in hIL-3-stimulated HUVEC, the recruitment of STAT5A and STAT5B, by the β common (βc) subunit of the IL-3R, was not affected by PAF receptor blockade.
Collapse
Affiliation(s)
| | - Lorenzo Del Sorbo
- †Fisiopatologia Clinica,Università di Torino, Torino, Italy
- Dipartimento di
| | | | | | | | | | | | | |
Collapse
|
77
|
Zhang Y, Pasparakis M, Kollias G, Simons M. Myocyte-dependent regulation of endothelial cell syndecan-4 expression. Role of TNF-alpha. J Biol Chem 1999; 274:14786-90. [PMID: 10329676 DOI: 10.1074/jbc.274.21.14786] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Syndecan-4 is a unique member of the syndecan gene family that has the ability to bind and activate protein kinase C-alpha. Whereas increased syndecan-4 levels have been noted in ischemic hearts, little is known regarding the regulation of its expression. To investigate the role of cardiac myocytes in induction of syndecan-4 expression, human endothelial cells (ECV304) were exposed to a medium conditioned by primary mouse cardiac myocytes or H9c2 cells. The medium conditioned by hypoxic but not normal myocytes was able to induce syndecan-4 expression in ECV cells. Western analysis of the conditioned medium demonstrated an increased presence of tumor necrosis factor-alpha (TNF-alpha) in the medium conditioned by hypoxic but not normal myoblasts. Primary cardiac myocytes collected from the wild type C57/129 but not the homozygous TNF-alpha-/- knockout mice were able to induce syndecan-4 expression in ECV cells when cultured under hypoxic conditions. In vitro studies demonstrated that TNF-alpha induced endothelial cell syndecan-4 expression by both increasing syndecan-4 gene expression in an NF-kappaB-dependent manner and by prolonging syndecan-4 mRNA half-life. We conclude that TNF-alpha is the principal factor produced by the ischemic myocytes that is responsible for induction of endothelial cell syndecan-4 expression and that this requires both transcriptional and posttranscriptional mechanisms.
Collapse
Affiliation(s)
- Y Zhang
- Angiogenesis Research Center, Cardiovascular Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachussetts 02215, USA
| | | | | | | |
Collapse
|
78
|
Del Sorbo L, DeMartino A, Biancone L, Bussolati B, Conaldi PG, Toniolo A, Camussi G. The synthesis of platelet-activating factor modulates chemotaxis of monocytes induced by HIV-1 Tat. Eur J Immunol 1999; 29:1513-21. [PMID: 10359105 DOI: 10.1002/(sici)1521-4141(199905)29:05<1513::aid-immu1513>3.0.co;2-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
HIV-1 Tat protein has been shown to induce chemotaxis and recruitment of monocytes. In the present study, we evaluated whether HIV-1 Tat protein was able to induce the synthesis of platelet-activating factor (PAF), which is a potent mediator of cell motility, and whether the synthesis of PAF was instrumental in triggering Tat-induced monocyte chemotaxis. The results obtained indicate that Tat, but not gp120 and gp41, induced a time-dependent synthesis of PAF from monocytes at concentration as low as 0.1 ng/ml. As inferred by the inhibitory effect of anti-Flt-1 antibody and by the desensitization of monocytes following preincubation with vascular endothelial growth factor, the synthesis of PAF by monocytes stimulated with Tat was induced by activation of vascular endothelial growth factor receptor 1. Moreover, the Tat-induced chemotaxis of monocytes was abrogated both by WEB 2170 and by CV 3988, two chemically unrelated PAF receptor antagonists, suggesting that the synthesized PAF modulates the chemotactic response of monocytes to Tat. In conclusion, the results of the present study indicate that Tat-induced PAF synthesis plays a critical role in triggering the events involved in the migratory response of monocytes.
Collapse
Affiliation(s)
- L Del Sorbo
- Dipartimento di Medicina Interna, Università di Torino, Italy
| | | | | | | | | | | | | |
Collapse
|
79
|
Brizzi MF, Battaglia E, Montrucchio G, Dentelli P, Del Sorbo L, Garbarino G, Pegoraro L, Camussi G. Thrombopoietin stimulates endothelial cell motility and neoangiogenesis by a platelet-activating factor-dependent mechanism. Circ Res 1999; 84:785-96. [PMID: 10205146 DOI: 10.1161/01.res.84.7.785] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In this study, we demonstrate that human umbilical cord vein-derived endothelial cells (HUVECs) expressed c-Mpl, the thrombopoietin (TPO) receptor, and that TPO activates HUVECs in vitro, as indicated by directional migration, synthesis of 1-alkyl-/1-acyl-platelet-activating factor (PAF) and interleukin-8 (IL-8), and phosphorylation of the signal transducers and activators of transcription (STAT) STAT1 and STAT5B. The observation that WEB 2170 and CV3988, 2 structurally unrelated PAF receptor antagonists, prevented the motility of HUVECs induced by TPO suggests a role of PAF as secondary mediator. Moreover, kinetic analysis of TPO-induced tyrosine phosphorylation of STAT demonstrated that STAT5B activation temporally correlated with the synthesis of PAF. PAF, in turn, induced a rapid tyrosine phosphorylation of STAT5B and PAF receptor blockade, by WEB 2170, preventing both TPO- and PAF-mediated STAT5B activation. The in vivo angiogenic effect of TPO, studied in a mouse model of Matrigel implantation, demonstrated that TPO induced a dose-dependent angiogenic response that required the presence of heparin. Moreover, the in vivo angiogenic effect of TPO was inhibited by the PAF receptor antagonist WEB 2170 but not by the anti-basic fibroblast growth factor neutralizing antibody. These results indicate that the effects of TPO are not restricted to cells of hematopoietic lineages, because TPO is able to activate endothelial cells and to induce an angiogenic response in which the recruitment of endothelial cells is mediated by the synthesis of PAF. Moreover, biochemical analysis supports the hypothesis that STAT5B may be involved in the signaling pathway leading to PAF-dependent angiogenesis.
Collapse
Affiliation(s)
- M F Brizzi
- Dipartimento di Medicina Interna, Università di Torino and Dipartimento di Scienze Cliniche e Biologiche, Torino, Italy
| | | | | | | | | | | | | | | |
Collapse
|
80
|
Artuc M, Hermes B, Steckelings UM, Grützkau A, Henz BM. Mast cells and their mediators in cutaneous wound healing--active participants or innocent bystanders? Exp Dermatol 1999; 8:1-16. [PMID: 10206716 DOI: 10.1111/j.1600-0625.1999.tb00342.x] [Citation(s) in RCA: 147] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Mast cells are traditionally viewed as effector cells of immediate type hypersensitivity reactions. There is, however, a growing body of evidence that the cells might play an important role in the maintenance of tissue homeostasis and repair. We here present our own data and those from the literature elucidating the possible role of mast cells during wound healing. Studies on the fate of mast cells in scars of varying ages suggest that these cells degranulate during wounding, with a marked decrease of chymase-positive cells, although the total number of cells does not decrease, based on SCF-receptor staining. Mast cells contain a plethora of preformed mediators like heparin, histamine, tryptase, chymase, VEGF and TNF-alpha which, on release during the initial stages of wound healing, affect bleeding and subsequent coagulation and acute inflammation. Various additional vasoactive and chemotactic, rapidly generated mediators (C3a, C5a, LTB4, LTC4, PAF) will contribute to these processes, whereas mast cell-derived proinflammatory and growth promoting peptide mediators (VEGF, FGF-2, PDGF, TGF-beta, NGF, IL-4, IL-8) contribute to neoangiogenesis, fibrinogenesis or re-epithelization during the repair process. The increasing number of tryptase-positive mast cells in older scars suggest that these cells continue to be exposed to specific chemotactic, growth- and differentiation-promoting factors throughout the process of tissue remodelling. All these data indicate that mast cells contribute in a major way to wound healing. their role as potential initiators of or as contributors to this process, compared to other cell types, will however have to be further elucidated.
Collapse
Affiliation(s)
- M Artuc
- Department of Dermatology, Charité-Virchow Clinic, Humboldt University, Berlin, Germany
| | | | | | | | | |
Collapse
|
81
|
Montrucchio G, Sapino A, Bussolati B, Ghisolfi G, Rizea-Savu S, Silvestro L, Lupia E, Camussi G. Potential angiogenic role of platelet-activating factor in human breast cancer. THE AMERICAN JOURNAL OF PATHOLOGY 1998; 153:1589-96. [PMID: 9811351 PMCID: PMC1853395 DOI: 10.1016/s0002-9440(10)65747-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
This study investigated the presence of platelet-activating factor (PAF) in the lipid extracts of 18 primary breast carcinomas and 20 control breast tissues. The amount of PAF detected in breast carcinomas was significantly higher than in controls. The mass spectrometric analysis of PAF-bioactive lipid extract from breast carcinomas showed the presence of several molecular species of PAF, including C16-alkylPAF, C18-lysophosphatidylcholine (LPC), C16-LPC, lyso-PAF, and C16-acylPAF. The amount of bioactive PAF extracted from breast specimens significantly correlated with tumor vascularization revealed by the number of CD34-and CD31-positive cells. As C16-alkylPAF was previously shown to induce angiogenesis in vivo, we evaluated whether the thin layer chromatography-purified lipid extracts of breast specimens elicited neoangiogenesis in a murine model of subcutaneous Matrigel injection. The lipid extracts from specimens of breast carcinoma containing high levels of PAF bioactivity, but not from breast carcinomas containing low levels of PAF bioactivity or from normal breast tissue, induced a significant angiogenic response. This angiogenic response was significantly inhibited by the PAF receptor antagonist WEB 2170. T47D and MCF7 breast cancer cell lines, but not an immortalized nontumor breast cell line (MCF10), released PAF in the culture medium. A significant in vivo neoangiogenic response, inhibited by WEB 2170, was elicited by T47D and MCF7 but not by MCF10 culture medium. These results indicate that an increased concentration of PAF is present in tumors with high microvessel density and that PAF may account for the neoangiogenic activity induced in mice by the lipid extracts obtained from breast cancer. A contribution of PAF in the neovascularization of human breast cancer is suggested.
Collapse
Affiliation(s)
- G Montrucchio
- Dipartimento de Fisiopatologia Clinica, Università di Torino, Italy
| | | | | | | | | | | | | | | |
Collapse
|
82
|
Giraudo E, Primo L, Audero E, Gerber HP, Koolwijk P, Soker S, Klagsbrun M, Ferrara N, Bussolino F. Tumor necrosis factor-alpha regulates expression of vascular endothelial growth factor receptor-2 and of its co-receptor neuropilin-1 in human vascular endothelial cells. J Biol Chem 1998; 273:22128-35. [PMID: 9705358 DOI: 10.1074/jbc.273.34.22128] [Citation(s) in RCA: 201] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor-alpha (TNF-alpha) modulates gene expression in endothelial cells and is angiogenic in vivo. TNF-alpha does not activate in vitro migration and proliferation of endothelium, and its angiogenic activity is elicited by synthesis of direct angiogenic inducers or of proteases. Here, we show that TNF-alpha up-regulates in a dose- and time-dependent manner the expression and the function of vascular endothelial growth factor receptor-2 (VEGFR-2) as well as the expression of its co-receptor neuropilin-1 in human endothelium. As inferred by nuclear run-on assay and transient expression of VEGFR-2 promoter-based reporter gene construct, the cytokine increased the transcription of the VEGFR-2 gene. Mithramycin, an inhibitor of binding of nuclear transcription factor Sp1 to the promoter consensus sequence, blocked activation of VEGFR-2, suggesting that the up-regulation of the receptor required Sp1 binding sites. TNF-alpha increased the cellular amounts of VEGFR-2 protein and tripled the high affinity 125I-VEGF-A165 capacity without affecting the Kd of ligand-receptor interaction. As a consequence, TNF-alpha enhanced the migration and the wound healing triggered by VEGF-A165. Since VEGFR-2 mediates angiogenic signals in endothelium, our data indicate that its up-regulation is another mechanism by which TNF-alpha is angiogenic and may provide insight into the mechanism of neovascularization as occurs in TNF-alpha-mediated pathological settings.
Collapse
Affiliation(s)
- E Giraudo
- Vascular Biology Laboratory, Department of Genetics, Biology and Biochemistry, Medical School, University of Torino, Torino, 10126 Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Ahmed A, Dearn S, Shams M, Li XF, Sangha RK, Rola-Pleszczynski M, Jiang J. Localization, quantification, and activation of platelet-activating factor receptor in human endometrium during the menstrual cycle: PAF stimulates NO, VEGF, and FAKpp125. FASEB J 1998; 12:831-43. [PMID: 9657523 DOI: 10.1096/fasebj.12.10.831] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Implantation is characterized by an inflammatory-like response with expansion of extracellular fluid volume, increased vascular permeability, and vasodilatation. These effects are believed to be mediated at the paracrine level by prostaglandin E2 and platelet-activating factor (PAF), but the cellular mechanism (or mechanisms) remains largely unknown. We demonstrate that PAF receptor (PAF-R) immunoreactivity and mRNA are detected in proliferative and secretory endometrial glands, however, the responsiveness of endometrium to physiological concentrations of PAF is confined predominantly to the secretory endometrium. Semiquantitative reverse transcription-polymerase chain reaction revealed that PAF-R transcript levels were highest in the mid-late proliferative and late secretory phases of the cycle. Interaction of PAF with its receptor resulted in the rapid release of nitric oxide (NO), increased expression of vascular endothelial growth factor (VEGF), and activation of FAKpp125, a focal adhesion kinase, demonstrating that the PAF-R is functionally active. Inhibition of NO synthesis by NG-monomethyl-L-arginine produced dose-dependent attenuation of PAF-evoked NO release, indicating NOS activation; the dependency of PAF-evoked NO release on PKC and extracellular Ca2+ was confirmed by PKC inhibitor Ro 31-8220 and by the removal of extracellular Ca2+. PAF up-regulated VEGF gene expression in a concentration- and time-dependent fashion in human endometrial epithelial cell lysates. Transcription of VEGF was rapidly followed by secretion of the protein. These data support our premise that this autocoid acts as an angiogenic mediator in the regeneration of the endometrium after menses and as a vasodilator to promote blastocyst attachment during the implantation process.
Collapse
Affiliation(s)
- A Ahmed
- Department of Obstetrics and Gynaecology, Birmingham Women's Hospital, University of Birmingham, Edgbaston, UK.
| | | | | | | | | | | | | |
Collapse
|
84
|
Jackson JR, Bolognese B, Mangar CA, Hubbard WC, Marshall LA, Winkler JD. The role of platelet activating factor and other lipid mediators in inflammatory angiogenesis. BIOCHIMICA ET BIOPHYSICA ACTA 1998; 1392:145-52. [PMID: 9593866 DOI: 10.1016/s0005-2760(98)00012-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronic inflammatory diseases are often accompanied by intense angiogenesis. A model of inflammatory angiogenesis is the murine air pouch granuloma which has a hyperangiogenic component. Proinflammatory lipid mediator generation is also a hallmark of chronic inflammation and the role of endogenous production of these mediators in angiogenesis is not known. The 14 kDa phospholipase A2 (PLA2) deacylates phospholipid, liberating arachidonic acid, which is used for leukotriene production, and lysophospholipid, which can drive the production of platelet-activating factor (PAF). Therefore, SB 203347, an inhibitor of the 14 kDa PLA2, zileuton, an inhibitor of 5-lipoxygenase, and Ro 24-4736 a PAF receptor antagonist were evaluated for their effects in the murine air pouch granuloma. SB 203347 reduced both LTB4 and PAF, but not PGD2 levels measured in the day 6 granuloma. This correlated with a significant reduction in angiogenesis. Zileuton reduced LTB4 levels as expected, but did not significantly inhibit angiogenesis, whereas Ro 24-4736 potently reduced angiogenesis. These data support the hypothesis that PAF, and to a lesser extent leukotrienes contribute to the angiogenic phenotype in chronic inflammation.
Collapse
Affiliation(s)
- J R Jackson
- Department of Immunopharmacology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA.
| | | | | | | | | | | |
Collapse
|
85
|
Grignolo FM, Emanuelli L, Eandi CM, Brogliatti B. Ocular angiogenesis; a new in vivo experimental model. ACTA OPHTHALMOLOGICA SCANDINAVICA. SUPPLEMENT 1998:10-1. [PMID: 9589706 DOI: 10.1111/j.1600-0420.1997.tb00447.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
86
|
Affiliation(s)
- A Mantovani
- Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy.
| | | | | |
Collapse
|
87
|
Tsoukatos DC, Arborati M, Liapikos T, Clay KL, Murphy RC, Chapman MJ, Ninio E. Copper-catalyzed oxidation mediates PAF formation in human LDL subspecies. Protective role of PAF:acetylhydrolase in dense LDL. Arterioscler Thromb Vasc Biol 1997; 17:3505-12. [PMID: 9437199 DOI: 10.1161/01.atv.17.12.3505] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Free radical-mediated oxidation of cholesterol-rich LDL plays a key role in atherogenesis and involves the formation of oxidized phospholipids with proinflammatory biological activity. We evaluated the production of platelet-activating factor (PAF), a potent inflammatory mediator, in human LDL subspecies on copper-initiated oxidation (4 mumol/L CuCl2, 80 micrograms/mL for hours at 37 degrees C). PAF formation was determined by biological assay of HPLC-purified lipid extracts of copper-oxidized lipoproteins; chemical identity was confirmed by gas chromatographic and mass spectrometric analyses. PAF, characterized as the C16:0 molecular species, was preferentially produced in intermediate LDL (d = 1.029 to 1.039 g/mL) (8.6 +/- 5.7 pmol PAF/3 h per mg LDL protein) and light LDL (d = 1.019 to 1.029 g/mL), but was absent from dense LDL particles (d = 1.050 to 1.063 g/mL). As PAF:acetylhydrolase inactivates PAF and oxidized forms of phosphatidylcholine, we evaluated the relationship of lipoprotein-associated PAF:acetylhydrolase to PAF formation. We confirmed that PAF:acetylhydrolase activity was elevated in native, dense LDL (41.5 +/- 9.5 nmol/min per mg protein) but low in LDL subspecies of light and intermediate density (d 1.020 to 1.039 g/mL) (3.5 +/- 1.6 nmol/min per mg protein) [Tselepis et al, Arterioscler Thromb Vasc Biol. 1995;15:1764-1773]. On copper-mediated oxidation for 3 hours at 37 degrees C, dense LDL particles conserved 20 +/- 14% of their initial enzymatic activity; in contrast, PAF:acetylhydrolase activity was abolished in light and intermediate LDL subspecies. Clearly, the elevated PAF:acetylhydrolase activity of dense LDL efficiently diminishes the potential inflammatory role of endogenously formed PAF; nonetheless, formation of proatherogenic lysophospholipids results. In contrast, LDL particles of the light and intermediate subclasses can accumulate PAF on oxidative modification.
Collapse
Affiliation(s)
- D C Tsoukatos
- Department of Chemistry, University of Ioannina, Greece
| | | | | | | | | | | | | |
Collapse
|
88
|
Im SY, Han SJ, Ko HM, Choi JH, Chun SB, Lee DG, Ha TY, Lee HK. Involvement of nuclear factor-kappa B in platelet-activating factor-mediated tumor necrosis factor-alpha expression. Eur J Immunol 1997; 27:2800-4. [PMID: 9394802 DOI: 10.1002/eji.1830271109] [Citation(s) in RCA: 54] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Tumor necrosis factor (TNF)-alpha and platelet-activating factor (PAF) are important mediators of inflammatory reactions, and their release is controlled by a positive feedback network. However, the regulatory mechanisms underlying the interaction of these two molecules are unknown. Within 10 min of the injection of lipopolysaccharide (LPS) into C57BL/6 mice, effects inducible by PAF such as anaphylactic shock-like symptoms, disseminated intravascular coagulation, and hemorrhage in renal medullae were observed, and all these pathological changes were prevented by the PAF antagonist, BN 50739. The plasma level of PAF after LPS injection reached a peak at 5 min. TNF-alpha gene expression was evident 20 min after LPS injection and was maximal at 40 min, and the level of serum TNF-alpha reached a peak at 1 h. Pretreatment with BN 50739 inhibited LPS-induced TNF-alpha gene expression and protein synthesis in a dose-dependent manner. Injection of PAF or treatment of the macrophage cell line, J774A.1, with PAF activated the transcription factor, nuclear factor (NF)-kappa B, which is essential for inducible TNF-alpha transcription. The activation of NF-kappa B by PAF preceded the LPS-mediated TNF-alpha gene expression. Pretreatment with BN 50739 inhibited LPS-induced mobilization of NF-kappa B in a dose-dependent manner in vivo as well as in vitro. These data suggest that PAF, which is released immediately or shortly after LPS injection, induces the expression of TNF-alpha through the activation of NF-kappa B.
Collapse
Affiliation(s)
- S Y Im
- Department of Microbiology, College of Natural Sciences, Chonnam National University, Kwangju, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
89
|
Montrucchio G, Lupia E, de Martino A, Battaglia E, Arese M, Tizzani A, Bussolino F, Camussi G. Nitric oxide mediates angiogenesis induced in vivo by platelet-activating factor and tumor necrosis factor-alpha. THE AMERICAN JOURNAL OF PATHOLOGY 1997; 151:557-63. [PMID: 9250168 PMCID: PMC1858000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We evaluated the role of an endogenous production of nitric oxide (NO) in the in vitro migration of endothelial cells and in the in vivo angiogenic response elicited by platelet-activating factor (PAF), tumor necrosis factor-alpha (TNF), and basic fibroblast growth factor (bFGF). The NO synthase inhibitor, N omega-nitro-L-arginine-methyl ester (L-NAME), but not its enantiomer D-NAME, prevented chemotaxis of endothelial cells induced in vitro by PAF and by TNF. The motogenic activity of TNF was also inhibited by WEB 2170, a specific PAF-receptor antagonist. In contrast, chemotaxis induced by bFGF was not prevented by L-NAME or by WEB 2170. Angiogenesis was studied in vivo in a murine model in which Matrigel was used as a vehicle for the delivery of mediators. In this model, the angiogenesis induced by PAF and TNF was inhibited by WEB 2170 and L-NAME but not by D-NAME. In contrast, angiogenesis induced by bFGF was not affected by L-NAME or by WEB 2170. TNF, but not bFGF, induced PAF synthesis within Matrigel. These results suggest that NO mediates the angiogenesis induced by PAF as well as that induced by TNF, which is dependent on the production of PAF. In contrast, the angiogenic effect of bFGF appears to be both PAF and NO independent.
Collapse
Affiliation(s)
- G Montrucchio
- Dipartimento di Fisiopatologia Clinica, Università di Torino, Italy
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Biancone L, Martino AD, Orlandi V, Conaldi PG, Toniolo A, Camussi G. Development of inflammatory angiogenesis by local stimulation of Fas in vivo. J Exp Med 1997; 186:147-52. [PMID: 9207009 PMCID: PMC2198950 DOI: 10.1084/jem.186.1.147] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Fas-Fas ligand interaction is thought to be a crucial mechanism in controlling lymphocyte expansion by inducing lymphocyte apoptosis. However, Fas is also broadly expressed on nonlymphoid cells, where its function in vivo remains to be determined. In this study, we describe the development of inflammatory angiogenesis induced by agonistic anti-Fas mAb Jo2 in a murine model where Matrigel is used as a vehicle for the delivery of mediators. The subcutaneous implants in mice of Matrigel containing mAb Jo2 became rapidly infiltrated by endothelial cells and by scattered monocytes and macrophages. After formation and canalization of new vessels, marked intravascular accumulation and extravasation of neutrophils were observed. Several mast cells were also detected in the inflammatory infiltrate. The phenomenon was dose and time dependent and required the presence of heparin. The dependency on activation of Fas is suggested by the observation that the inflammatory angiogenesis was restricted to the agonistic anti-Fas mAb and it was absent in lpr Fas-mutant mice. Apoptotic cells were not detectable at any time inside the implant or in the surrounding tissue, suggesting that angiogenesis and cell infiltration did not result from recruitment of phagocytes by apoptotic cells but rather by a stimulatory signal through Fas-engagement. These findings suggest a role for Fas-Fas ligand interaction in promoting local angiogenesis and inflammation.
Collapse
Affiliation(s)
- L Biancone
- Chair of Nephrology, Department of Clinical and Biological Sciences, University of Pavia, Varese, Italy
| | | | | | | | | | | |
Collapse
|
91
|
Yoshida S, Ono M, Shono T, Izumi H, Ishibashi T, Suzuki H, Kuwano M. Involvement of interleukin-8, vascular endothelial growth factor, and basic fibroblast growth factor in tumor necrosis factor alpha-dependent angiogenesis. Mol Cell Biol 1997; 17:4015-23. [PMID: 9199336 PMCID: PMC232254 DOI: 10.1128/mcb.17.7.4015] [Citation(s) in RCA: 482] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Tumor necrosis factor alpha (TNF-alpha) is a macrophage/monocyte-derived polypeptide which modulates the expression of various genes in vascular endothelial cells and induces angiogenesis. However, the underlying mechanism by which TNF-alpha mediates angiogenesis is not completely understood. In this study, we assessed whether TNF-alpha-induced angiogenesis is mediated through TNF-alpha itself or indirectly through other TNF-alpha-induced angiogenesis-promoting factors. Cellular mRNA levels of interleukin-8 (IL-8), vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), and their receptors were increased after the treatment of human microvascular endothelial cells with TNF-alpha (100 U/ml). TNF-alpha-dependent tubular morphogenesis in vascular endothelial cells was inhibited by the administration of anti-IL-8, anti-VEGF, and anti-bFGF antibodies, and coadministration of all three antibodies almost completely abrogated tubular formation. Moreover, treatment with Sp1, NF-kappaB, and c-Jun antisense oligonucleotides inhibited TNF-alpha-dependent tubular morphogenesis by microvascular endothelial cells. Administration of a NF-kappaB antisense oligonucleotide almost completely inhibited TNF-alpha-dependent IL-8 production and partially abrogated TNF-alpha-dependent VEGF production, and an Sp1 antisense sequence partially inhibited TNF-alpha-dependent production of VEGF. A c-Jun antisense oligonucleotide significantly inhibited TNF-alpha-dependent bFGF production but did not affect the production of IL-8 and VEGF. Administration of an anti-IL-8 or anti-VEGF antibody also blocked TNF-alpha-induced neovascularization in the rabbit cornea in vivo. Thus, angiogenesis by TNF-alpha appears to be modulated through various angiogenic factors, both in vitro and in vivo, and this pathway is controlled through paracrine and/or autocrine mechanisms.
Collapse
Affiliation(s)
- S Yoshida
- Department of Biochemistry, Kyushu University School of Medicine, Fukuoka, Japan
| | | | | | | | | | | | | |
Collapse
|
92
|
Abstract
In the literature, the role of interleukin-1 (IL-1) as an angiogen is controversial. The ability of IL-1-alpha to induce de novo angiogenesis in adult rats was studied using the mesenteric window angiogenesis assay (MWAA). Murine recombinant IL-1-alpha was injected intraperitoneally twice daily on Days 0 to 4 at 11.8 pM, 118 pM, and 1.18 nM and groups of animals were sacrificed on Days 7, 14, 21 and 28; controls received the vehicle. Angiogenesis was quantified in terms of microvascular spatial extension and density using technically independent microscopic techniques and image analysis. Compared with the vehicle control, the treatment with IL-1-alpha at doses of 118 pM and 1.18 nM induced statistically significant angiogenesis throughout the study period, whereas IL-1-alpha at 11.8 pM did not induce significant angiogenesis in statistical terms until Days 21 and 28. Compared with the previously reported angiogenic response to VEGF165, bFGF, IL-8, and TNF-alpha using the rat MWAA and the same standardized experimental protocol, the IL-1-alpha treatment displayed a higher degree of efficacy and potency than that of bFGF, IL-8, and TNF-alpha. Moreover, the duration of the significant response to IL-1-alpha exceeded that of bFGF, IL-8, and TNF-alpha. The present data indicate that IL-1-alpha at near-physiologic doses is a very effective angiogenic factor in the system used here. The response may well be multifactorially mediated, as is discussed, and the molecular mechanisms which are involved remain to be clarified.
Collapse
Affiliation(s)
- K Norrby
- Department of Pathology, Göteborg University, Göteborg, S-413 45, Sweden
| |
Collapse
|
93
|
Abstract
Angiogenesis, the formation of new microvessels from parent microvessels, involves remodeling the basement membrane and interstitial extracellular matrix (ECM) using degrading proteases produced by the endothelial cells (ECs) and other adjacent cells, and the synthesis of ECM molecules by these cells. Degraded ECM releases previously bound heparin-binding cytokines (and growth factors) which are able to act as ligands to high-affinity receptors on various target cells, including ECs. The EC carries receptors for a number of cytokines which are produced by neighboring cells or released from the ECM and which can either induce or suppress the angiogenic phenotype of the EC. ECs are able to synthesize and secrete cytokines with auto- and paracrine effects. Angiogenesis, which virtually never occurs physiologically in adult tissues (except in the ovary, the endometrium and the placenta), is essential in wound healing and inflammation. Angiogenesis is, in fact, strictly controlled by a redundancy of pro- and anti-angiogenic paracrine peptide molecules, some of which have recently been described. The expression and synthesis of two distinct anti-angiogenic factors is, for example, controlled by the p53 tumor suppressor gene. In certain hypoxic conditions, chronic inflammatory diseases and syndromes, angiogenesis is of pathogenic and prognostic significance. Angiogenesis is, moreover, essential for the growth and metastatic spread of solid tumors. This indicates the potential for developing new therapeutic strategies not only for tumors but also in diseases such as rheumatoid arthritis, psoriasis, liver cirrhosis and diabetic retinopathy. Moreover, the therapeutic induction of angiogenesis in ischemic tissues using recombinant cytokines is also promising for clinical application. In fact, the first successful human gene therapy for stimulating angiogenesis has recently been reported.
Collapse
Affiliation(s)
- K Norrby
- Department of Pathology, Göteborg University, Sahlgrenska University Hospital, Sweden
| |
Collapse
|
94
|
Stengel D, Antonucci M, Arborati M, Hourton D, Griglio S, Chapman MJ, Ninio E. Expression of the PAF receptor in human monocyte-derived macrophages is downregulated by oxidized LDL: relevance to the inflammatory phase of atherogenesis. Arterioscler Thromb Vasc Biol 1997; 17:954-62. [PMID: 9157961 DOI: 10.1161/01.atv.17.5.954] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Human monocyte-derived macrophages play a major role in the initiation and progression of atherosclerotic lesions as a result of the production of a wide spectrum of proinflammatory and prothrombotic factors. Among such factors is a potent inflammatory phospholipid, platelet-activating factor (PAF), which is produced after macrophage activation. Because the cells involved in PAF biosynthesis are typically targets for the bioactions of PAF via specific cell surface receptors, we evaluated the expression of the PAF receptor in human monocyte-derived macrophages. Oxidized LDL (oxLDL) exerts multiple cellular effects that enhance lesion progression; we therefore investigated the potential modulation of expression of the macrophage PAF receptor by oxLDL. [3H]PAF bound to adherent human macrophages with a K(d) of 2.1 nmol/L and a B(max) of 19 fmol/10(6) cells; approximately 5300 binding sites per cell were detected. OxLDL (100 microg protein per milliliter) induced a twofold decrease in cellular PAF binding after 3 hours at 37 degrees C. Analysis of macrophage mRNA by reverse transcription-polymerase chain reaction (RT-PCR) revealed two forms corresponding to the PAF receptor, of which the leukocyte type (type 1 promoter) predominated. Expression of PAF receptor mRNA, evaluated by quantitative RT-PCR using an actin or a GAPDH mimic, was progressively reduced (up to 70%) by oxLDL up to 6 hours and remained low for at least 24 hours. Such downregulation was reversible after incubation of the cells for 24 hours in oxLDL-free medium. Addition of forskolin (3 micromol/L) or dibutyryl cAMP (1 mmol/L) to macrophage cultures reproduced the oxLDL-mediated inhibition of PAF receptor expression; carbamyl PAF reduced PAF binding and PAF mRNA to a similar degree (approximately 50%). These data demonstrate that atherogenic oxLDL downregulates the expression of both cellular PAF receptors and PAF receptor mRNA in macrophages, consistent with both a diminished bioresponse to PAF and decreased cell motility. Such diminished bioresponse to a powerful antacoid reflects the suppression of an acute inflammatory reaction, thereby leading to chronic, low-level inflammation, such as that characteristic of fatty streaks and more advanced atherosclerotic plaques.
Collapse
Affiliation(s)
- D Stengel
- INSERM Unité 321, Unité de Recherche sur les Lipoprotéines et l'Athérogénèse, Hôpital de la Pitié, Paris, France.
| | | | | | | | | | | | | |
Collapse
|
95
|
Mantovani A, Bussolino F, Introna M. Cytokine regulation of endothelial cell function: from molecular level to the bedside. IMMUNOLOGY TODAY 1997; 18:231-40. [PMID: 9153955 DOI: 10.1016/s0167-5699(97)81662-3] [Citation(s) in RCA: 292] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- A Mantovani
- Istituto di Ricerche Farmacologiche, Mario Negri, Milano, Italy.
| | | | | |
Collapse
|
96
|
Im SY, Choi JH, Ko HM, Han SJ, Chun SB, Lee HK, Ha TY. A protective role of platelet-activating factor in murine candidiasis. Infect Immun 1997; 65:1321-6. [PMID: 9119469 PMCID: PMC175135 DOI: 10.1128/iai.65.4.1321-1326.1997] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Platelet-activating factor (PAF) is a potent phospholipid-derived modulator of immunological and inflammatory processes. In this study, the role of exogenous and endogenous PAF in resistance to infection with Candida albicans was investigated. Administration of PAF following a lethal challenge of C. albicans significantly protected mice from death and reduced the number of organisms in the kidneys. Neutralization of endogenous PAF with the PAF antagonist BN50739 shortened the mean survival time and increased the number of C. albicans cells per kidney. Shortly after infection of mice (30 min), significant levels of PAF were detected in the serum. PAF-induced protection appears to be mediated through the actions of tumor necrosis factor alpha (TNF-alpha), since pretreatment with anti-TNF-alpha before each injection of PAF abrogated the majority of PAF-induced enhanced resistance. Administration of PAF in vivo elevated serum TNF-alpha levels and TNF-alpha mRNA expression in the kidney. Production of TNF-alpha was markedly diminished by pretreatment with the PAF antagonist BN50739 prior to infection with C. albicans. We conclude that PAF, which is produced during infection with C. albicans, plays an important role in determining the level of resistance to this infectious microorganism. This effect of PAF appears to be mediated, at least in part, through the induction of TNF-alpha.
Collapse
Affiliation(s)
- S Y Im
- Department of Microbiology, College of Natural Sciences, Chonnam National University, Kwangju, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
97
|
Wang H, Tan X, Chang H, Gonzalez-Crussi F, Remick DG, Hsueh W. Regulation of platelet-activating factor receptor gene expression in vivo by endotoxin, platelet-activating factor and endogenous tumour necrosis factor. Biochem J 1997; 322 ( Pt 2):603-8. [PMID: 9065783 PMCID: PMC1218232 DOI: 10.1042/bj3220603] [Citation(s) in RCA: 51] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A competitive PCR assay was developed to quantify platelet-activating factor (PAF) receptor (PAF-R) transcripts in rat tissues using a synthetic RNA as a competitor. We found PAF-R mRNA constitutively expressed in the eight organs tested, with the ileum containing the highest concentration [(3.49+/-0.15) x 10(7) molecules/microg of RNA]. Significant but lower levels were also detected in the jejunum, spleen, lungs, kidneys, heart, stomach and liver. Furthermore we defined the regulatory role of inflammatory mediators in ileal PAF-R gene expression using a rat model of intestinal injury induced by PAF or lipopolysaccharide (LPS). Injection of LPS or low-dose PAF resulted in a marked increase in ileal PAF-R mRNA within 30 min. The up-regulation on PAF-R elicited by PAF was biphasic, peaking first at 90 min, then again at 6 h. In contrast, LPS elicited a weak monophasic response. The second phase of PAF-R mRNA increase after PAF administration was completely abolished by WEB 2170, a PAF antagonist, and partially inhibited by antitumour necrosis factor (TNF) antibody. These observations indicate the involvement of endogenous PAF and TNF in this event. In conclusion, we found: (a) preferential PAF-R expression in the ileum, suggesting a role for PAF in intestinal inflammation; (b) induction of PAF-R expression in vivo by its own agonist; (c) a complex regulation of PAR-R gene expression in vivo involving a network of various pro-inflammatory mediators.
Collapse
Affiliation(s)
- H Wang
- Department of Pathology, Northwestern University, Chicago, IL 60614, U.S.A
| | | | | | | | | | | |
Collapse
|
98
|
Grant DS, Kleinman HK. Regulation of capillary formation by laminin and other components of the extracellular matrix. EXS 1997; 79:317-33. [PMID: 9002225 DOI: 10.1007/978-3-0348-9006-9_13] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The process of angiogenesis (vessel formation) and the resulting stabilization of the mature vessel are complex events that are highly regulated and require signals from both serum and the extracellular matrix. Endothelial cells rest on a specialized thin extracellular matrix known as the basement membrane. Endothelial cells lining normal blood vessels are usually quiescent. When a proper stimulus is present, angiogenesis beings when endothelial cells degrade their basement membrane and invade the surrounding extravascular matrix. Formation of new vessels involves the migration and proliferation of cells. To assist the cells in their migration, the extravascular matrix provides an environment rich in stromal collagen fibers, fibrin, hyaluoronic acid, vitronectin and fibronectin. Once the endothelial cells assemble to form a new vessel, the cells secrete a basement membrane that helps to stabilize and maintain the vessel wall. The basement membrane adheres tightly to cells comprising the vessel wall, provides inductive signals, and plays a important role in the homeostasis of new vessels. We have demonstrated that two major components of the basement membrane, laminin and collagen IV, possess endothelial cell binding sites which regulate vessel stability. In this chapter, we will define the role of these molecules in endothelial cell behavior.
Collapse
Affiliation(s)
- D S Grant
- Cardeza Foundation for Hematological Research, Department of Medicine, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | |
Collapse
|
99
|
Dougherty ST, Eaves CJ, McBride WH, Dougherty GJ. Molecular mechanisms regulating TNF-alpha production by tumor-associated macrophages. Cancer Lett 1997; 111:27-37. [PMID: 9022125 DOI: 10.1016/s0304-3835(96)04490-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The molecular mechanisms that regulate the production and/or functional activity of intratumoral tumor necrosis factor-alpha (TNF-alpha) remain poorly defined. To begin to address this issue we have examined the level of TNF-alpha mRNA and protein produced by macrophages present within immunogenic Fsa-R and non-immunogenic Fsa-N tumors grown in syngeneic Lps(d) C3H/HeJ and Lps(n) C3H/HeN mice. The results obtained indicate that macrophages isolated from tumors grown in Lps(d) C3H/HeJ mice express 5-10-fold less TNF-alpha than equivalent cells present in tumors grown in Lps(n) C3H/HeN mice. These data suggest that the mechanisms that operate within the tumor microenvironment to induce the production of TNF-alpha act, at least in part, via the same signal transduction pathway that is defective in Lps(d) C3H/HeJ mice. Interestingly, despite such differences in TNF-alpha production, tumors inoculated into C3H/HeJ and C3H/HeN mice grew at a similar rate and contained an almost identical proportion of macrophages. Moreover, tumor cells purified from tumors grown in C3H/HeJ and C3H/HeN mice produced similar quantities of the TNF-alpha-inducible cytokine GM-CSF. Thus, although differences in the level of TNF-alpha produced within tumors grown in C3H/HeN and C3H/HeJ mice are readily demonstrable, such differences appear to have little direct impact on the outcome of tumor growth.
Collapse
Affiliation(s)
- S T Dougherty
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, British Columbia, Canada
| | | | | | | |
Collapse
|
100
|
Bussolino F, Albini A, Camussi G, Presta M, Viglietto G, Ziche M, Persico G. Role of soluble mediators in angiogenesis. Eur J Cancer 1996; 32A:2401-12. [PMID: 9059328 DOI: 10.1016/s0959-8049(96)00390-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- F Bussolino
- Dipartimento di Genetica, Biologia e Chimica Medica, University of Torino, Italy
| | | | | | | | | | | | | |
Collapse
|