Shear stress-stimulated endothelial cells induce smooth muscle cell chemotaxis via platelet-derived growth factor-BB and interleukin-1α  Alan Dardik,

Slides:



Advertisements
Similar presentations
Volume 56, Issue 5, Pages (November 1999)
Advertisements

IL-18 Downregulates Collagen Production in Human Dermal Fibroblasts via the ERK Pathway  Hee Jung Kim, Seok Bean Song, Jung Min Choi, Kyung Moon Kim,
Volume 132, Issue 1, Pages (January 2007)
Inhibition of phosphatidylinositol 3-kinase and protein kinase C attenuates extracellular matrix protein-induced vascular smooth muscle cell chemotaxis 
Platelet-Derived Growth Factor-BB Mediates Cell Migration through Induction of Activating Transcription Factor 4 and Tenascin-C  Kristine P. Malabanan,
Chung S. Lim, MRCS, PhD, Serafim Kiriakidis, PhD, Ewa M
The Fumagillin Analogue TNP-470 Inhibits DNA Synthesis of Vascular Smooth Muscle Cells Stimulated by Platelet-Derived Growth Factor and Insulin-like Growth.
Heparin inhibits thrombin-induced mitogen-activated protein kinase signaling in arterial smooth muscle cells  Ulf Hedin, MD, PhD, Günter Daum, PhD, Alexander.
High mobility group box 1 promotes endothelial cell angiogenic behavior in vitro and improves muscle perfusion in vivo in response to ischemic injury 
Synthetic smooth muscle cell phenotype is associated with increased nicotinamide adenine dinucleotide phosphate oxidase activity: Effect on collagen secretion 
Patrick C. H. Hsieh, MD, Richard D. Kenagy, PhD, Eileen R
Regulation of vascular smooth muscle cell expression and function of matrix metalloproteinases is mediated by estrogen and progesterone exposure  Oscar.
Chung S. Lim, MRCS, PhD, Serafim Kiriakidis, PhD, Ewa M
Differential effects of orbital and laminar shear stress on endothelial cells  Alan Dardik, MD, PhD, Leiling Chen, MD, Jared Frattini, MD, Hidenori Asada,
Sustained orbital shear stress stimulates smooth muscle cell proliferation via the extracellular signal-regulated protein kinase 1/2 pathway  Hidenori.
Bertrand Poussier, MD, Alfredo C
Domain-dependent action of urokinase on smooth muscle cell responses
Urokinase-induced smooth muscle cell responses require distinct signaling pathways: A role for the epidermal growth factor receptor  Suzanne M. Nicholl,
Darren J. Bridgewater, Jackie Ho, Victor Sauro, Douglas G. Matsell 
The cyclolignan picropodophyllin attenuates intimal hyperplasia after rat carotid balloon injury by blocking insulin-like growth factor-1 receptor signaling 
Transforming growth factor-β increases vascular smooth muscle cell proliferation through the Smad3 and extracellular signal-regulated kinase mitogen-activated.
Cell migration in response to the amino-terminal fragment of urokinase requires epidermal growth factor receptor activation through an ADAM-mediated mechanism 
Volume 122, Issue 2, Pages (February 2002)
Wai-ki Yiu, MBBS, MRCS, Stephen W. K. Cheng, MS, FRCS, Bauer E
Overexpression of mutated IκBα inhibits vascular smooth muscle cell proliferation and intimal hyperplasia formation  Brian S Zuckerbraun, MD, Carol A.
Volume 56, Issue 5, Pages (November 1999)
Hiroyuki Itoh, MD, Peter R
Omar Araim, MD, James Ballantyne, Andrew L. Waterhouse, PhD, Bauer E
Angiotensin II-induced growth of vascular smooth muscle cells requires an Src- dependent activation of the epidermal growth factor receptor1  Dirk Bokemeyer,
Ambient pressure upregulates nitric oxide synthase in a phosphorylated-extracellular regulated kinase– and protein kinase C–dependent manner  Angela G.
Tissue factor pathway inhibitor-2 is induced by fluid shear stress in vascular smooth muscle cells and affects cell proliferation and survival  Johan.
Transforming growth factor (TGF)-β1-induced human endometrial stromal cell decidualization through extracellular signal-regulated kinase and Smad activation.
Evidence for JNK-dependent up-regulation of proteoglycan synthesis and for activation of JNK1 following cyclical mechanical stimulation in a human chondrocyte.
Richard R. Keen, MD, Kevin D
Volume 56, Issue 4, Pages (October 1999)
Smooth muscle cells cultured from human saphenous vein exhibit increased proliferation, invasion, and mitogen-activated protein kinase activation in vitro.
Matrix metalloproteinases modulated by protein kinase Cε mediate resistin-induced migration of human coronary artery smooth muscle cells  Qinxue Ding,
Modulation of vascular smooth muscle cell alignment by cyclic strain is dependent on reactive oxygen species and P38 mitogen-activated protein kinase 
Akio Horiguchi, Mototsugu Oya, Ken Marumo, Masaru Murai 
Volume 68, Issue 1, Pages (July 2005)
Evidence for low-density lipoprotein–induced expression of connective tissue growth factor in mesangial cells  Mimi Sohn, Yan Tan, Richard L. Klein, Ayad.
The proliferative response to platelet-derived growth factor of smooth muscle cells isolated from synthetic vascular grafts in a canine model  David J.
Sphingosine-1-phosphate–induced smooth muscle cell migration involves the mammalian target of rapamycin  William J. Tanski, MD, Suzanne M. Nicholl, PhD,
Inhibition of phosphatidylinositol 3-kinase and protein kinase C attenuates extracellular matrix protein-induced vascular smooth muscle cell chemotaxis 
Platelet-derived growth factor is a cofactor in the induction of 1α(I) procollagen expression by transforming growth factor β1 in smooth muscle cells 
Gabriele Di Luozzo, MD, Jaya Bhargava, PhD, Richard J. Powell, MD 
Holger Lawall, MD, Peter Bramlage, MD, PhD, Berthold Amann, MD 
Protein kinase C-δ regulates migration and proliferation of vascular smooth muscle cells through the extracellular signal-regulated kinase 1/2  Bo Liu,
Volume 128, Issue 4, Pages (April 2005)
Nitric oxide inhibition increases matrix metalloproteinase–9 expression by rat aortic smooth muscle cells in vitro  Gilbert R. Upchurch, MD, John W. Ford,
Kellie J. White, Vincent J. Maffei, Marvin Newton-West, Robert A
Endothelial cell activation of the smooth muscle cell phosphoinositide 3-kinase/Akt pathway promotes differentiation  David J. Brown, MD, Eva M. Rzucidlo,
Taeseung Lee, MD, Nowokere Esemuede, MD, Bauer E
Antisense basic fibroblast growth factor alters the time course of mitogen-activated protein kinase in arterialized vein graft remodeling  Akimasa Yamashita,
In vitro differences between smooth muscle cells derived from varicose veins and normal veins  Ying Xiao, PhD, Zhibin Huang, MD, Henghui Yin, PhD, Ying.
IL-18 Downregulates Collagen Production in Human Dermal Fibroblasts via the ERK Pathway  Hee Jung Kim, Seok Bean Song, Jung Min Choi, Kyung Moon Kim,
Smooth muscle cell migration and proliferation are mediated by distinct phases of activation of the intracellular messenger mitogen-activated protein.
The effect of growth factors, cytokines, and extracellular matrix proteins on fibronectin production in human vascular smooth muscle cells  Terry L. Kaiura,
Vivian Gahtan, MD, Xiu-Jie Wang, MD, Masataka Ikeda, MD, Alliric I
Volume 70, Issue 5, Pages (September 2006)
Angiogenesis and arteriogenesis in limb ischemia
Melanoma Cells Control HA Synthesis in Peritumoral Fibroblasts via PDGF-AA and PDGF-CC: Impact on Melanoma Cell Proliferation  Anja Willenberg, Anja Saalbach,
Activation of integrin receptors is required for growth factor–induced smooth muscle cell dysfunction  Kyotaro Mawatari, MD, Bo Liu, PhD, K.Craig Kent,
Interleukin-1β inhibits PDGF-BB−induced migration by cooperating with PDGF-BB to induce cyclooxygenase-2 expression in baboon aortic smooth muscle cells 
Oscillatory shear stress increases smooth muscle cell proliferation and akt phosphorylation  Masae Haga, MD, Akimasa Yamashita, MD, PhD, Jacek Paszkowiak,
Autocrine-regulated airway smooth muscle cell migration is dependent on IL-17–induced growth-related oncogenes  Laila A. Al-Alwan, PhD, Ying Chang, PhD,
IGF-1 regulation of type II collagen and MMP-13 expression in rat endplate chondrocytes via distinct signaling pathways  M. Zhang, Ph.D., Q. Zhou, M.D.,
Effects of somatostatin, somatostatin analogs, and endothelial cell somatostatin gene transfer on smooth muscle cell proliferation in vitro  Rajabrata.
Platelet-derived growth factor and extracellular matrix proteins provide a synergistic stimulus for human vascular smooth muscle cell migration  Peter.
Susan M. Nesselroth, MDa, Alliric I
Presentation transcript:

Shear stress-stimulated endothelial cells induce smooth muscle cell chemotaxis via platelet-derived growth factor-BB and interleukin-1α  Alan Dardik, MD, PhD, Akimasa Yamashita, MD, PhD, Faisal Aziz, MD, Hidenori Asada, MD, Bauer E. Sumpio, MD, PhD  Journal of Vascular Surgery  Volume 41, Issue 2, Pages 321-331 (February 2005) DOI: 10.1016/j.jvs.2004.11.016 Copyright © 2005 The Society for Vascular Surgery Terms and Conditions

Fig 1 Shear stress-stimulated endothelial cell-derived conditioned medium (CM) stimulates smooth muscle cell (SMC) chemotaxis. A, Stimulation of SMC migration in response to CM. The increase in SMC migration in response to medium conditioned by shear stress (SS), corresponding to arterial levels of SS (CM 210 rpm), or positive controls (FBS 10%, PDGF-BB) was significant compared with static conditions (SFM or CM static) or PDGF-AA (n=3, *P < .05); medium conditioned by low levels of SS (CM 30 rpm) or cyclic strain (CM CS) did not stimulate SMC migration. PDGF-BB and PDGF-AA were 0.5 ng/mL, in SFM (SFM, serum-free medium; FBS, fetal bovine serum; PDGF, platelet-derived growth factor). B, SMC migration in response to CM is time-dependent. The difference in migration due to CM obtained under static (0 rpm, static-CM) and SS conditions (210 rpm, SS-CM) is significant (n = 7, P <.0001, ANOVA) as is the difference over time (P < .0001). C, SMC migration in response to CM depends on SMC protein synthesis after 16 but not 4 hours. The difference in SMC migration due to SS-CM, at 16 hours, between groups receiving actinomycin D or not, is significant (time 16 hours, last 2 bars; n = 7; *P = .003, ANOVA). Journal of Vascular Surgery 2005 41, 321-331DOI: (10.1016/j.jvs.2004.11.016) Copyright © 2005 The Society for Vascular Surgery Terms and Conditions

Fig 2 Conditioned medium stimulates smooth muscle cell (SMC) migration via platelet-derived growth factor-BB (PDGF-BB) and interleukin-1α (IL-1α). A, Enzyme-linked immunoabsorbent assay (ELISA) determination of the PDGF-BB concentration (pg/mL) in serum-free medium (SFM), fetal bovine serum (FBS) (10%), or conditioned medium (CM) under static conditions (0 rpm), 30 rpm SS, 210 rpm shear stress (SS), or cyclic strain (CS), after 16 hours. The difference between the amount of PDGF-BB in CM 210 rpm and the other groups of CM is significant (n = 4; *P = .02, ANOVA). B, Dose-dependent inhibition of SMC migration with anti-PDGF-BB but not anti-PDGF-AA. Inclusion of anti-PDGF-BB (10 μg/mL) decreased SMC migration 37% in response to CM and 82% in response to PDGF-BB (5 ng/mL; n = 3; *P < .0001, ANOVA). C, Dose-dependent inhibition of SMC migration with neutralizing antibodies (0 to 10 ng/mL) directed against IL-1α. Maximal inhibition (62%) occurred with 10 ng/mL anti-IL-1α (n = 4, *P = .0006, ANOVA). Journal of Vascular Surgery 2005 41, 321-331DOI: (10.1016/j.jvs.2004.11.016) Copyright © 2005 The Society for Vascular Surgery Terms and Conditions

Fig 3 Interleukin-1α (IL-1α) stimulates smooth muscle cell (SMC) migration. A, Dose-dependent stimulation of SMC migration with IL-1α. Maximal SMC migration occurred in response to 0.1 pg/mL (n = 5, P < .0001,* ANOVA); identical concentration of IL-1ra produced no migration (n = 4, P = .023, ANOVA). B, Specificity of IL-1α induced SMC migration. IL-1α–induced SMC migration was inhibited in the presence of anti-IL-1α (10 ng/mL, n = 4, P = .0006, ANOVA) but not in the presence of a similar isotype antibody (anti-P-ERK, 10 ng/mL; p=.34). IL-1α induced SMC migration was also inhibited in the presence of anti-IL-1α receptor (p=.005, ANOVA) (anti-P ERK, antiphospho extracellular signal-regulated protein kinase) . C, Lack of synergy of PDGF-BB and IL-1α to stimulate or inhibit SMC migration. The inclusion of IL-1α (0.1 pg/mL) produced no additional SMC migration beyond that induced by PDGF-BB (0.5 ng/mL) alone (11% increase; n = 3, P = .99, ANOVA). The inclusion of anti-IL-1α (10 ng/mL) produced no additional inhibition of SMC migration due to CM beyond that inhibited by anti-PDGF-BB (10 ng/mL) alone (1% decrease; n = 5, P =.99). Journal of Vascular Surgery 2005 41, 321-331DOI: (10.1016/j.jvs.2004.11.016) Copyright © 2005 The Society for Vascular Surgery Terms and Conditions

Fig 4 Inhibition of smooth muscle cell (SMC) chemotaxis by extracellular signal-regulated protein kinase (ERK1/2) upstream pathway-inhibitor PD98059. Inclusion of PD98059 (10 μmol/L) inhibited SMC migration stimulated by conditioned medium (CM) (n = 3, P = .004, ANOVA), platelet-derived growth factor-BB (PDGF-BB) (0.5 ng/mL; P = .002), or IL-1α (0.1 pg/mL; P = .01). Journal of Vascular Surgery 2005 41, 321-331DOI: (10.1016/j.jvs.2004.11.016) Copyright © 2005 The Society for Vascular Surgery Terms and Conditions

Fig 5 Smooth muscle cell (SMC) extracellular signal-regulated protein kinase (ERK1/2) phosphorylation is stimulated by endothelial cell conditioned medium, platelet-derived growth factor-BB (PDGF-BB), and interleukin-1α (IL-1α). The bar graph represents mean of phosphorylated ERK1/2 normalized to total ERK1/2 bands (n = 3). * P < .05 compared with control conditions. The photomicrograph shows a representative experiment of phosphorylated ERK1/2 bands on Western blot; total ERK1/2 bands demonstrated equivalent loading (data not shown). Shear stress-stimulated SMC ERK1/2 phosphorylation was also inhibited in the presence of PD98059 (10 μM, data not shown). PDGF-BB was 0.5 ng/mL; IL-1α was 0.1 pg/mL. Anti-PDGF-BB, anti-PDGF-AA, and anti-IL-1α were 10 ng/mL. Journal of Vascular Surgery 2005 41, 321-331DOI: (10.1016/j.jvs.2004.11.016) Copyright © 2005 The Society for Vascular Surgery Terms and Conditions