Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Immunol ; 192(7): 3390-8, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24585879

ABSTRACT

RhoA-mediated cytoskeletal rearrangements in endothelial cells (ECs) play an active role in leukocyte transendothelial cell migration (TEM), a normal physiological process in which leukocytes cross the endothelium to enter the underlying tissue. Although much has been learned about RhoA signaling pathways downstream from ICAM-1 in ECs, little is known about the consequences of the tractional forces that leukocytes generate on ECs as they migrate over the surface before TEM. We have found that after applying mechanical forces to ICAM-1 clusters, there is an increase in cellular stiffening and enhanced RhoA signaling compared with ICAM-1 clustering alone. We have identified that leukemia-associated Rho guanine nucleotide exchange factor (LARG), also known as Rho GEF 12 (ARHGEF12) acts downstream of clustered ICAM-1 to increase RhoA activity, and that this pathway is further enhanced by mechanical force on ICAM-1. Depletion of LARG decreases leukocyte crawling and inhibits TEM. To our knowledge, this is the first report of endothelial LARG regulating leukocyte behavior and EC stiffening in response to tractional forces generated by leukocytes.


Subject(s)
Endothelial Cells/immunology , Intercellular Adhesion Molecule-1/immunology , Mechanotransduction, Cellular/immunology , Rho Guanine Nucleotide Exchange Factors/immunology , Transendothelial and Transepithelial Migration/immunology , Blotting, Western , Cells, Cultured , Cytochalasin D/pharmacology , Cytoskeleton/drug effects , Cytoskeleton/immunology , Cytoskeleton/metabolism , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Infant, Newborn , Intercellular Adhesion Molecule-1/metabolism , Microscopy, Fluorescence , Nucleic Acid Synthesis Inhibitors/pharmacology , RNA Interference , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction/immunology , Stress, Mechanical
2.
Elife ; 122023 03 29.
Article in English | MEDLINE | ID: mdl-36989130

ABSTRACT

Endothelial cells line all blood vessels, where they coordinate blood vessel formation and the blood-tissue barrier via regulation of cell-cell junctions. The nucleus also regulates endothelial cell behaviors, but it is unclear how the nucleus contributes to endothelial cell activities at the cell periphery. Here, we show that the nuclear-localized linker of the nucleoskeleton and cytoskeleton (LINC) complex protein SUN1 regulates vascular sprouting and endothelial cell-cell junction morphology and function. Loss of murine endothelial Sun1 impaired blood vessel formation and destabilized junctions, angiogenic sprouts formed but retracted in SUN1-depleted sprouts, and zebrafish vessels lacking Sun1b had aberrant junctions and defective cell-cell connections. At the cellular level, SUN1 stabilized endothelial cell-cell junctions, promoted junction function, and regulated contractility. Mechanistically, SUN1 depletion altered cell behaviors via the cytoskeleton without changing transcriptional profiles. Reduced peripheral microtubule density, fewer junction contacts, and increased catastrophes accompanied SUN1 loss, and microtubule depolymerization phenocopied effects on junctions. Depletion of GEF-H1, a microtubule-regulated Rho activator, or the LINC complex protein nesprin-1 rescued defective junctions of SUN1-depleted endothelial cells. Thus, endothelial SUN1 regulates peripheral cell-cell junctions from the nucleus via LINC complex-based microtubule interactions that affect peripheral microtubule dynamics and Rho-regulated contractility, and this long-range regulation is important for proper blood vessel sprouting and junction integrity.


Subject(s)
Endothelial Cells , Microtubule-Associated Proteins , Animals , Mice , Microtubule-Associated Proteins/metabolism , Endothelial Cells/metabolism , Zebrafish/metabolism , Nuclear Proteins/metabolism , Microtubules/metabolism , Intercellular Junctions/metabolism
3.
J Biol Chem ; 286(14): 12141-8, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21242305

ABSTRACT

SmgGDS is an atypical guanine nucleotide exchange factor (GEF) that promotes both cell proliferation and migration and is up-regulated in several types of cancer. SmgGDS has been previously shown to activate a wide variety of small GTPases, including the Ras family members Rap1a, Rap1b, and K-Ras, as well as the Rho family members Cdc42, Rac1, Rac2, RhoA, and RhoB. In contrast, here we show that SmgGDS exclusively activates RhoA and RhoC among a large panel of purified GTPases. Consistent with the well known properties of GEFs, this activation is catalytic, and SmgGDS preferentially binds to nucleotide-depleted RhoA relative to either GDP- or GTPγS-bound forms. However, mutational analyses indicate that SmgGDS utilizes a distinct exchange mechanism compared with canonical GEFs and in contrast to known GEFs requires RhoA to retain a polybasic region for activation. A homology model of SmgGDS highlights an electronegative surface patch and a highly conserved binding groove. Mutation of either area ablates the ability of SmgGDS to activate RhoA. Finally, the in vitro specificity of SmgGDS for RhoA and RhoC is retained in cells. Together, these results indicate that SmgGDS is a bona fide GEF that specifically activates RhoA and RhoC through a unique mechanism not used by other Rho family exchange factors.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Protein Isoforms/metabolism , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Blotting, Western , Cell Line , Chromatography, Gel , Circular Dichroism , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , Humans , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , rho GTP-Binding Proteins/chemistry , rho GTP-Binding Proteins/genetics , rhoA GTP-Binding Protein/chemistry , rhoA GTP-Binding Protein/genetics
4.
Am J Pathol ; 177(4): 2091-102, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20802176

ABSTRACT

Rac1, a subunit of NADPH oxidase, plays an important role in directed endothelial cell motility. We reported previously that Rac1 activation was necessary for choroidal endothelial cell migration across the retinal pigment epithelium, a critical step in the development of vision-threatening neovascular age-related macular degeneration. Here we explored the roles of Rac1 and NADPH oxidase activation in response to vascular endothelial growth factor treatment in vitro and in a model of laser-induced choroidal neovascularization. We found that vascular endothelial growth factor induced the activation of Rac1 and of NADPH oxidase in cultured human choroidal endothelial cells. Further, vascular endothelial growth factor led to heightened generation of reactive oxygen species from cultured human choroidal endothelial cells, which was prevented by the NADPH oxidase inhibitors, apocynin and diphenyleneiodonium, or the antioxidant, N-acetyl-L-cysteine. In a model of laser-induced injury, inhibition of NADPH oxidase with apocynin significantly reduced reactive oxygen species levels as measured by dihydroethidium fluorescence and the volume of laser-induced choroidal neovascularization. Mice lacking functional p47phox, a subunit of NADPH oxidase, had reduced dihydroethidium fluorescence and choroidal neovascularization compared with wild-type controls. Taken together, these results indicate that vascular endothelial growth factor activates Rac1 upstream from NADPH oxidase in human choroidal endothelial cells and increases generation of reactive oxygen species, contributing to choroidal neovascularization. These steps may contributed to the pathology of neovascular age-related macular degeneration.


Subject(s)
Choroid/blood supply , Choroidal Neovascularization/metabolism , Endothelium, Vascular/metabolism , NADPH Oxidases/metabolism , Pigment Epithelium of Eye/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Acetylcysteine/pharmacology , Adult , Animals , Blotting, Western , Cell Movement/drug effects , Cells, Cultured , Choroid/metabolism , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/etiology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Humans , Immunoenzyme Techniques , Lasers/adverse effects , Macular Degeneration/etiology , Macular Degeneration/metabolism , Macular Degeneration/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidases/antagonists & inhibitors , Onium Compounds/pharmacology , Phosphoproteins/physiology , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
5.
J Biol Chem ; 284(38): 25602-11, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19633358

ABSTRACT

Vascular permeability is a complex process involving the coordinated regulation of multiple signaling pathways in the endothelial cell. It has long been documented that vascular endothelial growth factor (VEGF) greatly enhances microvascular permeability; however, the molecular mechanisms controlling VEGF-induced permeability remain unknown. Treatment of microvascular endothelial cells with VEGF led to an increase in reactive oxygen species (ROS) production. ROS are required for VEGF-induced permeability as treatment with the free radical scavenger, N-acetylcysteine, inhibited this effect. Additionally, treatment with VEGF caused ROS-dependent tyrosine phosphorylation of both vascular-endothelial (VE)-cadherin and beta-catenin. Rac1 was required for the VEGF-induced increase in permeability and adherens junction protein phosphorylation. Knockdown of Rac1 inhibited VEGF-induced ROS production consistent with Rac lying upstream of ROS in this pathway. Collectively, these data suggest that VEGF leads to a Rac-mediated generation of ROS, which, in turn, elevates the tyrosine phosphorylation of VE-cadherin and beta-catenin, ultimately regulating adherens junction integrity.


Subject(s)
Adherens Junctions/metabolism , Capillary Permeability/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Reactive Oxygen Species/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Acetylcysteine/pharmacology , Adherens Junctions/genetics , Antigens, CD/metabolism , Cadherins/metabolism , Capillary Permeability/physiology , Cells, Cultured , Free Radical Scavengers/pharmacology , Humans , Phosphorylation/drug effects , Phosphorylation/physiology , Vascular Endothelial Growth Factor A/genetics , beta Catenin/metabolism , rac1 GTP-Binding Protein/metabolism
6.
Philos Trans R Soc Lond B Biol Sci ; 374(1779): 20180229, 2019 08 19.
Article in English | MEDLINE | ID: mdl-31431179

ABSTRACT

Cells respond and adapt to their physical environments and to the mechanical forces that they experience. The translation of physical forces into biochemical signalling pathways is known as mechanotransduction. In this review, we focus on two aspects of mechanotransduction. First, we consider how forces exerted on cell adhesion molecules at the cell surface regulate the RhoA signalling pathway by controlling the activities of guanine nucleotide exchange factors (GEFs) and GTPase activating proteins (GAPs). In the second part of the review, we discuss how the nucleus contributes to mechanotransduction as a physical structure connected to the cytoskeleton. We focus on recent studies that have either severed the connections between the nucleus and the cytoskeleton, or that have entirely removed the nucleus from cells. These actions reduce the levels of active RhoA, thereby altering the mechanical properties of cells and decreasing their ability to generate tension and respond to external mechanical forces. This article is part of a discussion meeting issue 'Forces in cancer: interdisciplinary approaches in tumour mechanobiology'.


Subject(s)
Cell Membrane/physiology , Cell Nucleus/physiology , Mechanotransduction, Cellular/physiology , Signal Transduction/physiology , rhoA GTP-Binding Protein/physiology , Humans
7.
PLoS One ; 14(12): e0225051, 2019.
Article in English | MEDLINE | ID: mdl-31805065

ABSTRACT

As a key homeostasis regulator in mammals, the MERTK receptor tyrosine kinase is crucial for efferocytosis, a process that requires remodeling of the cell membrane and adjacent actin cytoskeleton. Membrane and cytoskeletal reorganization also occur in endothelial cells during inflammation, particularly during neutrophil transendothelial migration (TEM) and during changes in permeability. However, MERTK's function in endothelial cells remains unclear. This study evaluated the contribution of endothelial MERTK to neutrophil TEM and endothelial barrier function. In vitro experiments using primary human pulmonary microvascular endothelial cells found that neutrophil TEM across the endothelial monolayers was enhanced when MERTK expression in endothelial cells was reduced by siRNA knockdown. Examination of endothelial barrier function revealed increased passage of dextran across the MERTK-depleted monolayers, suggesting that MERTK helps maintain endothelial barrier function. MERTK knockdown also altered adherens junction structure, decreased junction protein levels, and reduced basal Rac1 activity in endothelial cells, providing potential mechanisms of how MERTK regulates endothelial barrier function. To study MERTK's function in vivo, inflammation in the lungs of global Mertk-/- mice was examined during acute pneumonia. In response to P. aeruginosa, more neutrophils were recruited to the lungs of Mertk-/- than wildtype mice. Vascular leakage of Evans blue dye into the lung tissue was also greater in Mertk-/- mice. To analyze endothelial MERTK's involvement in these processes, we generated inducible endothelial cell-specific (iEC) Mertk-/- mice. When similarly challenged with P. aeruginosa, iEC Mertk-/- mice demonstrated no difference in neutrophil TEM into the inflamed lungs or in vascular permeability compared to control mice. These results suggest that deletion of MERTK in human pulmonary microvascular endothelial cells in vitro and in all cells in vivo aggravates the inflammatory response. However, selective MERTK deletion in endothelial cells in vivo failed to replicate this response.


Subject(s)
Endothelial Cells/metabolism , Inflammation/metabolism , Lung/metabolism , c-Mer Tyrosine Kinase/metabolism , Adherens Junctions/metabolism , Animals , Capillary Permeability/physiology , Child , Female , Gene Knockdown Techniques , Humans , Mice , Mice, Knockout , c-Mer Tyrosine Kinase/genetics , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
8.
Mol Biol Cell ; 29(18): 2165-2175, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29995590

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is an incurable disease of the lung that is characterized by excessive deposition of extracellular matrix (ECM), resulting in disruption of normal lung function. The signals regulating fibrosis include both transforming growth factor beta (TGF-ß) and tissue rigidity and a major signaling pathway implicated in fibrosis involves activation of the GTPase RhoA. During studies exploring how elevated RhoA activity is sustained in IPF, we discovered that not only is RhoA activated by profibrotic stimuli but also that the expression of Rnd3, a major antagonist of RhoA activity, and the activity of p190RhoGAP (p190), a Rnd3 effector, are both suppressed in IPF fibroblasts. Restoration of Rnd3 levels in IPF fibroblasts results in an increase in p190 activity, a decrease in RhoA activity and a decrease in the overall fibrotic phenotype. We also find that treatment with IPF drugs nintedanib and pirfenidone decreases the fibrotic phenotype and RhoA activity through up-regulation of Rnd3 expression and p190 activity. These data provide evidence for a pathway in IPF where fibroblasts down-regulate Rnd3 levels and p190 activity to enhance RhoA activity and drive the fibrotic phenotype.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Repressor Proteins/metabolism , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Cell Line , Down-Regulation , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/pathology , Indoles/pharmacology , Phenotype , Pyridones/pharmacology , Signal Transduction , Transforming Growth Factor beta , Up-Regulation
9.
Cancer Cell Int ; 6: 8, 2006 Mar 28.
Article in English | MEDLINE | ID: mdl-16569238

ABSTRACT

BACKGROUND: Plasminogen activators are known to play a key role in the remodeling of bone matrix which occurs during tumor progression, bone metastasis and bone growth. Dysfunctional remodeling of bone matrix gives rise to the osteoblastic and osteolytic lesions seen in association with metastatic cancers. The molecular mechanisms responsible for the development of these lesions are not well understood. Studies were undertaken to address the role of the plasminogen activator system in the regulation of fibronectin matrix assembly in the osteoblast-like cell line, MG-63. RESULTS: Treatment of MG-63 cells with P25, a peptide ligand for uPAR, resulted in an increase in assembly of fibronectin matrix which was associated with an increase in the number of activated beta1 integrins on the cell surface. Overexpression of uPAR in MG-63 cells increased the effect of P25 on fibronectin matrix assembly and beta1 integrin activation. P25 had no effect on uPAR null fibroblasts, confirming a role for uPAR in this process. The addition of plasminogen activator inhibitor Type I (PAI-1) to cells increased the P25-induced fibronectin polymerization, as well as the number of activated integrins. This positive regulation of PAI-1 on fibronectin assembly was independent of PAI-1's anti-proteinase activity, but acted through PAI-1 binding to the somatomedin B domain of vitronectin. CONCLUSION: These results indicate that vitronectin modulates fibronectin matrix assembly in osteosarcoma cells through a novel mechanism involving cross-talk through the plasminogen activator system.

10.
Cancer Res ; 75(7): 1244-54, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25634211

ABSTRACT

Endothelial-to-mesenchymal transition (EndMT) occurs during development and underlies the pathophysiology of multiple diseases. In tumors, unscheduled EndMT generates cancer-associated myofibroblasts that fuel inflammation and fibrosis, and may contribute to vascular dysfunction that promotes tumor progression. We report that freshly isolated subpopulations of tumor-specific endothelial cells (TEC) from a spontaneous mammary tumor model undergo distinct forms of EndMT in response to TGFß stimulation. Although some TECs strikingly upregulate α smooth muscle actin (SMA), a principal marker of EndMT and activated myofibroblasts, counterpart normal mammary gland endothelial cells (NEC) showed little change in SMA expression after TGFß treatment. Compared with NECs, SMA(+) TECs were 40% less motile in wound-healing assays and formed more stable vascular-like networks in vitro when challenged with TGFß. Lineage tracing using ZsGreen(Cdh5-Cre) reporter mice confirmed that only a fraction of vessels in breast tumors contain SMA(+) TECs, suggesting that not all endothelial cells (EC) respond identically to TGFß in vivo. Indeed, examination of 84 TGFß-regulated target genes revealed entirely different genetic signatures in TGFß-stimulated NEC and TEC cultures. Finally, we found that basic FGF (bFGF) exerts potent inhibitory effects on many TGFß-regulated genes but operates in tandem with TGFß to upregulate others. ECs challenged with TGFß secrete bFGF, which blocks SMA expression in secondary cultures, suggesting a cell-autonomous or lateral-inhibitory mechanism for impeding mesenchymal differentiation. Together, our results suggest that TGFß-driven EndMT produces a spectrum of EC phenotypes with different functions that could underlie the plasticity and heterogeneity of the tumor vasculature.


Subject(s)
Endothelial Cells/physiology , Transforming Growth Factor beta/physiology , Animals , Cell Line, Tumor , Cell Movement , Cell Separation , Cell Transdifferentiation , Female , Fibroblast Growth Factor 2/physiology , Mammary Neoplasms, Experimental/pathology , Mice, Transgenic , Neoplasm Transplantation
11.
Methods Mol Biol ; 1046: 335-42, 2013.
Article in English | MEDLINE | ID: mdl-23868598

ABSTRACT

VE-cadherin phosphorylation and binding partner status are important indicators of endothelial permeability. Here we describe several techniques aimed at discerning total tyrosine phosphorylation levels of VE-cadherin, VE-cadherin phosphorylation on specific tyrosine residues, and the ability of VE-cadherin to bind its binding partner beta-catenin. Taken together, these approaches to studying VE-cadherin status on microvascular endothelial cells are excellent complements to traditional permeability assays.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Capillary Permeability , Molecular Biology/methods , Antigens, CD/genetics , Cadherins/genetics , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Phosphorylation , Protein Binding , Tyrosine/metabolism , beta Catenin/metabolism
12.
Methods Mol Biol ; 763: 281-90, 2011.
Article in English | MEDLINE | ID: mdl-21874459

ABSTRACT

Endothelial cells lining the vessels of the vasculature and the cell-cell junctions, which join them, -provide the primary barrier to the passage of fluids, immune cells, and macromolecules between the bloodstream and the tissues. Appropriate and dynamic regulation of this barrier is required during normal -physiological processes; however, if not tightly controlled, increased permeability of the endothelium can also contribute to many pathological situations, including chronic inflammatory diseases and edema. The development of in vitro methods to study endothelial barrier function has been key in the identification of molecular mechanisms underlying many of these disease states. In this chapter, we describe three complementary approaches to measure endothelial monolayer permeability and barrier function in vitro.


Subject(s)
Endothelial Cells/metabolism , Endothelium/metabolism , Intercellular Junctions/metabolism , Potentiometry/methods , Antigens, CD/analysis , Cadherins/analysis , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Dextrans/analysis , Diffusion Chambers, Culture , Electric Impedance , Endothelial Cells/cytology , Endothelium/cytology , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/analysis , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Junctions/drug effects , Permeability/drug effects , Thionucleotides/pharmacology , Thrombin/pharmacology , Vascular Endothelial Growth Factor A/pharmacology
13.
Mol Biol Cell ; 21(9): 1629-42, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20237158

ABSTRACT

RhoG is a member of the Rac-like subgroup of Rho GTPases and has been linked to a variety of different cellular functions. Nevertheless, many aspects of RhoG upstream and downstream signaling remain unclear; in particular, few extracellular stimuli that modulate RhoG activity have been identified. Here, we describe that stimulation of epithelial cells with epidermal growth factor leads to strong and rapid activation of RhoG. Importantly, this rapid activation was not observed with other growth factors tested. The kinetics of RhoG activation after epidermal growth factor (EGF) stimulation parallel the previously described Rac1 activation. However, we show that both GTPases are activated independently of one another. Kinase inhibition studies indicate that the rapid activation of RhoG and Rac1 after EGF treatment requires the activity of the EGF receptor kinase, but neither phosphatidylinositol 3-kinase nor Src kinases. By using nucleotide-free RhoG pull-down assays and small interfering RNA-mediated knockdown studies, we further show that guanine-nucleotide exchange factors (GEFs) of the Vav family mediate EGF-induced rapid activation of RhoG. In addition, we found that in certain cell types the recently described RhoG GEF PLEKHG6 can also contribute to the rapid activation of RhoG after EGF stimulation. Finally, we present results that show that RhoG has functions in EGF-stimulated cell migration and in regulating EGF receptor internalization.


Subject(s)
Epidermal Growth Factor/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Endocytosis/drug effects , ErbB Receptors/metabolism , Guanine Nucleotide Exchange Factors/genetics , HeLa Cells , Humans , Mice , NIH 3T3 Cells , Phosphorylation/drug effects , Platelet-Derived Growth Factor/pharmacology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Vascular Endothelial Growth Factor A/pharmacology , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/genetics
14.
J Cell Sci ; 121(Pt 22): 3693-703, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18957516

ABSTRACT

The relationship between the plasminogen activator system and integrin function is well documented but incompletely understood. The mechanism of uPAR-mediated signaling across the membrane and the molecular basis of uPAR-dependent activation of integrins remain important issues. The present study was undertaken to identify the molecular intermediates involved in the uPAR signaling pathway controlling alpha5beta1-integrin activation and fibronectin polymerization. Disruption of lipid rafts with MbetaCD or depletion of caveolin-1 by siRNA led to the inhibition of uPAR-dependent integrin activation and stimulation of fibronectin polymerization in human dermal fibroblasts. The data indicate a dual role for caveolin-1 in the uPAR signaling pathway, leading to integrin activation. Caveolin-1 functions initially as a membrane adaptor or scaffold to mediate uPAR-dependent activation of Src and EGFR. Subsequently, in its phosphorylated form, caveolin-1 acts as an accessory molecule to direct trafficking of activated EGFR to focal adhesions. These studies provide a novel paradigm for the regulation of crosstalk among integrins, growth-factor receptors and uPAR.


Subject(s)
Caveolin 1/metabolism , Fibronectins/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Signal Transduction , Animals , Caveolin 1/genetics , Cells, Cultured , ErbB Receptors/genetics , ErbB Receptors/metabolism , Fibroblasts/metabolism , Fibronectins/genetics , Humans , Integrins/genetics , Integrins/metabolism , Mice , Phosphorylation , Receptors, Urokinase Plasminogen Activator/genetics
15.
J Biol Chem ; 281(14): 9450-9, 2006 Apr 07.
Article in English | MEDLINE | ID: mdl-16461772

ABSTRACT

Previous studies have indicated that the urokinase-type plasminogen activator receptor (uPAR) can functionally interact with integrins thereby modulating integrin activity. We have previously demonstrated that treatment of fibroblasts with the uPAR ligand, P25, results in an increase in the activation of the beta1 integrin and a 35-fold increase in fibronectin matrix assembly (Monaghan, E., Gueorguiev, V., Wilkins-Port, C., and McKeown-Longo, P. J. (2004) J. Biol. Chem. 279, 1400-1407). Experiments were conducted to address the mechanism of uPAR regulation of matrix assembly. Treatment of fibroblasts with P25 led to an increase in the activation of the epidermal growth factor receptor (EGFR) and a colocalization of activated EGFR with beta1 integrins in cell matrix contacts. The effects of P25 on matrix assembly and beta1 integrin activation were inhibited by pretreatment with EGFR or Src kinase inhibitors, suggesting a role for both Src and EGFR in integrin activation by uPAR. Phosphorylation of EGFR in response to P25 occurred on Tyr-845, an Src-dependent phosphorylation site and was inhibited by PP2, the Src kinase inhibitor, consistent with Src kinase lying upstream of EGFR and integrin activation. Cells null for Src kinases also showed a loss of P25-induced matrix assembly, integrin activation, and EGFR phosphorylation. These P25-induced effects were restored following Src re-expression. The effects of P25 were specific for uPAR as enhanced matrix assembly by P25 was not seen in uPAR-/- cells, but was restored upon uPAR re-expression. These data provide evidence for a novel pathway of fibronectin matrix assembly through the uPAR-dependent sequential activation of Src kinase, EGFR, and beta1 integrin.


Subject(s)
ErbB Receptors/biosynthesis , Fibronectins/biosynthesis , Mannose-Binding Lectins/physiology , Membrane Glycoproteins/physiology , Receptors, Cell Surface/physiology , Cell Culture Techniques , Extracellular Matrix , Fibroblasts , Humans , Integrin beta1/physiology , Ligands , Phosphorylation , Transcriptional Activation , Transfection , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL