Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Int J Mol Sci ; 24(11)2023 Jun 03.
Article in English | MEDLINE | ID: mdl-37298667

ABSTRACT

CXCL12, belonging to the CXC chemokine family, is a weak agonist of platelet aggregation. We previously reported that the combination of CXCL12 and collagen at low doses synergistically activates platelets via not CXCR7 but CXCR4, a specific receptor for CXCL12 on the plasma membrane. Recently, we reported that not Rho/Rho kinase, but Rac is involved in the platelet aggregation induced by this combination. Ristocetin is an activator of the von Willebrand factor that interacts with glycoprotein (GP) Ib/IX/V, which generates thromboxane A2 via phospholipase A2 activation, resulting in the release of the soluble CD40 ligand (sCD40L) from human platelets. In the present study, we investigated the effects of a combination of ristocetin and CXCL12 at low doses on human platelet activation and its underlying mechanisms. Simultaneous stimulation with ristocetin and CXCL12 at subthreshold doses synergistically induce platelet aggregation. A monoclonal antibody against not CXCR7 but CXCR4 suppressed platelet aggregation induced by the combination of ristocetin and CXCL12 at low doses. This combination induces a transient increase in the levels of both GTP-binding Rho and Rac, followed by an increase in phosphorylated cofilin. The ristocetin and CXCL12-induced platelet aggregation as well as the sCD40L release were remarkably enhanced by Y27362, an inhibitor of Rho-kinase, but reduced by NSC23766, an inhibitor of the Rac-guanine nucleotide exchange factor interaction. These results strongly suggest that the combination of ristocetin and CXCL12 at low doses synergistically induces human platelet activation via Rac and that this activation is negatively regulated by the simultaneous activation of Rho/Rho-kinase.


Subject(s)
Ristocetin , rho-Associated Kinases , Humans , Blood Platelets/metabolism , CD40 Ligand/metabolism , Chemokine CXCL12/pharmacology , Chemokine CXCL12/metabolism , Phosphorylation , Platelet Activation , Platelet Aggregation , Platelet Glycoprotein GPIb-IX Complex/metabolism , rho-Associated Kinases/metabolism , Ristocetin/metabolism , Ristocetin/pharmacology , von Willebrand Factor/metabolism , rac GTP-Binding Proteins/drug effects , rac GTP-Binding Proteins/metabolism
2.
Eur Respir J ; 41(1): 165-76, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22790920

ABSTRACT

The protective effects of prostacyclin and its stable analogue iloprost are mediated by elevation of intracellular cyclic AMP (cAMP) leading to enhancement of the peripheral actin cytoskeleton and cell-cell adhesive structures. This study tested the hypothesis that iloprost may exhibit protective effects against lung injury and endothelial barrier dysfunction induced by bacterial wall lipopolysaccharide (LPS). Endothelial barrier dysfunction was assessed by measurements of transendothelial permeability, morphologically and by analysis of LPS-activated inflammatory signalling. In vivo, C57BL/6J mice were challenged with LPS with or without iloprost or 8-bromoadenosine-3',5'-cyclic monophosphate (Br-cAMP) treatment. Lung injury was monitored by measurements of bronchoalveolar lavage protein content, cell count and Evans blue extravasation. Iloprost and Br-cAMP attenuated the disruption of the endothelial monolayer, and suppressed the activation of p38 mitogen-activated protein kinase (MAPK), the nuclear factor (NF)-κB pathway, Rho signalling, intercellular adhesion molecular (ICAM)-1 expression and neutrophil migration after LPS challenge. In vivo, iloprost was effective against LPS-induced protein and neutrophil accumulation in bronchoalveolar lavage fluid, and reduced myeloperoxidase activation, ICAM-1 expression and Evans blue extravasation in the lungs. Inhibition of Rac activity abolished the barrier-protective and anti-inflammatory effects of iloprost and Br-cAMP. Iloprost-induced elevation of intracellular cAMP triggers Rac signalling, which attenuates LPS-induced NF-κB and p38 MAPK inflammatory pathways and the Rho-dependent mechanism of endothelial permeability.


Subject(s)
Iloprost/therapeutic use , Lung Injury/drug therapy , Lung/drug effects , Lung/physiopathology , Animals , Cells, Cultured , Endothelium/drug effects , Endothelium/physiology , Lipopolysaccharides/administration & dosage , Lung Injury/chemically induced , Mice , Mice, Inbred C57BL , Neuropeptides/drug effects , Neuropeptides/physiology , rac GTP-Binding Proteins/drug effects , rac GTP-Binding Proteins/physiology , rac1 GTP-Binding Protein
3.
J Surg Res ; 183(2): 798-807, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23545410

ABSTRACT

Excessive neutrophil recruitment is a major feature in septic lung damage although the signaling mechanisms behind pulmonary infiltration of neutrophils in sepsis remain elusive. In the present study, we hypothesized that Rac1 might play an important role in pulmonary neutrophil accumulation and tissue injury in abdominal sepsis. Male C57BL/6 mice were treated with Rac1 inhibitor NSC23766 (5 mg/kg) before cecal ligation and puncture (CLP). Bronchoalveolar lavage fluid and lung tissue were collected for the quantification of neutrophil recruitment and edema and CXC chemokine formation. Blood was collected for the determination of Mac-1 on neutrophils and proinflammatory compounds in plasma. Gene expression of CXC chemokines and tumor necrosis factor alpha was determined by quantitative reverse transcription-polymerase chain reaction in alveolar macrophages. Rac1 activity was increased in lungs from septic animals, and NSC23766 significantly decreased pulmonary activity of Rac1 induced by CLP. Administration of NSC23766 markedly reduced CLP-triggered neutrophil infiltration, edema formation, and tissue damage in the lung. Inhibition of Rac1 decreased CLP-induced neutrophil expression of Mac-1 and pulmonary formation of CXC chemokines. Moreover, NSC23766 abolished the sepsis-evoked elevation of messenger RNA levels of CXC chemokines and tumor necrosis factor alpha in alveolar macrophages. Rac1 inhibition decreased the CLP-induced increase in plasma levels of high mobility group protein B1 and interleukin 6, indicating a role of Rac1 in systemic inflammation. In conclusion, our results demonstrate that Rac1 signaling plays a key role in regulating pulmonary infiltration of neutrophils and tissue injury via regulation of chemokine production in the lung and Mac-1 expression on neutrophils in abdominal sepsis. Thus, targeting Rac1 activity might be a useful strategy to protect the lung in abdominal sepsis.


Subject(s)
Chemokines, CXC/metabolism , Macrophage-1 Antigen/metabolism , Neuropeptides/physiology , Pneumonia/microbiology , Pneumonia/physiopathology , Sepsis/complications , Signal Transduction/physiology , rac GTP-Binding Proteins/physiology , Aminoquinolines/pharmacology , Animals , Cecum/injuries , Cell Movement/drug effects , Cell Movement/physiology , Disease Models, Animal , HMGB1 Protein/metabolism , In Vitro Techniques , Ligation/adverse effects , Male , Mice , Mice, Inbred C57BL , Neuropeptides/antagonists & inhibitors , Neuropeptides/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/pathology , Pneumonia/pathology , Punctures/adverse effects , Pyrimidines/pharmacology , Sepsis/etiology , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/drug effects , rac1 GTP-Binding Protein
4.
Nat Cell Biol ; 6(3): 268-74, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15039778

ABSTRACT

The non-receptor tyrosine kinase Abl participates in receptor tyrosine kinase (RTK)-induced actin cytoskeleton remodelling, a signalling pathway in which the function of Rac is pivotal. More importantly, the activity of Rac is indispensable for the leukaemogenic ability of the BCR-Abl oncoprotein. Thus, Rac might function downstream of Abl and be activated by it. Here, we elucidate the molecular mechanisms through which Abl signals to Rac in RTK-activated pathways. We show that Sos-1, a dual guanine nucleotide-exchange factor (GEF), is phosphorylated on tyrosine, after activation of RTKs, in an Abl-dependent manner. Sos-1 and Abl interact in vivo, and Abl-induced tyrosine phosphorylation of Sos-1 is sufficient to elicit its Rac-GEF activity in vitro. Genetic or pharmacological interference with Abl (and the related kinase Arg) resulted in a marked decrease in Rac activation induced by physiological doses of growth factors. Thus, our data identify the molecular connections of a pathway RTKs-Abl-Sos-1-Rac that is involved in signal transduction and actin remodelling.


Subject(s)
Actin Cytoskeleton/enzymology , Proto-Oncogene Proteins c-abl/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , SOS1 Protein/metabolism , rac GTP-Binding Proteins/metabolism , Animals , COS Cells , Growth Substances/pharmacology , Humans , Phosphorylation/drug effects , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/genetics , Receptor Protein-Tyrosine Kinases/drug effects , SOS1 Protein/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Tyrosine/metabolism , rac GTP-Binding Proteins/drug effects
5.
Nat Med ; 7(9): 1041-7, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11533708

ABSTRACT

Cyclooxygenase-2 (COX-2), a key enzyme in arachidonic acid metabolism, is overexpressed in many cancers. Inhibition of COX-2 by nonsteroidal anti-inflammatory drugs (NSAIDs) reduces the risk of cancer development in humans and suppresses tumor growth in animal models. The anti-cancer effect of NSAIDs seems to involve suppression of tumor angiogenesis, but the underlying mechanism is not completely understood. Integrin alpha V beta 3 is an adhesion receptor critically involved in mediating tumor angiogenesis. Here we show that inhibition of endothelial-cell COX-2 by NSAIDs suppresses alpha V beta 3-dependent activation of the small GTPases Cdc42 and Rac, resulting in inhibition of endothelial-cell spreading and migration in vitro and suppression of fibroblast growth factor-2-induced angiogenesis in vivo. These results establish a novel functional link between COX-2, integrin alpha V beta 3 and Cdc42-/Rac-dependent endothelial-cell migration. Moreover, they provide a rationale to the understanding of the anti-angiogenic activity of NSAIDs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Endothelium, Vascular/cytology , Receptors, Vitronectin/metabolism , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism , Animals , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Fibroblast Growth Factor 2/pharmacology , Humans , Isoenzymes/antagonists & inhibitors , Membrane Proteins , Mice , Mice, Nude , Neovascularization, Physiologic/drug effects , Nitrobenzenes/pharmacology , Prostaglandin-Endoperoxide Synthases , Receptors, Vitronectin/antagonists & inhibitors , Sulfonamides/pharmacology , Thromboxane A2/pharmacology , cdc42 GTP-Binding Protein/drug effects , rac GTP-Binding Proteins/drug effects
6.
J Pharmacol Exp Ther ; 333(2): 393-403, 2010 May.
Article in English | MEDLINE | ID: mdl-20110378

ABSTRACT

Signaling through G protein-coupled receptors (GPCRs) promotes breast cancer metastasis. G proteins convey GPCR signals by dissociating into Galpha and Gbetagamma subunits. The aim of the present study was to determine whether blockade of Gbetagamma signaling suppresses breast cancer cell migration and invasion, which are critical components of metastasis. Conditioned media (CM) of NIH-3T3 fibroblasts are widely used as chemoattractants in in vitro cancer metastasis studies. Expression of a Gbetagamma scavenger peptide attenuated NIH-3T3 CM-induced migration and invasion of both metastatic breast cancer MDA-MB-231 and MDA-MB-436 cells by 40 to 50% without effects on cell viability. Migration and invasion of cells in response to NIH-3T3 CM were also blocked by 8-(4,5,6-trihydroxy-3-oxo-3H-xanthen-9-yl)-1-naph-thalene-carboxylic acid) (M119K), a Gbetagamma inhibitor, with maximum inhibition exceeding 80% and half-maximal inhibitory concentration (IC50) values of 1 to 2 microM. M119K also attenuated Rac-dependent formation of lamellipodia, a key structure required for metastasis. Constitutively active Rac1 rescued Gbetagamma blockade-mediated inhibition of breast cancer cell migration, whereas dominant negative Rac1 inhibited cell migration similar to Gbetagamma blockade. Furthermore, M119K suppressed Gi protein-coupled CXC chemokine receptor 4 (CXCR4)-dependent MDA-MB-231 cell migration by 80% with an IC50 value of 1 microM, whereas tyrosine kinase receptor-dependent cell migration was significantly less inhibited. However, CXCR4-dependent inhibition of adenylyl cyclase, a Gialpha-mediated response in MDA-MB-231 cells, was not blocked by M119K but was blocked by pertussis toxin, which selectively inactivates Gialpha. This report is the first to directly demonstrate the role of Gbetagamma in cancer cell migration and invasion and suggests that targeting Gbetagamma signaling pathways may provide a novel strategy for suppressing breast cancer metastasis.


Subject(s)
Breast Neoplasms/physiopathology , Cell Movement/physiology , GTP-Binding Protein beta Subunits/pharmacology , GTP-Binding Protein gamma Subunits/pharmacology , Neoplasm Invasiveness/physiopathology , Adenylyl Cyclases/drug effects , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/physiology , Cyclohexanes/pharmacology , Female , Humans , Microscopy, Fluorescence , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/physiopathology , Peptide Fragments/physiology , Pseudopodia/drug effects , Receptors, CXCR4/physiology , Recombinant Proteins , Signal Transduction/drug effects , Signal Transduction/physiology , Xanthenes/pharmacology , rac GTP-Binding Proteins/drug effects , rac GTP-Binding Proteins/physiology
7.
J Cell Biol ; 171(2): 361-71, 2005 Oct 24.
Article in English | MEDLINE | ID: mdl-16247032

ABSTRACT

Introduction of activated p21-activated kinase (PAK) is sufficient to release primary endothelial cells from contact inhibition of growth. Confluent cells display deficient activation of PAK and translocation of Rac to the plasma membrane at matrix adhesions. Targeting Rac to the plasma membrane rescues these cells from contact inhibition. PAK's ability to release human umbilical vein endothelial cells from contact inhibition is blocked by an unphosphorylatable form of its target Merlin, suggesting that PAK promotes mitogenesis by phosphorylating, and thus inactivating, Merlin. Merlin mutants, which are presumed to exert a dominant-negative effect, enable recruitment of Rac to matrix adhesions and promote mitogenesis in confluent cells. Small interference RNA-mediated knockdown of Merlin exerts the same effects. Dominant-negative Rac blocks PAK-mediated release from contact inhibition, implying that PAK functions upstream of Rac in this signaling pathway. These results provide a framework for understanding the tumor suppressor function of Merlin and indicate that Merlin mediates contact inhibition of growth by suppressing recruitment of Rac to matrix adhesions.


Subject(s)
Cell Membrane/metabolism , Contact Inhibition/physiology , Neurofibromin 2/physiology , rac GTP-Binding Proteins/metabolism , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Membrane/drug effects , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Humans , Neurofibromin 2/antagonists & inhibitors , Phosphorylation , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , p21-Activated Kinases , rac GTP-Binding Proteins/drug effects
8.
J Cell Biol ; 161(2): 381-91, 2003 Apr 28.
Article in English | MEDLINE | ID: mdl-12707308

ABSTRACT

Rho-GTPase has been implicated in axon outgrowth. However, not all of the critical steps controlled by Rho have been well characterized. Using cultured cerebellar granule neurons, we show here that stromal cell-derived factor (SDF)-1alpha, a neural chemokine, is a physiological ligand that can turn on two distinct Rho-dependent pathways with opposite consequences. A low concentration of the ligand stimulated a Rho-dependent pathway that mediated facilitation of axon elongation. In contrast, Rho/ROCK activation achieved by a higher concentration of SDF-1alpha caused repression of axon formation and induced no more increase in axon length. However, even at this higher concentration a Rho-dependent axon elongating activity could be recovered upon removal of ROCK activity using Y-27632. SDF-1alpha-induced axon elongating activity under ROCK inhibition was replicated by the dominant-active form of the mammalian homologue of the Drosophila gene Diaphanous (mDia)1 and counteracted by its dominant-negative form. Furthermore, RNAi knockdown of mDia1 abolished SDF-1alpha-induced axon elongation. Together, our results support a critical role for an SDF-1alpha/Rho/mDia1 pathway in mediating axon elongation.


Subject(s)
Carrier Proteins/metabolism , Cell Differentiation/genetics , Cerebellar Cortex/growth & development , Chemokines, CXC/metabolism , Growth Cones/metabolism , Protein Serine-Threonine Kinases/metabolism , rho GTP-Binding Proteins/metabolism , Actins/metabolism , Animals , Carrier Proteins/genetics , Cell Differentiation/drug effects , Cells, Cultured , Cerebellar Cortex/cytology , Cerebellar Cortex/metabolism , Chemokine CXCL12 , Chemokines, CXC/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Formins , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Growth Cones/drug effects , Growth Cones/ultrastructure , Intracellular Signaling Peptides and Proteins , Mice , Mutation , Protein Serine-Threonine Kinases/drug effects , RNA Interference/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , rac GTP-Binding Proteins/drug effects , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/drug effects , rho-Associated Kinases
9.
J Cell Biol ; 161(3): 547-56, 2003 May 12.
Article in English | MEDLINE | ID: mdl-12743104

ABSTRACT

In NIH3T3 cells, 0.001 nM of the synthetic androgen R1881 induces and stimulates association of androgen receptor (AR) with Src and phosphatidylinositol 3-kinase (Pl3-kinase), respectively, thereby triggering S-phase entry. 10 nM R1881 stimulates Rac activity and membrane ruffling in the absence of the receptor-Src-PI3-kinase complex assembly. The antiandrogen Casodex and specific inhibitors of Src and PI3-kinase prevent both hormonal effects, DNA synthesis and cytoskeletal changes. Neither low nor high R1881 concentration allows receptor nuclear translocation and receptor-dependent transcriptional activity in fibroblasts, although they harbor the classical murine AR. The very low amount of AR in NIH3T3 cells (7% of that present in LNCaP cells) activates the signaling pathways, but apparently is not sufficient to stimulate gene transcription. This view is supported by the appearance of receptor nuclear translocation as well as receptor-mediated transcriptional activity after overexpression of AR in fibroblasts. In addition, AR-negative Cos cells transiently transfected with a very low amount of hAR cDNA respond to low and high R1881 concentrations with signaling activation. Interestingly, they do not show significant transcriptional activation under the same experimental conditions. Fibroblasts are the first example of cells that respond to steroid hormones with activation of signaling pathways in the absence of endogenous receptor transcriptional activity. The data reported also show that hormone concentration can be crucial in determining the type of cell responsiveness.


Subject(s)
Androgens/metabolism , Cytoskeleton/metabolism , DNA/biosynthesis , Fibroblasts/metabolism , Receptors, Androgen/metabolism , 3T3 Cells , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Androgens/pharmacology , Animals , Antibodies/pharmacology , COS Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cytoskeleton/drug effects , Cytoskeleton/ultrastructure , DNA/drug effects , Dose-Response Relationship, Drug , Female , Fetus , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Male , Mice , Receptors, Androgen/drug effects , Receptors, Androgen/genetics , S Phase/drug effects , S Phase/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/metabolism , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Tumor Cells, Cultured , rac GTP-Binding Proteins/drug effects , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism
10.
Life Sci ; 221: 29-34, 2019 Mar 15.
Article in English | MEDLINE | ID: mdl-30721707

ABSTRACT

Vascular inflammatory responses play an important role in several cardiovascular diseases. Of the many pro-inflammatory vasoactive factors implicated in this process, is aldosterone, an important mediator of vascular oxidative stress. Statins, such as atorvastatin, are cholesterol-lowering drugs that have pleiotropic actions, including anti-oxidant properties independently of their cholesterol-lowering effect. This study investigated whether atorvastatin prevents aldosterone-induced VSMC inflammation by reducing reactive oxygen species (ROS) production. Vascular smooth muscle cells (VSMC) from WKY rats were treated with 1 µM atorvastatin for 60 min or for 72 h prior to aldosterone (10-7 mol/L) stimulation. Atorvastatin inhibited Rac1/2 and p47phox translocation from the cytosol to the membrane, as well as reduced aldosterone-induced ROS production. Atorvastatin also attenuated aldosterone-induced vascular inflammation and macrophage adhesion to VSMC. Similarly EHT1864, a Rac1/2 inhibitor, and tiron, ROS scavenger, reduced macrophage adhesion. Through its inhibitory effects on Rac1/2 activation and ROS production, atorvastatin reduces vascular ROS generation and inhibits VSMC inflammation. Our data suggest that in conditions associated with aldosterone-induced vascular damage, statins may have vasoprotective effects by inhibiting oxidative stress and inflammation.


Subject(s)
Aldosterone/metabolism , Atorvastatin/metabolism , Muscle, Smooth, Vascular/drug effects , Aldosterone/pharmacology , Angiotensin II , Animals , Antioxidants , Atorvastatin/pharmacology , Cells, Cultured , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Male , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle , NADPH Oxidases/drug effects , Oxidation-Reduction , Oxidative Stress/drug effects , Rats , Rats, Inbred WKY , Reactive Oxygen Species/metabolism , Signal Transduction , rac GTP-Binding Proteins/drug effects , rac1 GTP-Binding Protein/drug effects , RAC2 GTP-Binding Protein
11.
J Cell Physiol ; 217(1): 162-71, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18452153

ABSTRACT

The goal of this study was to determine the morphological and sub-cellular mechanical effects of Rac activation on fibroblasts within 3-D collagen matrices. Corneal fibroblasts were plated at low density inside 100 microm thick fibrillar collagen matrices and cultured for 1-2 days in serum-free media. Time-lapse imaging was then performed using Nomarski DIC. After an acclimation period, perfusion was switched to media containing PDGF. In some experiments, Y-27632 or blebbistatin were used to inhibit Rho-kinase (ROCK) or myosin II, respectively. PDGF activated Rac and induced cell spreading, which resulted in an increase in cell length, cell area, and the number of pseudopodial processes. Tractional forces were generated by extending pseudopodia, as indicated by centripetal displacement and realignment of collagen fibrils. Interestingly, the pattern of pseudopodial extension and local collagen fibril realignment was highly dependent upon the initial orientation of fibrils at the leading edge. Following ROCK or myosin II inhibition, significant ECM relaxation was observed, but small displacements of collagen fibrils continued to be detected at the tips of pseudopodia. Taken together, the data suggests that during Rac-induced cell spreading within 3-D matrices, there is a shift in the distribution of forces from the center to the periphery of corneal fibroblasts. ROCK mediates the generation of large myosin II-based tractional forces during cell spreading within 3-D collagen matrices, however residual forces can be generated at the tips of extending pseudopodia that are both ROCK and myosin II-independent.


Subject(s)
Cell Movement/physiology , Extracellular Matrix , Fibroblasts/cytology , rac GTP-Binding Proteins/metabolism , Biomechanical Phenomena , Cell Line , Cell Movement/drug effects , Cell Shape/physiology , Collagen , Cornea/cytology , Enzyme Inhibitors/pharmacology , Extracellular Matrix/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Myosin Type II/metabolism , Platelet-Derived Growth Factor/pharmacology , Pseudopodia/drug effects , Pseudopodia/metabolism , rac GTP-Binding Proteins/drug effects
12.
FASEB J ; 21(8): 1665-74, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17317725

ABSTRACT

Macrophage cyclooxygenase-2 (COX-2) plays an important role in prostaglandin E2 and thromboxane A2 production. Statins are inhibitors of HMG CoA (3-Hydroxy-3-methylglutaryl coenzyme A) reductases and cholesterol synthesis, which block the expression of several inflammatory proteins independent of their capacity to lower endogenous cholesterol. In the present study, we investigated the effect of simvastatin and mevastatin on COX-2 induction in human monocytic cell line U937 and analyzed the underlying mechanisms. Pretreatment of U937 cells with simvastatin or mevastatin for 24 h resulted in a significant reduction in the lipopolysaccharide (LPS)-dependent induction of prostaglandin E2, thromboxane A2 synthesis, and COX-2 expression. Mevalonate, the direct metabolite of HMG CoA reductase, and farnesyl pyrophosphate and geranylgeranyl-pyrophosphate, intermediates of the mevalonate pathway, significantly reversed the inhibitory effect of statins on COX-2. An inhibitor of geranylgeranyl transferases, GGTI-286 mimicked the effect of statins on COX-2 expression. Cytonecrotic factor-1 increased LPS-dependent expression of COX-2. Treatment of cells with NSC 23766, an inhibitor of Rac, which we demonstrated to block Rac 2 activation, resulted in an inhibition of the LPS-dependent expression of COX-2. Whereas no effect was obtained with RhoA/C blocker, C3 exoenzyme. Gel retardation experiments and NFkappaB-p65 transcription factor assay showed that simvastatin and NSC 23766 decrease significantly NF-kappaB complex formation. In macrophages, the antiinflammatory effects of statins are mediated in part through the inhibition of COX-2 and prostanoids. Rac GTPase protein is identified as one of the targets of statins in this regulation.


Subject(s)
Cyclooxygenase 2/genetics , Gene Expression Regulation/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Membrane Proteins/genetics , Monocytes/metabolism , Humans , Lipopolysaccharides/pharmacology , Lovastatin/analogs & derivatives , Lovastatin/pharmacology , Mevalonic Acid/metabolism , NF-kappa B/metabolism , Simvastatin/pharmacology , U937 Cells , rac GTP-Binding Proteins/drug effects , RAC2 GTP-Binding Protein
14.
Brain Res ; 1167: 92-100, 2007 Sep 05.
Article in English | MEDLINE | ID: mdl-17692294

ABSTRACT

Brain edema is the most life-threatening complication that occurs as a result of a number of insults to the brain. However, its therapeutic options are insufficiently effective. We have recently found that administration of pigment epithelium-derived factor (PEDF) inhibits retinal hyperpermeability in rats by counteracting biological effects of vascular endothelial growth factor (VEGF). In this study, we investigated whether PEDF could inhibit cold injury-induced brain edema in mice. Cold injury was induced by applying a pre-cooled metal probe on the parietal skull. VEGF and its receptor Flk-1 gene and/or protein expressions were up-regulated in the cold-injured brain. Cold injury induced brain edema, which was reduced by intraperitoneal injection of VEGF antibodies (Abs) or apocynin, an inhibitor of NADPH oxidase. PEDF mRNA and protein levels were up-regulated in response to cold injury. PEDF dose-dependently inhibited the brain edema, whose effect was neutralized by simultaneous treatments with anti-PEDF Abs. Although VEGF and Flk-1 gene and/or protein expressions were not suppressed by PEDF, PEDF or anti-VEGF Abs inhibited the cold injury-induced NADPH oxidase activity in the brain. Further, PEDF treatment inhibited activation of Rac-1, an essential component of NADPH oxidase in the cold-injured brain, while it did not affect mRNA levels of gp91phox, p22phox, or Rac-1. These results demonstrate that PEDF could inhibit the cold injury-induced brain edema by blocking the VEGF signaling to hyperpermeability through the suppression of NADPH oxidase via inhibition of Rac-1 activation. Our present study suggests that PEDF may be a novel therapeutic agent for the treatment of brain edema.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Edema/drug therapy , Brain Injuries/drug therapy , Cold Temperature/adverse effects , Eye Proteins/pharmacology , Nerve Growth Factors/pharmacology , Serpins/pharmacology , Acetophenones/pharmacology , Animals , Antibodies/pharmacology , Blood-Brain Barrier/physiopathology , Brain Edema/etiology , Brain Edema/physiopathology , Brain Injuries/complications , Brain Injuries/physiopathology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Eye Proteins/genetics , Eye Proteins/metabolism , Hypothermia, Induced , Male , Mice , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Serpins/genetics , Serpins/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Up-Regulation/drug effects , Up-Regulation/physiology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , rac GTP-Binding Proteins/drug effects , rac GTP-Binding Proteins/metabolism
15.
Mol Biol Cell ; 11(5): 1709-25, 2000 May.
Article in English | MEDLINE | ID: mdl-10793146

ABSTRACT

Hepatocyte growth factor (HGF), the ligand for the Met receptor tyrosine kinase, is a potent modulator of epithelial-mesenchymal transition and dispersal of epithelial cells, processes that play crucial roles in tumor development, invasion, and metastasis. Little is known about the Met-dependent proximal signals that regulate these events. We show that HGF stimulation of epithelial cells leads to activation of the Rho GTPases, Cdc42 and Rac, concomitant with the formation of filopodia and lamellipodia. Notably, HGF-dependent activation of Rac but not Cdc42 is dependent on phosphatidylinositol 3-kinase. Moreover, HGF-induced lamellipodia formation and cell spreading require phosphatidylinositol 3-kinase and are inhibited by dominant negative Cdc42 or Rac. HGF induces activation of the Cdc42/Rac-regulated p21-activated kinase (PAK) and c-Jun N-terminal kinase, and translocation of Rac, PAK, and Rho-dependent Rho-kinase to membrane ruffles. Use of dominant negative and activated mutants reveals an essential role for PAK but not Rho-kinase in HGF-induced epithelial cell spreading, whereas Rho-kinase activity is required for the formation of focal adhesions and stress fibers in response to HGF. We conclude that PAK and Rho-kinase play opposing roles in epithelial-mesenchymal transition induced by HGF, and provide new insight regarding the role of Cdc42 in these events.


Subject(s)
Epithelial Cells/cytology , Hepatocyte Growth Factor/pharmacology , Protein Serine-Threonine Kinases/metabolism , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Actins/drug effects , Actins/metabolism , Animals , Cell Line , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Dogs , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , JNK Mitogen-Activated Protein Kinases , Kidney/cytology , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/drug effects , Signal Transduction , cdc42 GTP-Binding Protein/drug effects , p21-Activated Kinases , rac GTP-Binding Proteins/drug effects
16.
Circ Res ; 90(3): 325-32, 2002 Feb 22.
Article in English | MEDLINE | ID: mdl-11861422

ABSTRACT

Previous studies demonstrated that sphingosine-1-phosphate (S1P) induced migration of human umbilical vein endothelial cells (HUVECs) whereas it inhibited that of vascular smooth muscle cells (SMCs). This study explored the molecular mechanisms underlying the contrasting S1P actions on vascular cell motility. In rat and human aortic SMCs, the chemoattractant platelet-derived growth factor B-chain (PDGF) induced rapid 5- to 6-fold increases in the cellular amount of GTP-bound, active form of Rac. S1P did not affect PDGF-stimulated tyrosine phosphorylation of PDGF-beta receptor, but strongly inhibited PDGF-induced Rac activation, with a dose-response relationship similar to that for inhibition of PDGF-elicited chemotaxis. Dihydrosphingosine-1-phosphate, which is a weaker agonist for the S1P receptors, but not an inactive ligand sphingosine, also inhibited PDGF-stimulated chemotaxis and Rac activation although to lesser extents compared with S1P, suggesting that negative regulation by S1P of both chemotaxis and Rac was a receptor-mediated process. In contrast, S1P by itself stimulated Rac activity in HUVECs. Among the five S1P receptor isoforms, SMCs prominently expressed Edg-5 mRNA, whereas HUVECs expressed abundant Edg-1 mRNA but lacked detectable expression of Edg-5 mRNA. Adenovirus-mediated expression of a dominant-negative form of either Rac or Cdc42, but not RhoA, markedly attenuated chemotaxis of SMCs and HUVECs toward PDGF and S1P, respectively. Overexpression of Edg-1 in SMCs and Edg-5 in HUVECs reduced S1P-induced inhibition and stimulation, respectively, of Rac activity and migration. These results together indicate that Edg isoform-specific, negative or positive regulation of cellular Rac activity is critically involved in S1P-mediated bimodal regulation of cell motility in SMCs and HUVECs.


Subject(s)
Cell Movement/physiology , Lysophospholipids/metabolism , Muscle, Smooth, Vascular/metabolism , Receptors, Cell Surface , Receptors, G-Protein-Coupled , Sphingosine/analogs & derivatives , Sphingosine/metabolism , rac GTP-Binding Proteins/metabolism , Animals , Blood Platelets/metabolism , Cell Movement/drug effects , Cell Surface Extensions/drug effects , Cells, Cultured , Chemotaxis/drug effects , Chemotaxis/physiology , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Enzyme Activation/drug effects , Humans , Immediate-Early Proteins/biosynthesis , Immediate-Early Proteins/genetics , Immediate-Early Proteins/pharmacology , Lysophospholipids/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Nuclear Proteins/pharmacology , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/pharmacology , Rats , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptors, Lysophosphatidic Acid , Receptors, Lysophospholipid , Sphingosine/pharmacology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription Factors/pharmacology , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/drug effects , rac GTP-Binding Proteins/genetics , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
17.
J Leukoc Biol ; 77(2): 257-66, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15561756

ABSTRACT

Patients with myelodysplasia suffer from recurrent bacterial infections as a result of differentiation defects of the myeloid lineage and a disturbed functioning of neutrophilic granulocytes. Important physiological activators of neutrophils are the cytokines interleukin-8/CXC chemokine ligand 8 (IL-8/CXCL8), which activates CXC chemokine receptor 1 and 2 (CXCR1 and CXCR2), and growth-related oncogene (GROalpha)/CXCL1, which stimulates only CXCR2. In this study, we show that migration toward IL-8/GROalpha gradients is decreased in myelodysplastic syndrome (MDS) neutrophils compared with healthy donors. We investigated the signal transduction pathways involved in IL-8/GROalpha-induced migration and showed that specific inhibitors for extracellular signal-regulated kinase (ERK)1/2 and phosphatidylinositol-3 kinase (PI-3K) abrogated neutrophil migration toward IL-8/GROalpha. In accordance with these results, we subsequently showed that IL-8/GROalpha-stimulated activation of ERK1/2 was substantially diminished in MDS neutrophils. Activation of the PI-3K downstream target protein kinase B/Akt was disturbed in MDS neutrophils when cells were activated with IL-8 but normal upon GROalpha stimulation. IL-8 stimulation resulted in higher migratory behavior and ERK1/2 activation than GROalpha stimulation, suggesting a greater importance of CXCR1. We then investigated IL-8-induced activation of the small GTPase Rac implicated in ERK1/2-dependent migration and found that it was less efficient in neutrophils from MDS patients compared with healthy donors. In contrast, IL-8 triggered a normal activation of the GTPases Ras and Ral, indicating that the observed defects were not a result of a general disturbance in CXCR1/2 signaling. In conclusion, our results demonstrate a disturbed CXCR1- and CXCR2-induced neutrophil chemotaxis in MDS patients, which might be the consequence of decreased Rac-ERK1/2 and PI-3K activation within these cells.


Subject(s)
Chemokines, CXC/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Interleukin-8/pharmacology , Myelodysplastic Syndromes/etiology , Neutrophils/cytology , Cell Movement/drug effects , Chemokine CXCL1 , Extracellular Signal-Regulated MAP Kinases/drug effects , Humans , Neutrophils/drug effects , Phosphorylation/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , rac GTP-Binding Proteins/drug effects , rac GTP-Binding Proteins/metabolism , ras Proteins/drug effects , ras Proteins/metabolism
18.
J Neurosci ; 22(18): 7968-81, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12223550

ABSTRACT

The role of small GTPases of the Rho family in synaptic functions has been addressed by analyzing the effects of lethal toxin (LT) from Clostridium sordellii strain IP82 (LT82) on neurotransmitter release at evoked identified synapses in the buccal ganglion of Aplysia. LT82 is a large monoglucosyltranferase that uses UDP-glucose as cofactor and glucosylates Rac (a small GTPase related to Rho), and Ras, Ral, and Rap (three GTPases of the Ras family). Intraneuronal application of LT (50 nm) rapidly inhibits evoked acetylcholine (ACh) release as monitored electrophysiologically. Injection of the catalytic domain of the toxin similarly blocked ACh release, but not when key amino acids needed for glucosylation were mutated. Intraneuronal application of competitive nucleotide sugars that differentially prevent glucosylation of Rac- and Ras-related GTPases, and the use of a toxin variant that affects a different spectrum of small GTPases, established that glucosylation of Rac is responsible for the reduction in ACh release. To determine the quantal release parameters affected by Rac glucosylation, we developed a nonstationary analysis of the fluctuations in postsynaptic response amplitudes that was performed before and after the toxin had acted or during toxin action. The results indicate that neither the quantal size nor the average probability for release were affected by lethal toxin action. ACh release blockage by LT82 was only caused by a reduction in the number of functional release sites. This reveals that after docking of synaptic vesicles, vesicular Rac stimulates a membrane effector (or effectors) essential for the fusion competence of the exocytotic sites.


Subject(s)
Binding Sites/physiology , Cell Membrane/physiology , Exocytosis/physiology , Membrane Fusion/physiology , rac GTP-Binding Proteins/metabolism , Acetylcholine/metabolism , Animals , Bacterial Toxins/pharmacology , Binding Sites/drug effects , Calcium/metabolism , Exocytosis/drug effects , Glycosylation/drug effects , In Vitro Techniques , Magnesium/metabolism , Microinjections , Neurons/drug effects , Neurons/metabolism , Patch-Clamp Techniques , Signal Transduction/drug effects , Signal Transduction/physiology , Synapses/drug effects , Synapses/metabolism , rac GTP-Binding Proteins/drug effects
19.
Mol Endocrinol ; 18(2): 359-72, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14615606

ABSTRACT

Insulin causes distinct cortical actin remodeling in muscle and fat cells, and interfering with actin dynamics halts glucose transporter 4 (GLUT4) translocation to the membrane. Phosphatidylinositol 3-kinase (PI3-K) and the small G protein Rac govern myocyte actin remodeling, whereas TC10 alpha contributes to adipocyte actin dynamics downstream of Cbl-associated protein (CAP) and Cbl, independently of PI3-K. Given the importance of insulin action in both cell types, it is paramount to determine whether signaling pathways and actin manifestations are cell type specific. We found CAP expression and insulin-mediated Cbl phosphorylation in differentiated myotubes but not in myoblasts. Unlike adipocytes, Cbl is phosphorylated on Y774 and Y731 in myotubes. TC10 alpha and beta-transcripts are amplified by RT-PCR in muscle cells, but the endogenous proteins are barely detectable using two unrelated antibodies. TC10 alpha transfected into myoblasts is activated by insulin despite the lack of CAP expression and Cbl phosphorylation. Moreover, dominant-negative TC10 alpha mutants do not prevent insulin-induced actin remodeling in either myoblasts or myotubes and do not interfere with insulin-mediated recruitment of c-myc epitope-tagged GLUT4 to the cell surface. In contrast to TC10 alpha, endogenous Rac is readily detectable in both muscle cells and adipocytes and binds GTP after insulin in a PI3-K-dependent manner. These data suggest that whereas individual components of the CAP to TC10 pathway are regulated by insulin, a functional TC10-dependent signaling pathway leading to actin remodeling and GLUT4 translocation may not operate in myocytes, as it does in adipocytes.


Subject(s)
Actins/metabolism , Adipocytes/metabolism , Insulin/metabolism , Muscle, Skeletal/metabolism , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Actins/drug effects , Adipocytes/drug effects , Animals , Cells, Cultured , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/metabolism , Glucose Transporter Type 4 , Guanosine Triphosphate/metabolism , Insulin/pharmacology , Mice , Monosaccharide Transport Proteins/drug effects , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Muscle Proteins/drug effects , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Transport , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-cbl , Ubiquitin-Protein Ligases/drug effects , Ubiquitin-Protein Ligases/metabolism , rac GTP-Binding Proteins/drug effects , rho GTP-Binding Proteins/drug effects , rho GTP-Binding Proteins/genetics
20.
J Leukoc Biol ; 76(1): 254-62, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15107457

ABSTRACT

The production of reactive oxygen species (ROS) by human neutrophils is imperative for their bactericidal activity. Proinflammatory agents such as granulocyte macrophage-colony stimulating factor (GM-CSF) can prime ROS production in response to chemoattractants such as N-formyl-L-methionyl-L-leucyl-L-phenylalanine (fMLP). In neutrophils from patients suffering from Myelodysplastic syndromes (MDS), a clonal, hematological disorder characterized by recurrent bacterial infections, this GM-CSF priming is severely impaired. In this study, we set out to further delineate the defects in neutrophils from MDS patients. We examined the effect of GM-CSF priming on fMLP-triggered activation of Rac, a small GTPase implicated in neutrophil ROS production. In contrast to healthy neutrophils, activation of Rac in response to fMLP was not enhanced by GM-CSF pretreatment in MDS neutrophils. Furthermore, activation of Rac was attenuated by pretreatment of neutrophils with the phosphatidylinositol 3-kinase (PI-3K) inhibitor LY294002. Unlike healthy neutrophils, fMLP-induced accumulation of the PI-3K lipid product PI(3,4,5)trisphosphate was not increased by GM-CSF pretreatment in MDS neutrophils. The disturbed Rac and PI-3K activation observed in MDS neutrophils did not appear to reflect a general GM-CSF or fMLP receptor-signaling defect, as fMLP-triggered Ras activation could be primed by GM-CSF in MDS and healthy neutrophils. Moreover, fMLP-induced activation of the GTPase Ral was also normal in neutrophils from MDS patients. Taken together, our data suggest that in neutrophils from MDS patients, a defect in priming of the PI-3K-Rac signaling pathway, located at the level of PI-3K, results in a decreased GM-CSF priming of ROS production.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/metabolism , Phosphatidylinositol Phosphates/metabolism , rac GTP-Binding Proteins/metabolism , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Humans , Myelodysplastic Syndromes , Neutrophil Activation/drug effects , Neutrophil Activation/physiology , Neutrophils/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Reactive Oxygen Species/metabolism , rac GTP-Binding Proteins/drug effects , ral GTP-Binding Proteins/drug effects , ral GTP-Binding Proteins/metabolism , ras Proteins/drug effects , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL