Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Semin Cancer Biol ; 54: 29-39, 2019 02.
Article in English | MEDLINE | ID: mdl-29621614

ABSTRACT

Ras oncoproteins play pivotal roles in both the development and maintenance of many tumor types. Unfortunately, these proteins are difficult to directly target using traditional pharmacological strategies, in part due to their lack of obvious binding pockets or allosteric sites. This obstacle has driven a considerable amount of research into pursuing alternative ways to effectively inhibit Ras, examples of which include inducing mislocalization to prevent Ras maturation and inactivating downstream proteins in Ras-driven signaling pathways. Ras proteins are archetypes of a superfamily of small GTPases that play specific roles in the regulation of many cellular processes, including vesicle trafficking, nuclear transport, cytoskeletal rearrangement, and cell cycle progression. Several other superfamily members have also been linked to the control of normal and cancer cell growth and survival. For example, Rap1 has high sequence similarity to Ras, has overlapping binding partners, and has been demonstrated to both oppose and mimic Ras-driven cancer phenotypes. Rap1 plays an important role in cell adhesion and integrin function in a variety of cell types. Mechanistically, Ras and Rap1 cooperate to initiate and sustain ERK signaling, which is activated in many malignancies and is the target of successful therapeutics. Here we review the role activated Rap1 in ERK signaling and other downstream pathways to promote invasion and cell migration and metastasis in various cancer types.


Subject(s)
rap1 GTP-Binding Proteins/metabolism , ras Proteins/metabolism , Animals , Biomarkers, Tumor , Cell Adhesion/genetics , Energy Metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic/drug effects , Humans , Integrins/genetics , Integrins/metabolism , Molecular Targeted Therapy , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction/drug effects , rap1 GTP-Binding Proteins/antagonists & inhibitors , rap1 GTP-Binding Proteins/genetics , ras Proteins/antagonists & inhibitors , ras Proteins/genetics
2.
J Immunol ; 196(7): 3019-31, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26921307

ABSTRACT

Regulation of thymocyte trafficking plays an important role during thymic selection, but our understanding of the molecular mechanisms underlying these processes is limited. In this study, we demonstrated that class III semaphorin E (sema3e), a guidance molecule during neural and vascular development, directly inhibited Rap1 activation and LFA-1-dependent adhesion through the GTPase-activating protein activity of plexin D1. Sema3e inhibited Rap1 activation of thymocytes in response to chemokines and TCR stimulation, LFA-mediated adhesion, and T cell-APC interactions. Immunological synapse (IS) formation in mature thymocytes on supported lipid bilayers was also attenuated by sema3e. Impaired IS formation was associated with reduced Rap1 activation on the contact surface and cell periphery. Moreover, a significant increase of CD4(+) thymocytes was detected in the medulla of mice with T cell lineage-specific deletion of plexin D1. Two-photon live imaging of thymic explants and slices revealed enhanced Rap1 activation and migration of CD69(+) double-positive and single-positive cells with plexin D1 deficiency. Our results demonstrate that sema3e/plexin D1 modulates IS formation and Ag-scanning activities of thymocytes within thymic tissues.


Subject(s)
Glycoproteins/metabolism , Immunological Synapses/metabolism , Immunomodulation , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Thymocytes/immunology , Thymocytes/metabolism , rap1 GTP-Binding Proteins/antagonists & inhibitors , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Cell Adhesion/genetics , Cell Communication , Cell Movement/genetics , Chemokines/metabolism , Cytoskeletal Proteins , Glycoproteins/genetics , Intracellular Signaling Peptides and Proteins , Lymphocyte Function-Associated Antigen-1/chemistry , Lymphocyte Function-Associated Antigen-1/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Protein Interaction Domains and Motifs , Semaphorins , Signal Transduction , rac GTP-Binding Proteins/metabolism
3.
Can J Physiol Pharmacol ; 94(5): 498-507, 2016 May.
Article in English | MEDLINE | ID: mdl-26854595

ABSTRACT

Hepatic stellate cells (HSCs) activation represents an essential event during alcoholic liver fibrosis (ALF). Previous studies have demonstrated that the rat HSCs could be significantly activated after exposure to 200 µmol/L acetaldehyde for 48 h, and the cAMP/PKA signaling pathways were also dramatically upregulated in activated HSCs isolated from alcoholic fibrotic rat liver. Exchange protein activated by cAMP (EPAC) is a family of guanine nucleotide exchange factors (GEFs) for the small Ras-like GTPases Rap, and is being considered as a vital mediator of cAMP signaling in parallel with the principal cAMP target protein kinase A (PKA). Our data showed that both cAMP/PKA and cAMP/EPAC signaling pathways were involved in acetaldehyde-induced HSCs. Acetaldehyde could reduce the expression of EPAC1 while enhancing the expression of EPAC2. The cAMP analog Me-cAMP, which stimulates the EPAC/Rap1 pathway, could significantly decrease the proliferation and collagen synthesis of acetaldehyde-induced HSCs. Furthermore, depletion of EPAC2, but not EPAC1, prevented the activation of HSC measured as the production of α-SMA and collagen type I and III, indicating that EPAC1 appears to have protective effects on acetaldehyde-induced HSCs. Curiously, activation of PKA or EPAC perhaps has opposite effects on the synthesis of collagen and α-SMA: EPAC activation by Me-cAMP increased the levels of GTP-bound (activated) Rap1 while PKA activation by Phe-cAMP had no significant effects on such binding. These results suggested that EPAC activation could inhibit the activation and proliferation of acetaldehyde-induced HSCs via Rap1.


Subject(s)
Guanine Nucleotide Exchange Factors/agonists , Hepatic Stellate Cells/drug effects , Liver Cirrhosis, Alcoholic/metabolism , rap1 GTP-Binding Proteins/agonists , Acetaldehyde/antagonists & inhibitors , Acetaldehyde/toxicity , Actins/agonists , Actins/antagonists & inhibitors , Actins/genetics , Actins/metabolism , Animals , Cell Line , Cell Proliferation/drug effects , Collagen Type I/agonists , Collagen Type I/antagonists & inhibitors , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type II/agonists , Collagen Type II/antagonists & inhibitors , Collagen Type II/genetics , Collagen Type II/metabolism , Cyclic AMP/agonists , Cyclic AMP/analogs & derivatives , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation/drug effects , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver Cirrhosis, Alcoholic/pathology , Liver Cirrhosis, Alcoholic/prevention & control , RNA Interference , Rats , Second Messenger Systems/drug effects , rap1 GTP-Binding Proteins/antagonists & inhibitors , rap1 GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/metabolism
4.
Biochem Pharmacol ; 192: 114750, 2021 10.
Article in English | MEDLINE | ID: mdl-34461118

ABSTRACT

Statins decrease the serum LDL-cholesterol concentration and reduce the risk for cardiovascular diseases but can cause myopathy, which may be related to mTORC inhibition. In the current study, we investigated which mTORC is inhibited by simvastatin and by which mechanisms. In C2C12 myoblasts and myotubes and mouse gastrocnemius, simvastatin was cytotoxic and inhibited S6rp and Akt Ser473 phosphorylation, indicating inhibition of mTORC1 and mTORC2, respectively. In contrast to simvastatin, the mTORC1 inhibitor rapamycin did not inhibit mTORC2 activity and was not cytotoxic. Like simvastatin, knock-down of Rictor, an essential component of mTORC2, impaired Akt Ser473 and S6rp phosphorylation and was cytotoxic for C2C12 myoblasts, suggesting that mTORC2 inhibition is an important myotoxic mechanism. The investigation of the mechanism of mTORC2 inhibition showed that simvastatin impaired Ras farnesylation, which was prevented by farnesol but without restoring mTORC2 activity. In comparison, Rap1 knock-down reduced mTORC2 activity and was cytotoxic for C2C12 myoblasts. Simvastatin impaired Rap1 geranylgeranylation and function, which was prevented by geranylgeraniol. In addition, simvastatin and the complex III inhibitor antimycin A caused mitochondrial superoxide accumulation and impaired the activity of mTORC2, which could partially be prevented by the antioxidant MitoTEMPO. In conclusion, mTORC2 inhibition is an important mechanism of simvastatin-induced myotoxicity. Simvastatin inhibits mTORC2 by impairing geranylgeranylation of Rap1 and by inducing mitochondrial dysfunction.


Subject(s)
Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Mitochondria/drug effects , Muscle, Skeletal/drug effects , Prenylation/drug effects , Simvastatin/toxicity , rap1 GTP-Binding Proteins/antagonists & inhibitors , Animals , Cell Line , Drug Delivery Systems/methods , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Male , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Myoblasts/drug effects , Myoblasts/metabolism , Prenylation/physiology , Simvastatin/administration & dosage , rap1 GTP-Binding Proteins/metabolism
5.
Biomed Pharmacother ; 139: 111488, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33957564

ABSTRACT

Despite the renal expression of P2Y12, the purinergic receptor for adenosine diphosphate, few data are available to discuss the renotherapeutic potential of ticagrelor, one of its reversible blockers. Indeed, the tonic inhibitory effect of this receptor has been linked to the activation of exchange protein activated by cyclic adenosine monophosphate-1 (Epac-1) protein through the cyclic adenosine monophosphate cascade. Epac-1 is considered a crossroad protein, where its activation has been documented to manage renal injury models. Hence, the current study aimed to investigate the possible therapeutic effectiveness of ticagrelor, against renal ischemia/reperfusion (I/R) model with emphasis on the involvement of Epac-1 signaling pathway using R-CE3F4, a selective Epac-1 blocker. Accordingly, rats were randomized into four groups; viz., sham-operated, renal I/R, I/R post-treated with ticagrelor for 3 days, and ticagrelor + R-CE3F4. Treatment with ticagrelor ameliorated the I/R-mediated structural alterations and improved renal function manifested by the reduction in serum BUN and creatinine. On the molecular level, ticagrelor enhanced renal Epac-1 mRNA expression, Rap-1 activation (Rap-1-GTP) and SOCS-3 level. On the contrary, it inhibited the protein expression of JAK-2/STAT-3 hub, TNF-α and MDA contents, as well as caspase-3 activity. Additionally, ticagrelor enhanced the protein expression/content of AKT/Nrf-2/HO-1 axis. All these beneficial effects were obviously antagonized upon using R-CE3F4. In conclusion, ticagrelor reno-therapeutic effect is partly mediated through modulating the Epac-1/Rap-1-GTP, AKT/Nrf-2/HO-1 and JAK-2/STAT-3/SOCS-3 trajectories, pathways that integrate to afford novel explanations to its anti-inflammatory, anti-oxidant, and anti-apoptotic potentials.


Subject(s)
Acute Kidney Injury/drug therapy , Guanine Nucleotide Exchange Factors/drug effects , Purinergic P2Y Receptor Antagonists/therapeutic use , Reperfusion Injury/drug therapy , Signal Transduction/drug effects , Ticagrelor/therapeutic use , rap1 GTP-Binding Proteins/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Ischemia , Kidney Function Tests , Male , Rats , Rats, Wistar , Renal Circulation/drug effects , rap1 GTP-Binding Proteins/antagonists & inhibitors
6.
J Cell Biol ; 161(2): 417-27, 2003 Apr 28.
Article in English | MEDLINE | ID: mdl-12707305

ABSTRACT

Chemokines arrest circulating lymphocytes within the vasculature through the rapid up-regulation of leukocyte integrin adhesive activity, promoting subsequent lymphocyte transmigration. However, the key regulatory molecules regulating this process have remained elusive. Here, we demonstrate that Rap1 plays a pivotal role in chemokine-induced integrin activation and migration. Rap1 was activated by secondary lymphoid tissue chemokine (SLC; CCL21) and stromal-derived factor 1 (CXCL4) treatment in lymphocytes within seconds. Inhibition of Rap1 by Spa1, a Rap1-specific GTPase-activating protein, abrogated chemokine-stimulated lymphocyte rapid adhesion to endothelial cells under flow via intercellular adhesion molecule 1. Expression of a dominant active Rap1V12 in lymphocytes stimulated shear-resistant adhesion, robust cell migration on immobilized intercellular adhesion molecule 1 and vascular cell adhesion molecule 1, and transendothelial migration under flow. We also demonstrated that Rap1V12 expression in lymphocytes induced a polarized morphology, accompanied by the redistribution of CXCR4 and CD44 to the leading edge and uropod, respectively. Spa1 effectively suppressed this polarization after SLC treatment. This unique characteristic of Rap1 may control chemokine-induced lymphocyte extravasation.


Subject(s)
Cell Polarity/physiology , Chemokines/metabolism , Chemotaxis, Leukocyte/physiology , Endothelium, Vascular/metabolism , GTPase-Activating Proteins , Integrins/metabolism , Lymphocytes/metabolism , rap1 GTP-Binding Proteins/metabolism , Animals , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Polarity/drug effects , Cells, Cultured , Chemokine CCL21 , Chemokine CXCL12 , Chemokines, CC/metabolism , Chemokines, CC/pharmacology , Chemokines, CXC/metabolism , Chemokines, CXC/pharmacology , Chemotaxis, Leukocyte/drug effects , Hemodynamics/drug effects , Hemodynamics/physiology , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Lymphocytes/drug effects , Mice , Mice, Inbred BALB C , Nuclear Proteins/metabolism , Nuclear Proteins/pharmacology , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , rap1 GTP-Binding Proteins/antagonists & inhibitors , rap1 GTP-Binding Proteins/genetics
7.
Cell Death Dis ; 10(7): 535, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31296845

ABSTRACT

Excitotoxic neuronal death induced by high concentrations of glutamate is a pathological event common to multiple acute or chronic neurodegenerative diseases. Excitotoxicity is mediated through overactivation of the N-Methyl-D-aspartate type of ionotropic glutamate receptors (NMDARs). Physiological stimulation of NMDARs triggers their endocytosis from the neuronal surface, inducing synaptic activity and survival. However almost nothing is known about the internalization of overactivated NMDARs and their interacting proteins, and how this endocytic process is connected with neuronal death has been poorly explored. Kinase D-interacting substrate of 220 kDa (Kidins220), also known as ankyrin repeat-rich membrane spanning (ARMS), is a component of NMDAR complexes essential for neuronal viability by the control of ERK activation. Here we have investigated Kidins220 endocytosis induced by NMDAR overstimulation and the participation of this internalization step in the molecular mechanisms of excitotoxicity. We show that excitotoxicity induces Kidins220 and GluN1 traffic to the Golgi apparatus (GA) before Kidins220 is degraded by the protease calpain. We also find that excitotoxicity triggers an early activation of Rap1-GTPase followed by its inactivation. Kidins220 excitotoxic endocytosis and subsequent calpain-mediated downregulation governs this late inactivation of Rap1 that is associated to decreases in ERK activity preceding neuronal death. Furthermore, we identify the molecular mechanisms involved in the excitotoxic shutoff of Kidins220/Rap1/ERK prosurvival cascade that depends on calpain processing of Rap1-activation complexes. Our data fit in a model where Kidins220 targeting to the GA during early excitotoxicity would facilitate Rap1 activation and subsequent stimulation of ERK. At later times, activation of Golgi-associated calpain, would promote the degradation of GA-targeted Kidins220 and two additional components of the specific Rap1 activation complex, PDZ-GEF1, and S-SCAM. In this way, late excitotoxicity would turn off Rap1/ERK cascade and compromise neuronal survival.


Subject(s)
Calpain/metabolism , Golgi Apparatus/metabolism , Membrane Proteins/metabolism , Neurons/metabolism , Phosphoproteins/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , rap1 GTP-Binding Proteins/metabolism , Animals , Calpain/antagonists & inhibitors , Cell Death/drug effects , Cell Death/genetics , Cells, Cultured , Endocytosis/drug effects , Endocytosis/genetics , Endosomes/metabolism , Glutamic Acid/metabolism , Golgi Apparatus/drug effects , Membrane Proteins/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/drug effects , Neurons/enzymology , Neurons/ultrastructure , Phosphoproteins/genetics , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics , Signal Transduction/genetics , rab5 GTP-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/antagonists & inhibitors , rap1 GTP-Binding Proteins/genetics
8.
Mol Immunol ; 44(7): 1729-36, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17045653

ABSTRACT

The TLR agonists, flagellin (FLG) and lipopolysaccharide (LPS) stimulate functional activation and cytokine gene expression via the extracellular signal regulated kinase 1/2 (ERK1/2) MAP kinase cascade. However, the upstream mechanisms of these signaling events remain unknown. In mammals, the small GTP-binding protein Ras mediates ERK1/2 activation through activation of downstream effectors Raf-1-MEK1/2-ERK1/2 in response to a variety of stimuli. It is not clear whether this classic Ras cascade plays a role in TLR signaling in avian cells. In the present study, we investigated the role of Ras in FLG- and LPS-mediated signaling in ERK activation in chicken heterophils. Treatment of heterophils with LPS caused a rapid (within 5min) activation of Ras-GTP. The role of Ras activation in LPS-induced stimulation of ERK1/2 was corroborated when the specific Ras inhibitor, FTI-277, inhibited ERK1/2 activation. The classic Ras-mediated pathway of ERK1/2 activation by LPS was confirmed when the specific Raf-1 inhibitor, GW 5074, and the MEK1/2 inhibitor, U0126, both reduced ERK activation by 51-60%. Of more interest was that treatment of the heterophils with FLG did not activate Ras-GTP. Likewise, neither FTI-277 nor GW 5074 had any effect on FLG-mediated activation of ERK1/2. Another small GTPase, Rap1, has been shown to play a role in mammalian neutrophil function. Using a Rap1-GTP pull-down assay, we found that FLG stimulation, but not LPS, of avian heterophils induced a rapid and transient Rap1 activation. Rap1 has been shown to activate the ERK1/2 via a different Raf family member B-Raf whose downstream effector is MEK1/2. We show here that FLG stimulation of heterophils induces the phosphorylation of Rap1. The FLG induction of the Rap1-->B-Raf-->MEK1/2-->ERK1/2 cascade was confirmed by the reduction of ERK1/2 activation by the specific Rap1 inhibitor (GGTI-298) and U0126. The results demonstrate that for the first time that the small GTPase Ras family is involved in TLR signaling of avian heterophils with the TLR agonists LPS (Ras) and FLG (Rap1) inducing differential signaling cascades to activate the downstream ERK MAP kinase.


Subject(s)
Chickens/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Flagellin/immunology , Lipopolysaccharides/immunology , rap1 GTP-Binding Proteins/physiology , ras Proteins/physiology , Animals , Enzyme Activation , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Flagellin/pharmacology , Lipopolysaccharides/pharmacology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Monomeric GTP-Binding Proteins/antagonists & inhibitors , Monomeric GTP-Binding Proteins/physiology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/metabolism , Signal Transduction/drug effects , Toll-Like Receptors/agonists , rap1 GTP-Binding Proteins/antagonists & inhibitors , ras Proteins/antagonists & inhibitors
9.
PLoS One ; 12(3): e0173085, 2017.
Article in English | MEDLINE | ID: mdl-28278256

ABSTRACT

Previously, we reported that cAMP/PKA signaling is involved in GPER-mediated coronary relaxation by activating MLCP via inhibition of RhoA pathway. In the current study, we tested the hypothesis that activation of GPER induces coronary artery relaxation via inhibition of RhoA/Rho kinase pathway by cAMP downstream targets, exchange proteins directly activated by cAMP (Epac) as well as PKA. Our results show that Epac inhibitors, brefeldin A (BFA, 50 µM), or ESI-09 (20 µM), or CE3F4 (100 µM), all partially inhibited porcine coronary artery relaxation response to the selective GPER agonist, G-1 (0.3-3 µM); while concurrent administration of BFA and PKI (5 µM), a PKA inhibitor, almost completely blocked the relaxation effect of G-1. The Epac specific agonist, 8-CPT-2Me-cAMP (007, 1-100 µM), induced a concentration-dependent relaxation response. Furthermore, the activity of Ras-related protein 1 (Rap1) was up regulated by G-1 (1 µM) treatment of porcine coronary artery smooth muscle cells (CASMCs). Phosphorylation of vasodilator-stimulated phosphoprotein (p-VASP) was elevated by G-1 (1 µM) treatment, but not by 007 (50 µM); and the effect of G-1 on p-VASP was blocked by PKI, but not by ESI-09, an Epac antagonist. RhoA activity was similarly down regulated by G-1 and 007, whereas ESI-09 restored most of the reduced RhoA activity by G-1 treatment. Furthermore, G-1 decreased PGF2α-induced p-MYPT1, which was partially reversed with either ESI-09 or PKI; whereas, concurrent administration of ESI-09 and PKI totally prevented the inhibitory effect of G-1. The inhibitory effects of G-1 on p- MLC levels in CASMCs were mostly restored by either ESI-09 or PKI. These results demonstrate that activation of GPER induces coronary artery relaxation via concurrent inhibition of RhoA/Rho kinase by Epac/Rap1 and PKA. GPER could be a potential drug target for preventing and treating cardiovascular diseases.


Subject(s)
Coronary Vessels/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Receptors, Estrogen/metabolism , rap1 GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cells, Cultured , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclopentanes/pharmacology , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/metabolism , Hydrazones/pharmacology , Isoxazoles/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myosin-Light-Chain Phosphatase/metabolism , Phosphorylation/drug effects , Quinolines/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Swine , Thionucleotides/pharmacology , rap1 GTP-Binding Proteins/antagonists & inhibitors , rap1 GTP-Binding Proteins/genetics
10.
Thromb Res ; 118(4): 509-21, 2006.
Article in English | MEDLINE | ID: mdl-16380154

ABSTRACT

The abundant Rap1 in platelets becomes activated when these cells are stimulated by various agonists, but its function has remained unknown. In view of this, we developed an assay to quantitatively measure activated Rap1 and used it to determine relationships between Rap1 activation and several platelet functions: integrin alpha2beta1 activation, tyrosine phosphorylation, and the release reaction. We looked at how these processes are affected by the protein kinase C inhibitor BIMI, tyrosine kinase inhibitor PP2, PI 3-kinase inhibitor wortmannin, and ADP scavenger apyrase. In CRP (collagen related peptide)-activated platelets, all the inhibitors severely inhibited Rap1 activation, but had little effect on integrin alpha2beta1 activation, indicating that the integrin activation mechanism is different from the Rap1 activation mechanism, at least in GPVI-dependent activation. With p85alpha-null mouse platelets, we demonstrated that Rap1 activation involves PI 3-kinase p85alpha-dependent tyrosine phosphorylation. All the inhibitors similarly decreased Rap1 activation and the serotonin release reaction, and the inhibition of Rap1 activation was not due to the lack of released ADP. Our results indicate that platelet Rap1 activation is closely related to the release reaction and not to integrin alpha2beta1 activation in GPVI-mediated platelet activation.


Subject(s)
Blood Platelets/metabolism , Platelet Membrane Glycoproteins/metabolism , rap1 GTP-Binding Proteins/metabolism , Androstadienes/pharmacology , Apyrase/pharmacology , Collagen/metabolism , Humans , Integrin alpha2beta1/antagonists & inhibitors , Integrin alpha2beta1/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Tyrosine/metabolism , Wortmannin , rap1 GTP-Binding Proteins/antagonists & inhibitors
11.
Cancer Res ; 59(19): 4919-26, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10519405

ABSTRACT

Ras malignant transformation requires posttranslational modification by farnesyltransferase (FTase). Here we report on the design and antitumor activity, in monotherapy as well as in combination therapy with cytotoxic agents, of a novel class of non-thiol-containing peptidomimetic inhibitors of FTase and the closely related family member geranylgeranyltransferase I (GGTase I). The non-thiol-containing FTI-2148 is highly selective for FTase (IC50, 1.4 nM) over GGTase I (IC50, 1700 nM), whereas GGTI-2154 is highly selective for GGTase I (21 nM) over FTase (IC50, 5600 nM). In whole cells, the corresponding methylester prodrug FTI-2153 is >3000-fold more potent at inhibiting H-Ras (IC50, 10 nM) than Rap1A processing, whereas GGTI-2166 is over 100-fold more selective at inhibiting Rap1A (IC50, 300 nM) over H-Ras processing. Furthermore, FTI-2153 was highly effective at suppressing oncogenic H-Ras constitutive activation of mitogen-activated protein kinase and human tumor growth in soft agar. FTI-2148 suppressed the growth of the human lung adenocarcinoma A-549 cells in nude mice by 33, 67, and 91% in a dose-dependent manner. Combination therapy of FTI-2148 with either cisplatin, gemcitabine, or Taxol resulted in a greater antitumor efficacy than monotherapy. GGTI-2154 in similar antitumor efficacy experiments is less potent than FTI-2148 and inhibits tumor growth by 9, 27, and 46%. Combination therapy of GGTI-2154 with cisplatin, gemcitabine, or Taxol is also more effective. Finally, FTI-2148 and GGTI-2154 are 30- and 33-fold more selective and 30- and 16-fold more potent in whole cells than our previously reported thiol-containing FTI-276 and GGTI-297, respectively. Thus, our results demonstrate that this highly potent and selective novel class of non-thiol-containing peptidomimetics inhibits human tumor growth in whole animals and that combination therapy with cytotoxic agents is more beneficial than monotherapy.


Subject(s)
Adenocarcinoma/drug therapy , Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzamides/chemistry , Benzamides/therapeutic use , Enzyme Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Oligopeptides/chemistry , 3T3 Cells , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Antineoplastic Combined Chemotherapy Protocols/toxicity , Benzamides/toxicity , Cell Division/drug effects , Cisplatin/administration & dosage , Cisplatin/therapeutic use , Cisplatin/toxicity , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Deoxycytidine/toxicity , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/toxicity , Farnesyltranstransferase , Humans , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/drug effects , Molecular Structure , Paclitaxel/administration & dosage , Paclitaxel/therapeutic use , Paclitaxel/toxicity , Transplantation, Heterologous , Tumor Cells, Cultured , rap1 GTP-Binding Proteins/antagonists & inhibitors , ras Proteins/antagonists & inhibitors , Gemcitabine
12.
J Mol Med (Berl) ; 94(1): 13-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26423530

ABSTRACT

Platelets are critical for hemostasis, i.e., the body's ability to prevent blood loss at sites of vascular injury. They patrol the vasculature in a quiescent, non-adhesive state for approximately 10 days, after which they are removed from circulation by phagocytic cells of the reticulo-endothelial system. At sites of vascular injury, they promptly shift to an activated, adhesive state required for the formation of a hemostatic plug. The small GTPase RAP1 is a critical regulator of platelet adhesiveness. Our recent studies demonstrate that the antagonistic balance between the RAP1 regulators, CalDAG-GEFI and RASA3, is critical for the modulation of platelet adhesiveness, both in circulation and at sites of vascular injury. The RAP1 activator CalDAG-GEFI responds to small changes in the cytoplasmic calcium concentration and thus provides sensitivity and speed to the activation response, essential for efficient platelet adhesion under conditions of hemodynamic shear stress. The RAP1 inhibitor RASA3 ensures that circulating platelets remain quiescent by restraining CalDAG-GEFI-dependent RAP1 activation. Upon cellular stimulation, it is turned off by P2Y12 signaling to enable sustained RAP1 activation, required for the formation of a stable hemostatic plug. This review will summarize important studies that elucidated the signaling pathways that control RAP1 activation in platelets.


Subject(s)
Blood Platelets/physiology , Guanine Nucleotide Exchange Factors/metabolism , Platelet Activation/physiology , Platelet Adhesiveness/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Telomere-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/metabolism , Animals , Calcium/metabolism , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Humans , Mice , Mice, Knockout , Phosphatidylinositol 4,5-Diphosphate/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Purinergic P2Y12/metabolism , Shelterin Complex , Signal Transduction , Telomere-Binding Proteins/antagonists & inhibitors , Vascular System Injuries/physiopathology , rap1 GTP-Binding Proteins/antagonists & inhibitors
13.
Oncogene ; 21(39): 6101-12, 2002 Sep 05.
Article in English | MEDLINE | ID: mdl-12203122

ABSTRACT

Retinal pigmented epithelial (RPE) cell integrity is critical to the maintenance of retina functions and RPE cells do not proliferate in adults. The activation of RPE results in cell proliferation which may be associated with proliferative retinopathy and choroidal melanoma. Mitogen-activated protein kinase (MAPK) is believed to be a key participant in the response to mitogenic stimuli. We therefore investigated the involvement of the extracellular signal-regulated protein kinase (ERK) 1 and 2 during the induction of RPE cell proliferation. After foetal calf serum (FCS) stimulation activation of the Ras/Raf/ERK signalling pathway was detected by Western blotting and immunochemistry, with specific anti-phosphosignalling protein antibodies. Pharmacological and antisense (AS) oligonucleotide (ODN) strategies were used to analyse the signalling involved in FCS-induced RPE cell proliferation. Activation of the small G protein Ras and, to a lesser extent of Raf-1, the kinase directly downstream from Ras, was necessary to FCS-induced cell proliferation. MEK1/2 and ERK1/2 were activated during cell proliferation. Inhibition of MEK1/2 with UO 126 completely abolished ERK1/2 activation and reduced cell proliferation by 33-43%. ERK1/2 depletion by an AS ODN approach reduced cell proliferation by 27-33%, confirming the role of ERK1/2 in the FCS stimulation of RPE cells. We also investigated the role of PKA/cAMP, one of the major inhibitory pathways of ERK1/2. PKA blockade did not modify ERK1/2 activation or cell proliferation. In contrast, agents that increased cAMP concentration, abolished RPE proliferation, and MEK/ERK activation. Moreover, inhibition of the cAMP-activated small G protein Rap1, partially reversed the inhibitory effects of cAMP on cell proliferation and MEK/ERK activation. The requirement for Ras and ERK1/2, the lack of ERK1/2 regulation by PKA and the cAMP/Rap1 counter-regulatory pathway for ERK-mediated cell proliferation suggest complex regulation of signalling in RPE cells. These data may have important implications for the development of more selective models for retinal anti-proliferative therapies.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/pharmacology , Enzyme Inhibitors/pharmacology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Pigment Epithelium of Eye/drug effects , Signal Transduction/physiology , Animals , Blotting, Western , Cattle , Cell Division/drug effects , Humans , Immunoenzyme Techniques , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Oligonucleotides, Antisense/pharmacology , Phosphorylation , Pigment Epithelium of Eye/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/metabolism , rap1 GTP-Binding Proteins/antagonists & inhibitors , rap1 GTP-Binding Proteins/metabolism , ras Proteins/metabolism , src-Family Kinases/metabolism
14.
Thromb Haemost ; 93(2): 319-25, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15711749

ABSTRACT

The NO/cGMP signalling pathway strongly inhibits agonist-induced platelet aggregation. However, the molecular mechanisms involved are not completely defined. We have studied NO/cGMP effects on the activity of Rap 1, an abundant guanine-nucleotidebinding protein in platelets. Rap 1-GTP levels were reduced by NO-donors and activators of NO-sensitive soluble guanylyl cyclase. Four lines of evidence suggest that NO/cGMP effects are mediated by cGMP-dependent protein kinase (cGKI): (i) Rap 1 inhibition correlated with cGKI activity as measured by the phosphorylation state of VASP, an established substrate of cGKI, (ii) 8-pCPT-cGMP, a membrane permeable cGMP-analog and activator of cGKI, completely blocked Rap1 activation, (iii) Rp-8pCPT-cGMPS, a cGKI inhibitor, reversed NO effects and (iv) expression of cGKI in cGKI-deficient megakaryocytes inhibited Rap1 activation. NO/cGMP/cGKI effects were independent of the type of stimulus used for Rap1 activation. Thrombin-,ADP- and collagen-induced formation of Rap 1-GTP in platelets as well as turbulence-induced Rap 1 activation in megakaryocytes were inhibited. Furthermore, cGKI inhibited ADP-induced Rap 1 activation induced by the Galpha(i)-coupled P2Y12 receptor alone, i.e. independently of effects on Ca2+-signalling. From these studies we conclude that NO/cGMP inhibit Rap 1 activation in human platelets and that this effect is mediated by cGKI. Since Rap1 controls the function of integrin alpha(IIb)beta3, we propose that Rap 1 inhibition might play a central role in the anti-aggregatory actions of NO/cGMP.


Subject(s)
Blood Platelets/metabolism , Cyclic GMP-Dependent Protein Kinases/physiology , Cyclic GMP/metabolism , Nitric Oxide/metabolism , rap1 GTP-Binding Proteins/antagonists & inhibitors , Adenosine Diphosphate/pharmacology , Calcium Signaling , Cell Line , Collagen/pharmacology , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinases/genetics , Humans , Megakaryocytes/cytology , Megakaryocytes/metabolism , Nitric Oxide Donors/pharmacology , Transfection , rap1 GTP-Binding Proteins/metabolism
15.
Clin Exp Metastasis ; 32(6): 555-66, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26070429

ABSTRACT

The isoprenoid biosynthetic pathway (IBP) is critical for providing substrates for the post-translational modification of proteins key in regulating malignant cell properties, including proliferation, invasion, and migration. Inhibitors of the IBP, including statins and nitrogenous bisphosphonates, are used clinically for the treatment of hypercholesterolemia and bone disease respectively. The statins work predominantly in the liver, while the nitrogenous bisphosphonates are highly sequestered to bone. Inhibition of the entire IBP is limited by organ specificity and side effects resulting from depletion of all isoprenoids. We have developed a novel compound, disodium [(6Z,11E,15E)-9-[bis(sodiooxy)phosphoryl]-17-hydroxy-2,6,12,16-tetramethyheptadeca-2,6,11,15-tetraen-9-yl]phosphonate (GGOHBP), which selectively targets geranylgeranyl diphosphate synthase, reducing post-translational protein geranylgeranylation. Intracardiac injection of luciferase-expressing human-derived 22Rv1 PCa cells into SCID mice resulted in tumor development in bone (100 %), adrenal glands (72 %), mesentery (22 %), liver (17 %), and the thoracic cavity (6 %). Three weeks after tumor inoculation, daily subcutaneous (SQ) injections of 1.5 mg/kg GGOHBP or the vehicle were given for one month. Dissected tumors revealed a reduction in adrenal gland tumors corresponding to a 54 % (P < 0.005) reduction in total adrenal gland tumor weight of the treated mice as compared to vehicle-treated controls. Western blot analysis of the harvested tissues showed a reduction in Rap1A geranylgeranylation in adrenal glands and mesenteric tumors of the treated mice while non-tumorous tissues and control mice showed no Rap1A alteration. Our findings detail a novel bisphosphonate compound capable of preferentially altering the IBP in tumor-burdened adrenal glands of a murine model of PCa metastasis.


Subject(s)
Adrenal Gland Neoplasms/prevention & control , Diphosphonates/pharmacology , Disease Models, Animal , Prostatic Neoplasms/prevention & control , Protein Prenylation/drug effects , rap1 GTP-Binding Proteins/antagonists & inhibitors , Adrenal Gland Neoplasms/drug therapy , Adrenal Gland Neoplasms/metabolism , Adrenal Gland Neoplasms/secondary , Animals , Blotting, Western , Cell Proliferation/drug effects , Farnesyltranstransferase/metabolism , Humans , Male , Mice , Mice, SCID , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Tumor Burden , Tumor Cells, Cultured , rap1 GTP-Binding Proteins/metabolism
16.
Nat Cell Biol ; 17(1): 81-94, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25531777

ABSTRACT

We identified the Rho GTPase Rnd1 as a candidate metastasis suppressor in basal-like and triple-negative breast cancer through bioinformatics analysis. Depletion of Rnd1 disrupted epithelial adhesion and polarity, induced epithelial-to-mesenchymal transition, and cooperated with deregulated expression of c-Myc or loss of p53 to cause neoplastic conversion. Mechanistic studies revealed that Rnd1 suppresses Ras signalling by activating the GAP domain of Plexin B1, which inhibits Rap1. Rap1 inhibition in turn led to derepression of p120 Ras-GAP, which was able to inhibit Ras. Inactivation of Rnd1 in mammary epithelial cells induced highly undifferentiated and invasive tumours in mice. Conversely, Rnd1 expression inhibited spontaneous and experimental lung colonization in mouse models of metastasis. Genomic studies indicated that gene deletion in combination with epigenetic silencing or, more rarely, point mutation inactivates RND1 in human breast cancer. These results reveal a previously unappreciated mechanism through which Rnd1 restrains activation of Ras-MAPK signalling and breast tumour initiation and progression.


Subject(s)
Cell Transformation, Neoplastic/genetics , Epithelial-Mesenchymal Transition/genetics , Lung Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , rho GTP-Binding Proteins/physiology , Animals , Cell Adhesion/genetics , Cell Line, Tumor , Cell Polarity/genetics , Cell Proliferation/genetics , Cellular Senescence/genetics , Female , Humans , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-myc/biosynthesis , Receptors, Cell Surface/metabolism , Signal Transduction/genetics , Triple Negative Breast Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , p120 GTPase Activating Protein/biosynthesis , rap1 GTP-Binding Proteins/antagonists & inhibitors , ras GTPase-Activating Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/genetics
17.
Sci Rep ; 5: 8206, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25644492

ABSTRACT

Eph receptor (Eph) and ephrin signaling can play central roles in prostate cancer and other cancer types. Exposed to ephrin-A1 PC3 prostate cancer cells alter adhesion to extracellular matrix (ECM) proteins. However, whether PC3 cells increase or reduce adhesion, and by which mechanisms they change adhesion to the ECM remains to be characterized. Here, we assay how ephrin-A1 stimulates PC3 cells to adhere to ECM proteins using single-cell force spectroscopy. We find that PC3 cells binding to immobilized ephrin-A1 but not to solubilized ephrin-A1 specifically strengthen adhesion to collagen I. This Eph-ephrin-A1 signaling, which we suppose is based on mechanotransduction, stimulates ß1-subunit containing integrin adhesion via the protein kinase Akt and the guanine nucleotide-exchange factor cytohesin. Inhibiting the small GTPases, Rap1 or Rac1, generally lowered adhesion of PC3 prostate cancer cells. Our finding suggests a mechanism by which PC3 prostate cancer cells exposed to ephrins crosstalk to ß1-integrins and preferably metastasize in bone, a collagen I rich tissue.


Subject(s)
Collagen Type I/chemistry , Integrin beta1/metabolism , Receptors, Eph Family/metabolism , Animals , Cell Adhesion , Cell Communication , Cell Line, Tumor , Collagen Type I/metabolism , Ephrin-A1/chemistry , Ephrin-A1/pharmacology , HEK293 Cells , HeLa Cells , Humans , Immobilized Proteins/chemistry , Male , Mechanotransduction, Cellular/drug effects , Mice , Microscopy, Atomic Force , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/metabolism , rap1 GTP-Binding Proteins/antagonists & inhibitors , rap1 GTP-Binding Proteins/metabolism
18.
J Clin Invest ; 125(4): 1419-32, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25705885

ABSTRACT

The small GTPase RAP1 is critical for platelet activation and thrombus formation. RAP1 activity in platelets is controlled by the GEF CalDAG-GEFI and an unknown regulator that operates downstream of the adenosine diphosphate (ADP) receptor, P2Y12, a target of antithrombotic therapy. Here, we provide evidence that the GAP, RASA3, inhibits platelet activation and provides a link between P2Y12 and activation of the RAP1 signaling pathway. In mice, reduced expression of RASA3 led to premature platelet activation and markedly reduced the life span of circulating platelets. The increased platelet turnover and the resulting thrombocytopenia were reversed by concomitant deletion of the gene encoding CalDAG-GEFI. Rasa3 mutant platelets were hyperresponsive to agonist stimulation, both in vitro and in vivo. Moreover, activation of Rasa3 mutant platelets occurred independently of ADP feedback signaling and was insensitive to inhibitors of P2Y12 or PI3 kinase. Together, our results indicate that RASA3 ensures that circulating platelets remain quiescent by restraining CalDAG-GEFI/RAP1 signaling and suggest that P2Y12 signaling is required to inhibit RASA3 and enable sustained RAP1-dependent platelet activation and thrombus formation at sites of vascular injury. These findings provide insight into the antithrombotic effect of P2Y12 inhibitors and may lead to improved diagnosis and treatment of platelet-related disorders.


Subject(s)
GTPase-Activating Proteins/physiology , Platelet Activation/physiology , rap1 GTP-Binding Proteins/antagonists & inhibitors , Animals , Cellular Senescence , Clopidogrel , GTPase-Activating Proteins/genetics , Guanine Nucleotide Exchange Factors/deficiency , Hemostasis , Lymphopenia/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Platelet Activation/drug effects , Platelet Activation/genetics , Platelet Aggregation Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/physiology , Receptors, Purinergic P2Y12/physiology , Saphenous Vein/injuries , Splenectomy , Thrombocytopenia/genetics , Thrombocytopenia/surgery , Thrombopoiesis , Ticlopidine/analogs & derivatives , Ticlopidine/pharmacology , rap1 GTP-Binding Proteins/physiology
19.
Cell Signal ; 27(3): 532-44, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25562427

ABSTRACT

In addition to providing skeletal support, the bone is an endocrine organ that produces osteocalcin, whose uncarboxylated form (GluOC) increases insulin secretion either directly or indirectly by promoting incretin secretion. We have now investigated the signaling pathway by which GluOC increases expression of adiponectin in adipocytes. Activation of its putative receptor GPRC6A by GluOC induced the intracellular accumulation of cAMP and consequent activation of protein kinase A (PKA) in differentiated 3T3-L1 adipocytes. It also induced phosphorylation of CREB (cAMP response element binding protein), but this effect appeared to be mediated indirectly by extracellular signal-regulated kinase (ERK) rather than directly by PKA, given that it was attenuated by the ERK signaling inhibitor U0126. Activated PKA also induced activation of the tyrosine kinase Src, the small GTPase Rap1, an upstream of ERK and CREB phosphorylation. Activated CREB up-regulated the expression of peroxisome proliferator-activated receptor γ (PPARγ), which in turn led to induction of adiponectin expression. Finally, intermittent oral administration of GluOC in mice reduced the size of gonadal white adipocytes as well as increased the expression of PPARγ and adiponectin in these cells. Our results have thus revealed the signaling pathway by which GluOC induces adiponectin expression in adipocytes.


Subject(s)
Gene Expression Regulation , Osteocalcin/pharmacology , Signal Transduction/drug effects , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Adiponectin/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Butadienes/pharmacology , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation/drug effects , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Nitriles/pharmacology , PPAR gamma/metabolism , Phosphorylation/drug effects , RNA Interference , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , rap1 GTP-Binding Proteins/antagonists & inhibitors , rap1 GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/metabolism
20.
PLoS One ; 8(2): e57903, 2013.
Article in English | MEDLINE | ID: mdl-23469100

ABSTRACT

Rap1 and Rap2 are closely related proteins of the Ras family of small G-proteins. Rap1 is well known to regulate cell-cell adhesion. Here, we have analysed the effect of Rap-mediated signalling on endothelial permeability using electrical impedance measurements of HUVEC monolayers and subsequent determination of the barrier resistance, which is a measure for the ease with which ions can pass cell junctions. In line with its well-established effect on cell-cell junctions, depletion of Rap1 decreases, whereas activation of Rap1 increases barrier resistance. Despite its high sequence homology with Rap1, depletion of Rap2 has an opposite, enhancing, effect on barrier resistance. This effect can be mimicked by depletion of the Rap2 specific activator RasGEF1C and the Rap2 effector MAP4K4, establishing Rap2 signalling as an independent pathway controlling barrier resistance. As simultaneous depletion or activation of both Rap1 and Rap2 results in a barrier resistance comparable to control cells, Rap1 and Rap2 control barrier resistance in a reciprocal manner. This Rap1-antagonizing effect of Rap2 is established independent of junctional actin formation. These data establish that endothelial barrier resistance is determined by the combined antagonistic actions of Rap1 and Rap2.


Subject(s)
Endothelium/metabolism , rap GTP-Binding Proteins/antagonists & inhibitors , rap GTP-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/antagonists & inhibitors , rap1 GTP-Binding Proteins/metabolism , Gene Knockdown Techniques , HEK293 Cells , Humans , RNA, Small Interfering/genetics , rap GTP-Binding Proteins/deficiency , rap GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/deficiency , rap1 GTP-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL