Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
1.
Mol Psychiatry ; 26(7): 3223-3239, 2021 07.
Article in English | MEDLINE | ID: mdl-32651478

ABSTRACT

The neural molecular and biochemical response to stress is a distinct physiological process, and multiple lines of evidence indicate that the prefrontal cortex (PFC) is particularly sensitive to, and afflicted by, exposure to stress. Largely through this PFC dysfunction, stress has a characterized role in facilitating cognitive impairment, which is often dissociable from its effects on non-cognitive behaviors. The Rap1 small GTPase pathway has emerged as a commonly disrupted intracellular target in neuropsychiatric conditions, whether it be via alterations in Rap1 expression or through alterations in the expression of direct and specific upstream Rap1 activators and inhibitors. Here we demonstrate that escalating, intermittent stress increases Rap1 in mouse PFC synapses, results in cognitive impairments, and reduces the preponderance of mature dendritic spines in PFC neurons. Using viral-mediated gene transfer, we reveal that the hyper-induction of Rap1 in the PFC is sufficient to drive stress-relevant cognitive and synaptic phenotypes. These findings point to Rap1 as a critical mediator of stress-driven neuronal and behavioral pathology and highlight a previously unrecognized involvement for Rap1 in novelty-driven PFC engagement.


Subject(s)
Neuronal Plasticity , Prefrontal Cortex/physiopathology , Stress, Psychological/enzymology , rap1 GTP-Binding Proteins/physiology , Animals , Mice , Neurons , Synapses
2.
Sci Rep ; 10(1): 13221, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32764635

ABSTRACT

Integrin activation is associated with conformational regulation. In this study, we developed a system to evaluate conformational changes in α4ß7 integrin. We first inserted the PA tag into the plexin-semaphorin-integrin (PSI) domain of ß7 chain, which reacted with an anti-PA tag antibody (NZ-1) in an Mn2+-dependent manner. The small GTPase Rap1 deficiency, as well as chemokine stimulation and the introduction of the active form of Rap1, Rap1V12, enhanced the binding of NZ-1 to the PA-tagged mutant integrin, and increased the binding affinity to mucosal addressing cell adhesion molecule-1 (MAdCAM-1). Furthermore, we generated two kinds of hybridomas producing monoclonal antibodies (mAbs) that recognized Mn2+-dependent epitopes of ß7. Both epitopes were exposed to bind to mAbs on the cells by the introduction of Rap1V12. Although one epitope in the PSI domain of ß7 was exposed on Rap1-deficienct cells, the other epitope in the hybrid domain of ß7 was not. These data indicate that the conversion of Rap1-GDP to GTP exerts two distinct effects stepwise on the conformation of α4ß7. The induction of colitis by Rap1-deficient CD4+ effector/memory T cells suggests that the removal of constraining effect by Rap1-GDP on α4ß7 is sufficient for homing of these pathogenic T cells into colon lamina propria (LP).


Subject(s)
Antibodies, Monoclonal/immunology , Integrins/metabolism , rap1 GTP-Binding Proteins/physiology , Animals , Cell Line , Humans , Integrins/chemistry , Integrins/immunology , Jurkat Cells , Membrane Glycoproteins/chemistry , Mice , Peptides/immunology , Protein Conformation , Protein Domains , Rats , Structure-Activity Relationship
3.
Blood ; 136(10): 1180-1190, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32518959

ABSTRACT

Ras-related protein 1 (Rap1) is a major convergence point of the platelet-signaling pathways that result in talin-1 binding to the integrin ß cytoplasmic domain and consequent integrin activation, platelet aggregation, and effective hemostasis. The nature of the connection between Rap1 and talin-1 in integrin activation is an important remaining gap in our understanding of this process. Previous work identified a low-affinity Rap1-binding site in the talin-1 F0 domain that makes a small contribution to integrin activation in platelets. We recently identified an additional Rap1-binding site in the talin-1 F1 domain that makes a greater contribution than F0 in model systems. Here we generated mice bearing point mutations, which block Rap1 binding without affecting talin-1 expression, in either the talin-1 F1 domain (R118E) alone, which were viable, or in both the F0 and F1 domains (R35E,R118E), which were embryonic lethal. Loss of the Rap1-talin-1 F1 interaction in platelets markedly decreases talin-1-mediated activation of platelet ß1- and ß3-integrins. Integrin activation and platelet aggregation in mice whose platelets express only talin-1(R35E, R118E) are even more impaired, resembling the defect seen in platelets lacking both Rap1a and Rap1b. Although Rap1 is important in thrombopoiesis, platelet secretion, and surface exposure of phosphatidylserine, loss of the Rap1-talin-1 interaction in talin-1(R35E, R118E) platelets had little effect on these processes. These findings show that talin-1 is the principal direct effector of Rap1 GTPases that regulates platelet integrin activation in hemostasis.


Subject(s)
Integrin beta1/metabolism , Integrin beta3/metabolism , Point Mutation , Talin/physiology , Thrombopoiesis , rap GTP-Binding Proteins/physiology , rap1 GTP-Binding Proteins/physiology , Animals , Female , Integrin beta1/genetics , Integrin beta3/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Activation , Platelet Aggregation , Protein Domains , Signal Transduction
4.
Front Immunol ; 11: 738, 2020.
Article in English | MEDLINE | ID: mdl-32425937

ABSTRACT

Phagocytic integrins are endowed with the ability to engulf and dispose of particles of different natures. Evolutionarily conserved from worms to humans, they are involved in pathogen elimination and apoptotic and tumoral cell clearance. Research in the field of integrin-mediated phagocytosis has shed light on the molecular events controlling integrin activation and their effector functions. However, there are still some aspects of the regulation of the phagocytic process that need to be clarified. Here, we have revised the molecular events controlling phagocytic integrin activation and the downstream signaling driving particle engulfment, and we have focused particularly on αMß2/CR3, αXß2/CR4, and a brief mention of αVß5/αVß3integrins.


Subject(s)
Integrins/physiology , Phagocytosis/physiology , Adaptor Proteins, Signal Transducing/physiology , Animals , Apoptosis , Humans , Integrin alphaXbeta2/physiology , Integrins/chemistry , Macrophage-1 Antigen/physiology , Membrane Proteins/physiology , Protein-Tyrosine Kinases/physiology , Signal Transduction/physiology , Talin/physiology , rap1 GTP-Binding Proteins/physiology
5.
J Crohns Colitis ; 13(5): 648-658, 2019 Apr 26.
Article in English | MEDLINE | ID: mdl-30500874

ABSTRACT

BACKGROUND AND AIMS: Genome-wide association studies [GWASs] of European populations have identified numerous susceptibility loci for Crohn's disease [CD]. Susceptibility genes differ by ethnicity, however, so GWASs specific for Asian populations are required. This study aimed to clarify the Japanese-specific genetic background for CD by a GWAS using the Japonica array [JPA] and subsequent imputation with the 1KJPN reference panel. METHODS: Two independent Japanese case/control sets (Tohoku region [379 CD patients, 1621 controls] and Kyushu region [334 CD patients, 462 controls]) were included. GWASs were performed separately for each population, followed by a meta-analysis. Two additional replication sets [254 + 516 CD patients and 287 + 565 controls] were analysed for top hit single nucleotide polymorphisms [SNPs] from novel genomic regions. RESULTS: Genotype data of 4 335 144 SNPs from 713 Japanese CD patients and 2083 controls were analysed. SNPs located in TNFSF15 (rs78898421, Pmeta = 2.59 × 10-26, odds ratio [OR] = 2.10), HLA-DQB1 [rs184950714, pmeta = 3.56 × 10-19, OR = 2.05], ZNF365, and 4p14 loci were significantly associated with CD in Japanese individuals. Replication analyses were performed for four novel candidate loci [p <1 × 10-6], and rs488200 located upstream of RAP1A was significantly associated with CD [pcombined = 4.36 × 10-8, OR = 1.31]. Transcriptome analysis of CD4+ effector memory T cells from lamina propria mononuclear cells of CD patients revealed a significant association of rs488200 with RAP1A expression. CONCLUSIONS: RAP1A is a novel susceptibility locus for CD in the Japanese population.


Subject(s)
Crohn Disease/genetics , Genetic Predisposition to Disease/genetics , rap1 GTP-Binding Proteins/physiology , Adult , Case-Control Studies , Crohn Disease/epidemiology , Female , Gene Expression Profiling , Genome-Wide Association Study , Humans , Japan/epidemiology , Male , Polymorphism, Single Nucleotide/genetics , Young Adult , rap1 GTP-Binding Proteins/genetics
6.
Int J Mol Sci ; 19(10)2018 Sep 20.
Article in English | MEDLINE | ID: mdl-30241315

ABSTRACT

This review addresses the issue of the numerous roles played by Rap1 GTPase (guanosine triphosphatase) in different cell types, in terms of both physiology and pathology. It is one among a myriad of small G proteins with endogenous GTP-hydrolyzing activity that is considerably stimulated by posttranslational modifications (geranylgeranylation) or guanine nucleotide exchange factors (GEFs), and inhibited by GTPase-activating proteins (GAPs). Rap1 is a ubiquitous protein that plays an essential role in the control of metabolic processes, such as signal transduction from plasma membrane receptors, cytoskeleton rearrangements necessary for cell division, intracellular and substratum adhesion, as well as cell motility, which is needed for extravasation or fusion. We present several examples of how Rap1 affects cells and organs, pointing to possible molecular manipulations that could have application in the therapy of several diseases.


Subject(s)
rap1 GTP-Binding Proteins/physiology , Adaptive Immunity , Cell Differentiation , Cell Transformation, Neoplastic , Models, Molecular , Prenylation , Protein Processing, Post-Translational , Signal Transduction , rap1 GTP-Binding Proteins/chemistry , rap1 GTP-Binding Proteins/metabolism
7.
Proc Natl Acad Sci U S A ; 115(16): E3722-E3730, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29602807

ABSTRACT

Cell migration requires the coordination of an excitable signal transduction network involving Ras and PI3K pathways with cytoskeletal activity. We show that expressing activated Ras GTPase-family proteins in cells lacking PTEN or other mutations which increase cellular protrusiveness transforms cells into a persistently activated state. Leading- and trailing-edge markers were found exclusively at the cell perimeter and the cytosol, respectively, of the dramatically flattened cells. In addition, the lifetimes of dynamic actin puncta were increased where they overlapped with actin waves, suggesting a mechanism for the coupling between these two networks. All of these phenotypes could be reversed by inhibiting signal transduction. Strikingly, maintaining cells in this state of constant activation led to a form of cell death by catastrophic fragmentation. These findings provide insight into the feedback loops that control excitability of the signal transduction network, which drives migration.


Subject(s)
Dictyostelium/physiology , Protozoan Proteins/physiology , Signal Transduction/physiology , Actin Cytoskeleton/physiology , Actin Cytoskeleton/ultrastructure , Cell Adhesion , Cell Movement , Cell Shape , Chemotaxis , Dictyostelium/genetics , Dictyostelium/ultrastructure , Enzyme Activation , Microscopy, Fluorescence , Microscopy, Phase-Contrast , Mutation, Missense , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/physiology , Phenotype , Protozoan Proteins/genetics , Recombinant Proteins/metabolism , rap1 GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/physiology
8.
PLoS Genet ; 14(1): e1007195, 2018 01.
Article in English | MEDLINE | ID: mdl-29381707

ABSTRACT

Rasa3 is a GTPase activating protein of the GAP1 family which targets R-Ras and Rap1. Although catalytic inactivation or deletion of Rasa3 in mice leads to severe hemorrhages and embryonic lethality, the biological function and cellular location of Rasa3 underlying these defects remains unknown. Here, using a combination of loss of function studies in mouse and zebrafish as well as in vitro cell biology approaches, we identify a key role for Rasa3 in endothelial cells and vascular lumen integrity. Specific ablation of Rasa3 in the mouse endothelium, but not in megakaryocytes and platelets, lead to embryonic bleeding and death at mid-gestation, recapitulating the phenotype observed in full Rasa3 knock-out mice. Reduced plexus/sprouts formation and vascular lumenization defects were observed when Rasa3 was specifically inactivated in mouse endothelial cells at the postnatal or adult stages. Similar results were obtained in zebrafish after decreasing Rasa3 expression. In vitro, depletion of Rasa3 in cultured endothelial cells increased ß1 integrin activation and cell adhesion to extracellular matrix components, decreased cell migration and blocked tubulogenesis. During migration, these Rasa3-depleted cells exhibited larger and more mature adhesions resulting from a perturbed dynamics of adhesion assembly and disassembly which significantly increased their life time. These defects were due to a hyperactivation of the Rap1 GTPase and blockade of FAK/Src signaling. Finally, Rasa3-depleted cells showed reduced turnover of VE-cadherin-based adhesions resulting in more stable endothelial cell-cell adhesion and decreased endothelial permeability. Altogether, our results indicate that Rasa3 is a critical regulator of Rap1 in endothelial cells which controls adhesions properties and vascular lumen integrity; its specific endothelial cell inactivation results in occluded blood vessels, hemorrhages and early embryonic death in mouse, mimicking thus the Rasa3-/- mouse phenotype.


Subject(s)
Capillary Permeability/genetics , Cell Adhesion/genetics , Endothelial Cells/physiology , Endothelium, Vascular/metabolism , GTPase-Activating Proteins/physiology , rap1 GTP-Binding Proteins/physiology , Animals , Animals, Genetically Modified , Cells, Cultured , Embryo, Mammalian , Embryo, Nonmammalian , Female , GTPase-Activating Proteins/genetics , Human Umbilical Vein Endothelial Cells , Humans , Male , Megakaryocytes/physiology , Mice , Mice, Knockout , Signal Transduction , Zebrafish , rap1 GTP-Binding Proteins/genetics
9.
J Nippon Med Sch ; 84(4): 148-159, 2017.
Article in English | MEDLINE | ID: mdl-28978894

ABSTRACT

Endothelial cells lining blood vessels regulate vascular barrier function, which controls the passage of plasma proteins and circulating cells across the endothelium. In most normal adult tissues, endothelial cells preserve basal vascular permeability at a low level, while they increase permeability in response to inflammation. Therefore, vascular permeability is tightly controlled by a number of extracellular stimuli and mediators to maintain tissue homeostasis. Accordingly, impaired regulation of endothelial permeability causes various diseases, including chronic inflammation, asthma, edema, sepsis, acute respiratory distress syndrome, anaphylaxis, tumor angiogenesis, and diabetic retinopathy. Vascular endothelial (VE)-cadherin, a member of the classical cadherin superfamily, is a component of cell-to-cell adherens junctions in endothelial cells and plays an important role in regulating vascular permeability. VE-cadherin mediates intercellular adhesion through trans-interactions formed by its extracellular domain, while its cytoplasmic domain is anchored to the actin cytoskeleton via α- and ß-catenins, leading to stabilization of VE-cadherin at cell-cell junctions. VE-cadherin-mediated cell adhesions are dynamically, but tightly, controlled by mechanisms that involve protein phosphorylation and reorganization of the actomyosin cytoskeleton. Phosphorylation of VE-cadherin, and its associated-catenins, results in dissociation of the VE-cadherin/catenin complex and internalization of VE-cadherin, leading to increased vascular permeability. Furthermore, reorganization of the actomyosin cytoskeleton by Rap1, a small GTPase that belongs to the Ras subfamily, and Rho family small GTPases, regulates VE-cadherin-mediated cell adhesions to control vascular permeability. In this review, we describe recent progress in understanding the signaling mechanisms that enable dynamic regulation of VE-cadherin adhesions and vascular permeability. In addition, we discuss the possibility of novel therapeutic approaches targeting the signaling pathways controlling VE-cadherin-mediated cell adhesion in diseases associated with vascular hyper-permeability.


Subject(s)
Antigens, CD/metabolism , Antigens, CD/physiology , Cadherins/metabolism , Cadherins/physiology , Endothelial Cells/metabolism , Endothelial Cells/physiology , Intercellular Junctions/metabolism , Intercellular Junctions/physiology , Actin Cytoskeleton/metabolism , Cell Adhesion/genetics , Cell Adhesion/physiology , Humans , Phosphorylation , rap1 GTP-Binding Proteins/physiology
10.
Proc Natl Acad Sci U S A ; 113(11): 3036-41, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26929333

ABSTRACT

cAMP signaling plays a key role in regulating pain sensitivity. Here, we uncover a previously unidentified molecular mechanism in which direct phosphorylation of the exchange protein directly activated by cAMP 1 (EPAC1) by G protein kinase 2 (GRK2) suppresses Epac1-to-Rap1 signaling, thereby inhibiting persistent inflammatory pain. Epac1(-/-) mice are protected against inflammatory hyperalgesia in the complete Freund's adjuvant (CFA) model. Moreover, the Epac-specific inhibitor ESI-09 inhibits established CFA-induced mechanical hyperalgesia without affecting normal mechanical sensitivity. At the mechanistic level, CFA increased activity of the Epac target Rap1 in dorsal root ganglia of WT, but not of Epac1(-/-), mice. Using sensory neuron-specific overexpression of GRK2 or its kinase-dead mutant in vivo, we demonstrate that GRK2 inhibits CFA-induced hyperalgesia in a kinase activity-dependent manner. In vitro, GRK2 inhibits Epac1-to-Rap1 signaling by phosphorylation of Epac1 at Ser-108 in the Disheveled/Egl-10/pleckstrin domain. This phosphorylation event inhibits agonist-induced translocation of Epac1 to the plasma membrane, thereby reducing Rap1 activation. Finally, we show that GRK2 inhibits Epac1-mediated sensitization of the mechanosensor Piezo2 and that Piezo2 contributes to inflammatory mechanical hyperalgesia. Collectively, these findings identify a key role of Epac1 in chronic inflammatory pain and a molecular mechanism for controlling Epac1 activity and chronic pain through phosphorylation of Epac1 at Ser-108. Importantly, using the Epac inhibitor ESI-09, we validate Epac1 as a potential therapeutic target for chronic pain.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/physiology , Guanine Nucleotide Exchange Factors/physiology , Hyperalgesia/physiopathology , Inflammation/complications , Nociception/physiology , Pain/physiopathology , Amino Acid Sequence , Animals , Chronic Disease , Freund's Adjuvant/toxicity , Ganglia, Spinal/physiopathology , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , Hyperalgesia/etiology , Inflammation/chemically induced , Ion Channels/physiology , Mechanoreceptors/physiology , Mice , Mice, Knockout , Molecular Sequence Data , Nerve Tissue Proteins/physiology , Pain/etiology , Pain Threshold/physiology , Phosphorylation , Phosphoserine/metabolism , Protein Interaction Mapping , Protein Processing, Post-Translational , Protein Structure, Tertiary , Signal Transduction , rap1 GTP-Binding Proteins/physiology
11.
Biochim Biophys Acta ; 1863(4): 544-61, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26704387

ABSTRACT

Exocytosis of the sperm's single secretory granule, or acrosome, is a regulated exocytosis triggered by components of the egg's investments. In addition to external calcium, sperm exocytosis (termed the acrosome reaction) requires cAMP synthesized endogenously and calcium mobilized from the acrosome through IP3-sensitive channels. The relevant cAMP target is Epac. In the first part of this paper, we present a novel tool (the TAT-cAMP sponge) to investigate cAMP-related signaling pathways in response to progesterone as acrosome reaction trigger. The TAT-cAMP sponge consists of the cAMP-binding sites of protein kinase A regulatory subunit RIß fused to the protein transduction domain TAT of the human immunodeficiency virus-1. The sponge permeated into sperm, sequestered endogenous cAMP, and blocked exocytosis. Progesterone increased the population of sperm with Rap1-GTP, Rab3-GTP, and Rab27-GTP in the acrosomal region; pretreatment with the TAT-cAMP sponge prevented the activation of all three GTPases. In the second part of this manuscript, we show that phospholipase Cε (PLCε) is required for the acrosome reaction downstream of Rap1 and upstream of intra-acrosomal calcium mobilization. Last, we present direct evidence that cAMP, Epac, Rap1, and PLCε are necessary for calcium mobilization from sperm's secretory granule. In summary, we describe here a pathway that connects cAMP to calcium mobilization from the acrosome during sperm exocytosis. Never before had direct evidence for each step of the cascade been put together in the same study.


Subject(s)
Acrosome/metabolism , Calcium/metabolism , Cyclic AMP/metabolism , Spermatozoa/metabolism , Cyclic AMP/physiology , Exocytosis/genetics , Exocytosis/physiology , Guanine Nucleotide Exchange Factors/metabolism , Guanine Nucleotide Exchange Factors/physiology , Humans , Inositol Phosphates/metabolism , Inositol Phosphates/physiology , Male , Phosphoinositide Phospholipase C/metabolism , Phosphoinositide Phospholipase C/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Transfection , rap1 GTP-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/physiology
13.
Thromb Haemost ; 114(6): 1241-9, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26311310

ABSTRACT

In proliferative retinopathies, like proliferative diabetic retinopathy and retinopathy of prematurity (ROP), the hypoxia response is sustained by the failure of the retina to revascularise its ischaemic areas. Non-resolving retina ischaemia/hypoxia results in upregulation of pro-angiogenic factors and pathologic neovascularisation with ectopic, fragile neovessels. Promoting revascularisation of the retinal avascular area could interfere with this vicious cycle and lead to vessel normalisation. Here, we examined the function of endothelial junctional adhesion molecule-C (JAM-C) in the context of ROP. Endothelial-specific JAM-C-deficient (EC-JAM-C KO) mice and littermate JAM-C-proficient (EC-JAM-C WT) mice were subjected to the ROP model. An increase in total retinal vascularisation was found at p17 owing to endothelial JAM-C deficiency, which was the result of enhanced revascularisation and vessel normalisation, thereby leading to significantly reduced avascular area in EC-JAM-C KO mice. In contrast, pathologic neovessel formation was not affected by endothelial JAM-C deficiency. Consistent with improved vessel normalisation, tip cell formation at the interface between vascular and avascular area was higher in EC-JAM-C KO mice, as compared to their littermate controls. Consistently, JAM-C inactivation in endothelial cells resulted in increased spreading on fibronectin and enhanced sprouting in vitro in a manner dependent on ß1-integrin and on the activation of the small GTPase RAP1. Together, endothelial deletion of JAM-C promoted endothelial cell sprouting, and consequently vessel normalisation and revascularisation of the hypoxic retina without altering pathologic neovascularisation. Thus, targeting endothelial JAM-C may provide a novel therapeutic strategy for promoting revascularisation and vessel normalisation in the treatment of proliferative retinopathies.


Subject(s)
Endothelium, Vascular/physiopathology , Junctional Adhesion Molecule C/deficiency , Neovascularization, Pathologic/physiopathology , Retinal Vessels/physiopathology , Retinopathy of Prematurity/physiopathology , Vitreoretinopathy, Proliferative/physiopathology , Animals , Cell Adhesion , Cell Hypoxia , Cell Line , Cell Size , Cell Surface Extensions , Disease Models, Animal , Endothelial Cells , Endothelium, Vascular/pathology , Fibronectins , Human Umbilical Vein Endothelial Cells , Humans , Integrin beta1/physiology , Ischemia/physiopathology , Junctional Adhesion Molecule C/physiology , Mice , Mice, Knockout , Neovascularization, Pathologic/etiology , Organ Specificity , Platelet Endothelial Cell Adhesion Molecule-1/analysis , RNA Interference , RNA, Small Interfering/genetics , Retinal Vessels/ultrastructure , rap1 GTP-Binding Proteins/physiology
14.
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
15.
Circ Res ; 116(5): 827-35, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25520364

ABSTRACT

RATIONALE: Chemokine-controlled arterial leukocyte recruitment is a crucial process in atherosclerosis. Formyl peptide receptor 2 (FPR2) is a chemoattractant receptor that recognizes proinflammatory and proresolving ligands. The contribution of FPR2 and its proresolving ligand annexin A1 to atherosclerotic lesion formation is largely undefined. OBJECTIVE: Because of the ambivalence of FPR2 ligands, we here investigate the role of FPR2 and its resolving ligand annexin A1 in atherogenesis. METHODS AND RESULTS: Deletion of FPR2 or its ligand annexin A1 enhances atherosclerotic lesion formation, arterial myeloid cell adhesion, and recruitment. Mechanistically, we identify annexin A1 as an endogenous inhibitor of integrin activation evoked by the chemokines CCL5, CCL2, and CXCL1. Specifically, the annexin A1 fragment Ac2-26 counteracts conformational activation and clustering of integrins on myeloid cells evoked by CCL5, CCL2, and CXCL1 through inhibiting activation of the small GTPase Rap1. In vivo administration of Ac2-26 largely diminishes arterial recruitment of myeloid cells in a FPR2-dependent fashion. This effect is also observed in the presence of selective antagonists to CCR5, CCR2, or CXCR2, whereas Ac2-26 was without effect when all 3 chemokine receptors were antagonized simultaneously. Finally, repeated treatment with Ac2-26 reduces atherosclerotic lesion sizes and lesional macrophage accumulation. CONCLUSIONS: Instructing the annexin A1-FPR2 axis harbors a novel approach to target arterial leukocyte recruitment. With the ability of Ac2-26 to counteract integrin activation exerted by various chemokines, delivery of Ac2-26 may be superior in inhibition of arterial leukocyte recruitment when compared with blocking individual chemokine receptors.


Subject(s)
Annexin A1/physiology , Aortic Diseases/etiology , Atherosclerosis/etiology , Animals , Annexin A1/deficiency , Annexin A1/genetics , Annexin A1/pharmacology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Chemokine CCL2/physiology , Chemokine CCL5/physiology , Chemokine CXCL1/physiology , Chemotaxis/drug effects , Dietary Fats/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/physiology , Peptides/pharmacology , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR5/physiology , Receptors, Formyl Peptide/deficiency , Receptors, Formyl Peptide/physiology , Receptors, Interleukin-8B/antagonists & inhibitors , rap1 GTP-Binding Proteins/physiology
16.
Nat Commun ; 5: 4657, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25118589

ABSTRACT

Host immune cells can detect and destruct invading pathogens via pattern-recognition receptors. Small Rap GTPases act as conserved molecular switches coupling extracellular signals to various cellular responses, but their roles as regulators in Toll-like receptor (TLR) signalling have not been fully elucidated. Here we report that Ras guanine nucleotide-releasing protein 3 (RasGRP3), a guanine nucleotide-exchange factor activating Ras and Rap1, limits production of proinflammatory cytokines (especially IL-6) in macrophages by activating Rap1 on activation by low levels of TLR agonists. We demonstrate that RasGRP3, a dominant member of RasGRPs in macrophages, impairs TLR3/4/9-induced IL-6 production and relieves dextrane sulphate sodium-induced colitis and collagen-induced arthritis. In RasGRP3-deficient RAW264.7 cells obtained by CRISPR-Cas9 genome editing, TLR3/4/9-induced activation of Rap1 was inhibited while ERK1/2 activation was enhanced. Our study suggests that RasGRP3 limits inflammatory response by activating Rap1 on low-intensity pathogen infection, setting a threshold for preventing excessive inflammatory response.


Subject(s)
GTP Phosphohydrolases/physiology , Inflammation/physiopathology , Macrophages/physiology , Toll-Like Receptors/physiology , rap1 GTP-Binding Proteins/physiology , ras Guanine Nucleotide Exchange Factors/physiology , Animals , Arthritis, Experimental/pathology , Arthritis, Experimental/physiopathology , Cells, Cultured , Disease Models, Animal , Inflammation/pathology , Interleukin-6/physiology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/physiology , Toll-Like Receptor 3/physiology , Toll-Like Receptor 4/physiology , Toll-Like Receptor 9/physiology , ras Guanine Nucleotide Exchange Factors/deficiency , ras Guanine Nucleotide Exchange Factors/genetics
17.
Curr Biol ; 23(18): 1825-33, 2013 Sep 23.
Article in English | MEDLINE | ID: mdl-24012314

ABSTRACT

The establishment of a multicellular body plan requires coordinating changes in cell adhesion and the cytoskeleton to ensure proper cell shape and position within a tissue. Cell adhesion to the extracellular matrix (ECM) via integrins plays diverse, essential roles during animal embryogenesis and therefore must be precisely regulated. Talin, a FERM-domain containing protein, forms a direct link between integrin adhesion receptors and the actin cytoskeleton and is an important regulator of integrin function. Similar to other FERM proteins, talin makes an intramolecular interaction that could autoinhibit its activity. However, the functional consequence of such an interaction has not been previously explored in vivo. Here, we demonstrate that targeted disruption of talin autoinhibition gives rise to morphogenetic defects during fly development and specifically that dorsal closure (DC), a process that resembles wound healing, is delayed. Impairment of autoinhibition leads to reduced talin turnover at and increased talin and integrin recruitment to sites of integrin-ECM attachment. Finally, we present evidence that talin autoinhibition is regulated by Rap1-dependent signaling. Based on our data, we propose that talin autoinhibition provides a switch for modulating adhesion turnover and adhesion stability that is essential for morphogenesis.


Subject(s)
Drosophila/growth & development , Morphogenesis/genetics , Talin/genetics , Animals , Drosophila/embryology , Drosophila/genetics , Gene Expression Regulation, Developmental , Larva/genetics , Larva/growth & development , Mutation , Signal Transduction , Talin/physiology , rap1 GTP-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/physiology
18.
Proc Natl Acad Sci U S A ; 110(28): 11427-32, 2013 Jul 09.
Article in English | MEDLINE | ID: mdl-23798437

ABSTRACT

Rap1 is a small GTPase regulating cell-cell adhesion, cell-matrix adhesion, and actin rearrangements, all processes dynamically coordinated during cell spreading and endothelial barrier function. Here, we identify the adaptor protein ras-interacting protein 1 (Rasip1) as a Rap1-effector involved in cell spreading and endothelial barrier function. Using Förster resonance energy transfer, we show that Rasip1 interacts with active Rap1 in a cellular context. Rasip1 mediates Rap1-induced cell spreading through its interaction partner Rho GTPase-activating protein 29 (ArhGAP29), a GTPase activating protein for Rho proteins. Accordingly, the Rap1-Rasip1 complex induces cell spreading by inhibiting Rho signaling. The Rasip1-ArhGAP29 pathway also functions in Rap1-mediated regulation of endothelial junctions, which controls endothelial barrier function. In this process, Rasip1 cooperates with its close relative ras-association and dilute domain-containing protein (Radil) to inhibit Rho-mediated stress fiber formation and induces junctional tightening. These results reveal an effector pathway for Rap1 in the modulation of Rho signaling and actin dynamics, through which Rap1 modulates endothelial barrier function.


Subject(s)
Endothelium, Vascular/physiology , GTPase-Activating Proteins/physiology , Intracellular Signaling Peptides and Proteins/physiology , rap1 GTP-Binding Proteins/physiology , Cells, Cultured , Endothelium, Vascular/cytology , Humans , Protein Binding , Signal Transduction
19.
Cell Mol Life Sci ; 70(13): 2395-410, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23420480

ABSTRACT

Phagocytosis mediated by the complement receptor CR3 (also known as integrin αMß2 or Mac-1) is regulated by the recruitment of talin to the cytoplasmic tail of the ß2 integrin subunit. Talin recruitment to this integrin is dependent on Rap1 activation. However, the mechanism by which Rap1 regulates this event and CR3-dependent phagocytosis remains largely unknown. In the present work, we examined the role of the Rap1 effector RIAM, a talin-binding protein, in the regulation of complement-mediated phagocytosis. Using the human myeloid cell lines HL-60 and THP-1, we determined that knockdown of RIAM impaired αMß2 integrin affinity changes induced by stimuli fMLP and LPS. Phagocytosis of complement-opsonized RBC particles, but not of IgG-opsonized RBC particles, was impaired in RIAM knockdown cells. Rap1 activation via EPAC induced by 8-pCPT-2'-O-Me-cAMP resulted in an increase of complement-mediated phagocytosis that was abrogated by knockdown of RIAM in HL-60 and THP-1 cell lines and in macrophages derived from primary monocytes. Furthermore, recruitment of talin to ß2 integrin during complement-mediated phagocytosis was reduced in RIAM knockdown cells. These results indicate that RIAM is a critical component of the phagocytosis machinery downstream of Rap1 and mediates its function by recruiting talin to the phagocytic complement receptors.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Membrane Proteins/physiology , Phagocytosis/physiology , Adaptor Proteins, Signal Transducing/metabolism , CD18 Antigens/metabolism , CD18 Antigens/physiology , Cells, Cultured , Complement System Proteins/physiology , Gene Knockdown Techniques , HL-60 Cells , Humans , Macrophage-1 Antigen/physiology , Macrophages/cytology , Macrophages/metabolism , Membrane Proteins/metabolism , Models, Biological , Neutrophils/cytology , Neutrophils/metabolism , Talin/metabolism , Talin/physiology , rap1 GTP-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/physiology
20.
Neurochem Int ; 62(6): 848-53, 2013 May.
Article in English | MEDLINE | ID: mdl-23416045

ABSTRACT

Rap1 signaling is important for migration, differentiation, axonal growth, and during neuronal polarity. Rap1 can be activated by external stimuli, which in turn regulates specific guanine nucleotide exchange factors such as C3G, among others. Cdk5 functions are also important to neuronal migration and differentiation. Since we found that pharmacological inhibition of Cdk5 by using roscovitine reduced Rap1 protein levels in COS-7 cells and also C3G contains three putative phosphorylation sites for Cdk5, we examined whether the Cdk5-dependent phosphorylation of C3G could affect Rap1 expression and activity. We co-transfected C3G and tet-OFF system for p35 over-expression, an activator of Cdk5 activity into COS-7 cells, and then we evaluated phosphorylation in serine residues in C3G by immunoprecipitation and Western blot. We found that p35 over-expression increased C3G-serine-phosphorylation while inhibition of p35 expression by tetracycline or inhibition of Cdk5 activity with roscovitine decreased it. Interestingly, we found that MG-132, a proteasome inhibitor, rescue Rap1 protein levels in the presence of roscovitine. Besides, C3G-serine-phosphorylation and Rap1 protein levels were reduced in brain from Cdk5(-/-) as compared with the Cdk5(+/+) brain. Finally, we found that p35 over-expression increased Rap1 activity while inhibition of p35 expression by tetracycline or roscovitine decreased Rap1 activity. These results suggest that Cdk5-mediated serine-phosphorylation of C3G may control Rap1 stability and activity, and this may potentially impact various neuronal functions such as migration, differentiation, and polarity.


Subject(s)
Cyclin-Dependent Kinase 5/physiology , rap1 GTP-Binding Proteins/physiology , Amino Acid Sequence , Animals , Blotting, Western , COS Cells , Chlorocebus aethiops , Cysteine Proteinase Inhibitors , Guanine Nucleotide-Releasing Factor 2/physiology , Immunoprecipitation , Interleukin-12 Subunit p35/biosynthesis , Leupeptins/pharmacology , Membrane Fusion Proteins , Mice , Mice, Knockout , Molecular Sequence Data , Neurons/drug effects , Phosphorylation , Real-Time Polymerase Chain Reaction , Tetracycline/pharmacology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...