Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
1.
Blood ; 136(10): 1180-1190, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32518959

RESUMEN

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.


Asunto(s)
Integrina beta1/metabolismo , Integrina beta3/metabolismo , Mutación Puntual , Talina/fisiología , Trombopoyesis , Proteínas de Unión al GTP rap/fisiología , Proteínas de Unión al GTP rap1/fisiología , Animales , Femenino , Integrina beta1/genética , Integrina beta3/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Activación Plaquetaria , Agregación Plaquetaria , Dominios Proteicos , Transducción de Señal
2.
Mol Psychiatry ; 26(7): 3223-3239, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-32651478

RESUMEN

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.


Asunto(s)
Plasticidad Neuronal , Corteza Prefrontal/fisiopatología , Estrés Psicológico/enzimología , Proteínas de Unión al GTP rap1/fisiología , Animales , Ratones , Neuronas , Sinapsis
3.
PLoS Genet ; 14(1): e1007195, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29381707

RESUMEN

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.


Asunto(s)
Permeabilidad Capilar/genética , Adhesión Celular/genética , Células Endoteliales/fisiología , Endotelio Vascular/metabolismo , Proteínas Activadoras de GTPasa/fisiología , Proteínas de Unión al GTP rap1/fisiología , Animales , Animales Modificados Genéticamente , Células Cultivadas , Embrión de Mamíferos , Embrión no Mamífero , Femenino , Proteínas Activadoras de GTPasa/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Megacariocitos/fisiología , Ratones , Ratones Noqueados , Transducción de Señal , Pez Cebra , Proteínas de Unión al GTP rap1/genética
4.
Proc Natl Acad Sci U S A ; 115(16): E3722-E3730, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29602807

RESUMEN

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.


Asunto(s)
Dictyostelium/fisiología , Proteínas Protozoarias/fisiología , Transducción de Señal/fisiología , Citoesqueleto de Actina/fisiología , Citoesqueleto de Actina/ultraestructura , Adhesión Celular , Movimiento Celular , Forma de la Célula , Quimiotaxis , Dictyostelium/genética , Dictyostelium/ultraestructura , Activación Enzimática , Microscopía Fluorescente , Microscopía de Contraste de Fase , Mutación Missense , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/fisiología , Fenotipo , Proteínas Protozoarias/genética , Proteínas Recombinantes/metabolismo , Proteínas de Unión al GTP rap1/genética , Proteínas de Unión al GTP rap1/fisiología
5.
Proc Natl Acad Sci U S A ; 113(11): 3036-41, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26929333

RESUMEN

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.


Asunto(s)
Quinasa 2 del Receptor Acoplado a Proteína-G/fisiología , Factores de Intercambio de Guanina Nucleótido/fisiología , Hiperalgesia/fisiopatología , Inflamación/complicaciones , Nocicepción/fisiología , Dolor/fisiopatología , Secuencia de Aminoácidos , Animales , Enfermedad Crónica , Adyuvante de Freund/toxicidad , Ganglios Espinales/fisiopatología , Factores de Intercambio de Guanina Nucleótido/deficiencia , Factores de Intercambio de Guanina Nucleótido/genética , Hiperalgesia/etiología , Inflamación/inducido químicamente , Canales Iónicos/fisiología , Mecanorreceptores/fisiología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/fisiología , Dolor/etiología , Umbral del Dolor/fisiología , Fosforilación , Fosfoserina/metabolismo , Mapeo de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Transducción de Señal , Proteínas de Unión al GTP rap1/fisiología
6.
Int J Mol Sci ; 19(10)2018 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-30241315

RESUMEN

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.


Asunto(s)
Proteínas de Unión al GTP rap1/fisiología , Inmunidad Adaptativa , Diferenciación Celular , Transformación Celular Neoplásica , Modelos Moleculares , Prenilación , Procesamiento Proteico-Postraduccional , Transducción de Señal , Proteínas de Unión al GTP rap1/química , Proteínas de Unión al GTP rap1/metabolismo
7.
Biochim Biophys Acta ; 1863(4): 544-61, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26704387

RESUMEN

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.


Asunto(s)
Acrosoma/metabolismo , Calcio/metabolismo , AMP Cíclico/metabolismo , Espermatozoides/metabolismo , AMP Cíclico/fisiología , Exocitosis/genética , Exocitosis/fisiología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Factores de Intercambio de Guanina Nucleótido/fisiología , Humanos , Fosfatos de Inositol/metabolismo , Fosfatos de Inositol/fisiología , Masculino , Fosfoinositido Fosfolipasa C/metabolismo , Fosfoinositido Fosfolipasa C/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología , Transfección , Proteínas de Unión al GTP rap1/metabolismo , Proteínas de Unión al GTP rap1/fisiología
8.
Circ Res ; 116(5): 827-35, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25520364

RESUMEN

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.


Asunto(s)
Anexina A1/fisiología , Enfermedades de la Aorta/etiología , Aterosclerosis/etiología , Animales , Anexina A1/deficiencia , Anexina A1/genética , Anexina A1/farmacología , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/prevención & control , Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Aterosclerosis/patología , Aterosclerosis/prevención & control , Quimiocina CCL2/fisiología , Quimiocina CCL5/fisiología , Quimiocina CXCL1/fisiología , Quimiotaxis/efectos de los fármacos , Grasas de la Dieta/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/fisiología , Péptidos/farmacología , Receptores CCR2/antagonistas & inhibidores , Receptores CCR5/fisiología , Receptores de Formil Péptido/deficiencia , Receptores de Formil Péptido/fisiología , Receptores de Interleucina-8B/antagonistas & inhibidores , Proteínas de Unión al GTP rap1/fisiología
9.
Proc Natl Acad Sci U S A ; 110(28): 11427-32, 2013 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-23798437

RESUMEN

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.


Asunto(s)
Endotelio Vascular/fisiología , Proteínas Activadoras de GTPasa/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas de Unión al GTP rap1/fisiología , Células Cultivadas , Endotelio Vascular/citología , Humanos , Unión Proteica , Transducción de Señal
10.
Circ Res ; 110(5): 716-26, 2012 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-22282193

RESUMEN

RATIONALE: Vascular endothelial growth factor (VEGF), a major proangiogenic agent, exerts its proangiogenic action by binding to VEGF receptor 2 (VEGFR2), the activity of which is regulated by direct interactions with other cell surface proteins, including integrin α(V)ß(3). However, how the interaction between VEGFR2 and integrin α(V)ß(3) is regulated is not clear. OBJECTIVE: To investigate whether Necl-5/poliovirus receptor, an immunoglobulin-like molecule that is known to bind integrin α(V)ß(3), regulates the interaction between VEGFR2 and integrin α(V)ß(3), and to clarify the role of Necl-5 in the VEGF-induced angiogenesis. METHODS AND RESULTS: Necl-5-knockout mice displayed no obvious defect in vascular development; however, recovery of blood flow after hindlimb ischemia and the VEGF-induced neovascularization in implanted Matrigel plugs were impaired in Necl-5-knockout mice. To clarify the mechanism of the regulation of angiogenesis by Necl-5, we investigated the roles of Necl-5 in the VEGF-induced angiogenic responses in vitro. Knockdown of Necl-5 by siRNAs in human umbilical vein endothelial cells (HUVECs) inhibited the VEGF-induced capillary-like network formation on Matrigel, migration, and proliferation, and conversely, enhanced apoptosis. Coimmunoprecipitation assays showed the interaction of Necl-5 with VEGFR2, and knockdown of Necl-5 prevented the VEGF-induced interaction of integrin α(V)ß(3) with VEGFR2. Knockdown of Necl-5 suppressed the VEGFR2-mediated activation of downstream proangiogenic and survival signals, including Rap1, Akt, and endothelial nitric oxide synthase. CONCLUSIONS: These results demonstrate the critical role of Necl-5 in angiogenesis and suggest that Necl-5 may regulate the VEGF-induced angiogenesis by controlling the interaction of VEGFR2 with integrin α(v)ß(3), and the VEGFR2-mediated Rap1-Akt signaling pathway.


Asunto(s)
Antígenos de Neoplasias/fisiología , Moléculas de Adhesión Celular/fisiología , Endotelio Vascular/fisiología , Proteínas de Neoplasias/fisiología , Neovascularización Fisiológica/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología , Animales , Antígenos de Neoplasias/genética , Apoptosis/efectos de los fármacos , Moléculas de Adhesión Celular/deficiencia , Moléculas de Adhesión Celular/genética , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Humanos , Integrina alfaVbeta3/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Modelos Animales , Proteínas de Neoplasias/deficiencia , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogénicas c-akt/fisiología , ARN Interferente Pequeño/farmacología , Receptores Virales/efectos de los fármacos , Receptores Virales/fisiología , Transducción de Señal/fisiología , Proteínas de Unión al GTP rap1/fisiología
11.
Cell Mol Life Sci ; 70(13): 2395-410, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23420480

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas de la Membrana/fisiología , Fagocitosis/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antígenos CD18/metabolismo , Antígenos CD18/fisiología , Células Cultivadas , Proteínas del Sistema Complemento/fisiología , Técnicas de Silenciamiento del Gen , Células HL-60 , Humanos , Antígeno de Macrófago-1/fisiología , Macrófagos/citología , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Neutrófilos/citología , Neutrófilos/metabolismo , Talina/metabolismo , Talina/fisiología , Proteínas de Unión al GTP rap1/metabolismo , Proteínas de Unión al GTP rap1/fisiología
12.
J Am Soc Nephrol ; 22(5): 859-72, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21493776

RESUMEN

Renal ischemia-reperfusion injury is associated with the loss of tubular epithelial cell-cell and cell-matrix interactions which contribute to renal failure. The Epac-Rap signaling pathway is a potent regulator of cell-cell and cell-matrix adhesion. The cyclic AMP analogue 8-pCPT-2'-O-Me-cAMP has been shown to selectively activate Epac, whereas the addition of an acetoxymethyl (AM) ester to 8-pCPT-2'-O-Me-cAMP enhanced in vitro cellular uptake. Here we demonstrate that pharmacological activation of Epac-Rap signaling using acetoxymethyl-8-pCPT-2'-O-Me-cAMP preserves cell adhesions during hypoxia in vitro, maintaining the barrier function of the epithelial monolayer. Intrarenal administration in vivo of 8-pCPT-2'-O-Me-cAMP also reduced renal failure in a mouse model for ischemia-reperfusion injury. This was accompanied by decreased expression of the tubular cell stress marker clusterin-α, and lateral expression of ß-catenin after ischemia indicative of sustained tubular barrier function. Our study emphasizes the undervalued importance of maintaining tubular epithelial cell adhesion in renal ischemia and demonstrates the potential of pharmacological modulation of cell adhesion as a new therapeutic strategy to reduce the extent of injury in kidney disease and transplantation.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/fisiología , Isquemia/complicaciones , Riñón/irrigación sanguínea , Insuficiencia Renal/etiología , Transducción de Señal/fisiología , Estrés Fisiológico , Proteínas de Unión al GTP rap1/fisiología , Uniones Adherentes/fisiología , Animales , Adhesión Celular , Hipoxia de la Célula , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Adhesiones Focales , Túbulos Renales Proximales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
13.
Am J Physiol Gastrointest Liver Physiol ; 301(2): G385-400, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21546580

RESUMEN

Cholestatic liver disorders are accompanied by the hepatic accumulation of cytotoxic bile acids that induce cell death. Increases in cAMP protect hepatocytes from bile acid-induced apoptosis by a cAMP-guanine exchange factor (cAMP-GEF)/phosphoinositide-3-kinase (PI3K)/Akt pathway. The aim of these studies was to identify the downstream substrate in this pathway and to determine at what level in the apoptotic cascade cytoprotection occurs. Since inhibitory phosphorylation of glycogen synthase kinase-3 (GSK) occurs downstream of PI3K/Akt and this phosphorylation has been implicated in cell survival, we conducted studies to determine whether GSK was downstream in cAMP-GEF/PI3K/Akt-mediated cytoprotection. Our results show that treatment of hepatocytes with the cAMP-GEF-specific analog, 4-(4-chlorophenylthio)-2'-O-methyladenosine-3',5'-cAMP, results in PI3K-dependent phosphorylation of GSK. Direct chemical inhibition of GSK in rat hepatocytes or human HUH7-NTCP cells with several structurally and functionally distinct inhibitors including bromoindirubin-3'-oxime (BIO), maleimides (SB216763, SB415286), thiadiazolidine derivatives, and LiCl attenuates apoptosis induced by glycochenodeoxycholate (GCDC). In addition, genetic silencing of the GSK ß isoform with small interfering RNA attenuates GCDC apoptosis in HUH7-NTCP cells. Adenoviral inhibition of the Rap1 blocks both cAMP-GEF-mediated cytoprotection against GCDC-induced apoptosis and Akt/GSK3ß phosphorylation. GCDC-induced phosphorylation of the proapoptotic kinase, c-Jun NH(2)-terminal kinase (JNK) is inhibited by GSK inhibition or cAMP-GEF activation. GCDC-induced apoptosis is accompanied by phosphorylation of the endoplasmic reticulum stress markers pIEF2α and IRE-1, and pretreatment with the cAMP-GEF analog or GSK inhibitors prevents this phosphorylation. Collectively, our results support the presence of a cAMP/cAMP-GEF/Rap1/PI3K/Akt/GSKß survival pathway in hepatocytes that inhibits bile acid-induced JNK phosphorylation.


Asunto(s)
Apoptosis/fisiología , Glucógeno Sintasa Quinasa 3/fisiología , Factores de Intercambio de Guanina Nucleótido/fisiología , Hepatocitos/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Transducción de Señal/fisiología , Animales , Ácidos y Sales Biliares/farmacología , Carcinoma Hepatocelular , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , AMP Cíclico/fisiología , Citoprotección , Retículo Endoplásmico/fisiología , Ácido Glicoquenodesoxicólico/fisiología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Masculino , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt , Ratas , Ratas Wistar , Estrés Fisiológico/fisiología , Tionucleótidos , Células Tumorales Cultivadas , Proteínas de Unión al GTP rap1/fisiología
14.
Proc Natl Acad Sci U S A ; 105(51): 20511-6, 2008 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-19075231

RESUMEN

Memory formation is highly sensitive to specific patterns of training, but the cellular and molecular mechanisms underlying pattern sensitivity are not well understood. We explored this general question by using Aplysia californica as a model system. We examined the regulation of MAPK (ERK1/2) activation by small G proteins in the CNS by using different patterns of analog stimuli that mimic different patterns of behavioral training for memory induction. We first cloned and characterized the Aplysia homologs of the small G proteins, Ras and Rap1 (ApRas and ApRap, respectively). We next examined changes in ApRas and ApRap activity that accompany MAPK activation. Last, by delivering recombinant ApRas and ApRap into the CNS, we directly manipulated their activity and examined the resultant MAPK activation. We found that MAPK activation induced by analog training depends on the combined activity of ApRas and ApRap, rather than the individual activity of either one alone. Also, ApRas and ApRap have a complex role in MAPK activation: they can act as activators or inhibitors, depending on the specific pattern of the training. The pattern-sensitive regulation of MAPK by interactive ApRas and ApRap activity that we have identified could contribute to the molecular routing of different downstream effects of spatially localized MAPK required for the induction of specific pattern-sensitive forms of synaptic facilitation and memory.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Memoria , Proteínas de Unión al GTP Monoméricas/fisiología , Animales , Aplysia/fisiología , Sistema Nervioso Central , Clonación Molecular , Datos de Secuencia Molecular , Proteínas de Unión al GTP rap1/genética , Proteínas de Unión al GTP rap1/fisiología , Proteínas ras/genética , Proteínas ras/fisiología
15.
Biochim Biophys Acta ; 1788(4): 790-6, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19159611

RESUMEN

Rap proteins are Ras-like small GTP-binding proteins that amongst others are involved in the control of cell-cell and cell-matrix adhesion. Several Rap guanine nucleotide exchange factors (RapGEFs) function to activate Rap. These multi-domain proteins, which include C3G, Epacs, PDZ-GEFs, RapGRPs and DOCK4, are regulated by various different stimuli and may function at different levels in junction formation. Downstream of Rap, a number of effector proteins have been implicated in junctional control, most notably the adaptor proteins AF6 and KRIT/CCM1. In this review, we will highlight the latest findings on the Rap signaling network in the control of epithelial and endothelial cell-cell junctions.


Asunto(s)
Uniones Adherentes/fisiología , Factores de Intercambio de Guanina Nucleótido/fisiología , Transducción de Señal/fisiología , Uniones Estrechas/fisiología , Proteínas de Unión al GTP rap1/fisiología , Animales , Células Endoteliales/ultraestructura , Humanos , Estructura Terciaria de Proteína
16.
Am J Physiol Heart Circ Physiol ; 298(1): H136-43, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19880670

RESUMEN

We have previously reported that 1) inhibition of cyclooxygenase-2 and PGE(2) production reduces hypertrophy after myocardial infarction in mice and 2) PGE(2) acting through its EP4 receptor causes hypertrophy of neonatal ventricular myocytes (NVMs) via ERK1/2. It is known that EP4 couples to adenylate cyclase, cAMP, and PKA. The present study was designed to determine interactions between the cAMP-PKA pathway and ERK1/2 and to further characterize events downstream of ERK1/2. We hypothesized that PKA and the small GTPase Rap are upstream of ERK1/2 and that 90-kDa ribosomal S6 kinase (p90RSK) is activated downstream. Treatment of NVMs with PGE(2) activated Rap, and this activation was inhibited in part by an EP4 antagonist and PKA inhibition. Transfection of a dominant negative mutant of Rap reduced PGE(2) activation of ERK1/2. PGE(2) activation of p90RSK was also dependent on EP4, PKA, and Rap. We also tested the involvement of Rap, ERK1/2, and p90RSK in PGE(2) regulation of gene expression. PGE(2) stimulation of brain natriuretic peptide promoter activity was blocked by either ERK1/2 inhibition or a dominant negative mutation of p90RSK. PGE(2) stimulation of c-Fos was dependent on EP4, PKA, ERK1/2, and p90RSK, whereas only the latter two kinases were involved in PGE(2) regulation of early growth response-1. Finally, we tested the involvement of EP4-dependent signaling in the NVM growth response and found that the overexpression of EP4 increased NVM cell size. We conclude that EP4-dependent signaling in NVMs in part involves PKA, Rap, ERK1/2, and p90RSK and results in the increased expression of brain natriuretic peptide and c-Fos.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Dinoprostona/fisiología , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Miocitos Cardíacos/fisiología , Receptores de Prostaglandina E/fisiología , Proteínas Quinasas S6 Ribosómicas/fisiología , Transducción de Señal/fisiología , Proteínas de Unión al GTP rap1/fisiología , Adenoviridae/genética , Adenoviridae/fisiología , Animales , Animales Recién Nacidos , Western Blotting , Tamaño de la Célula , Células Cultivadas , Dinoprostona/farmacología , Expresión Génica/fisiología , Ratones , Miocitos Cardíacos/ultraestructura , Ratas , Ratas Sprague-Dawley , Subtipo EP4 de Receptores de Prostaglandina E , Transfección
17.
Blood ; 111(9): 4627-36, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18319399

RESUMEN

B-cell development is orchestrated by complex signaling networks. Rap1 is a member of the Ras superfamily of small GTP-binding proteins and has 2 isoforms, Rap1a and Rap1b. Although Rap1 has been suggested to have an important role in a variety of cellular processes, no direct evidence demonstrates a role for Rap1 in B-cell biology. In this study, we found that Rap1b was the dominant isoform of Rap1 in B cells. We discovered that Rap1b deficiency in mice barely affected early development of B cells but markedly reduced marginal zone (MZ) B cells in the spleen and mature B cells in peripheral and mucosal lymph nodes. Rap1b-deficient B cells displayed normal survival and proliferation in vivo and in vitro. However, Rap1b-deficient B cells had impaired adhesion and reduced chemotaxis in vitro, and lessened homing to lymph nodes in vivo. Furthermore, we found that Rap1b deficiency had no marked effect on LPS-, BCR-, or SDF-1-induced activation of mitogen-activated protein kinases and AKT but clearly impaired SDF-1-mediated activation of Pyk-2, a key regulator of SDF-1-mediated B-cell migration. Thus, we have discovered a critical and distinct role of Rap1b in mature B-cell trafficking and development of MZ B cells.


Asunto(s)
Linfocitos B/fisiología , Quimiotaxis de Leucocito , Proteínas de Unión al GTP rap/fisiología , Proteínas de Unión al GTP rap1/fisiología , Animales , Linfocitos B/citología , Quimiocina CXCL12/metabolismo , Quinasa 2 de Adhesión Focal/metabolismo , Ganglios Linfáticos/citología , Ratones , Proteínas de Unión al GTP rap/deficiencia , Proteínas de Unión al GTP rap1/deficiencia
18.
Hum Reprod ; 25(9): 2229-38, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20663796

RESUMEN

BACKGROUND: The mononuclear villous cytotrophoblast (CTB) differentiates and fuses to the multinucleated syncytiotrophoblast (STB), which produces hCG and progesterone. cAMP-mediated intracellular pathways are involved in the process of endocrine differentiation and fusion (syncytialization). The exchange protein directly activated by cAMP (Epac) is a mediator of cAMP signaling. We examined the differential roles of Epac and protein kinase A (PKA) signaling in the cell fusion and differentiation of trophoblast-derived BeWo cells. METHODS: Epac1 and Epac2 were localized in human placental tissue (n = 9) by immunohistochemistry. The PKA-selective cAMP analog (N(6)-phenyl-cAMP, Phe) or Epac-selective cAMP analog (CPT) was tested for effects on hCG and progesterone production, and syncytialization in BeWo cells. The effect of knockdown of Epac or its downstream target molecule (Rap1) on syncytialization was evaluated. RESULTS: Epac1 and Epac2 proteins were expressed in villous CTB, STB, stroma, blood vessels and extravillous CTB of the placenta. Phe increased the expression of hCG alpha/beta mRNA and secretion of hCG protein in BeWo cells (P < 0.01 versus control). CPT-stimulated production of hCG (P < 0.05), albeit to a lesser extent than Phe. Progesterone production was also enhanced by Phe or CPT (P < 0.01 and P < 0.05, respectively). CPT or a stable cAMP analog (dibutyryl-cAMP: Db) increased the number of syncytialized BeWo cells (P < 0.01), whereas Phe did not stimulate fusion. CPT- or Db-induced syncytialization was observed, even in the presence of a PKA inhibitor. Knockdown of Epac1 or Rap1 repressed the Db-, CPT- or forskolin-induced cell fusion. CONCLUSIONS: The Epac signaling pathway may be associated with the cAMP-mediated functional differentiation and syncytialization of human trophoblasts.


Asunto(s)
Diferenciación Celular , Factores de Intercambio de Guanina Nucleótido/fisiología , Placenta/metabolismo , Transducción de Señal , Diferenciación Celular/efectos de los fármacos , Fusión Celular , Línea Celular , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Gonadotropina Coriónica/genética , Gonadotropina Coriónica/metabolismo , AMP Cíclico/análogos & derivados , AMP Cíclico/metabolismo , AMP Cíclico/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Especificidad de Órganos , Placenta/citología , Placenta/efectos de los fármacos , Embarazo , Trimestres del Embarazo , Progesterona/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Transducción de Señal/efectos de los fármacos , Trofoblastos/citología , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo , Proteínas de Unión al GTP rap1/genética , Proteínas de Unión al GTP rap1/fisiología
19.
Yakugaku Zasshi ; 130(11): 1413-20, 2010 Nov.
Artículo en Japonés | MEDLINE | ID: mdl-21048397

RESUMEN

Endothelial cells lining blood vessels are in tight contact with each other, thereby maintaining vascular integrity. Compromising vascular integrity leads to an increase in vascular permeability, which is associated with chronic inflammation, edema, and tumor angiogenesis. Vascular endothelial (VE)-cadherin is an endothelium-specific cell-cell adhesion molecule involved in endothelial barrier functions. We previously reported that cyclic AMP-elevating agonists such as prostaglandins and adrenomedullin potentiate VE-cadherin-dependent cell adhesion by inducing activation of Rap1 small GTPase through Epac. We further investigated the mechanism whereby Rap1 potentiates VE-cadherin-dependent cell adhesion, and found that Rap1 induces the formation of circumferential actin bundles along the cell-cell junctions. Although it has been believed that α-/ß-catenins anchor cadherin to the actin cytoskeleton to stabilize cadherin at cell-cell junctions (classical model), Nelson's and Weis' groups have recently suggested a new dynamic model in which α-/ß-catenins do not stably connect actin to cadherin. However, our study clearly indicated that the circumferential actin bundles anchor VE-cadherin to the cell-cell junctions through α-/ß-catenins. Thus Rap1 potentiates endothelial cell-cell junctions through the mechanism based on the static model.


Asunto(s)
Antígenos CD/fisiología , Cadherinas/fisiología , Moléculas de Adhesión Celular/fisiología , Adhesión Celular/fisiología , Células Endoteliales/fisiología , Uniones Intercelulares/fisiología , Transducción de Señal/fisiología , Proteínas de Unión al GTP rap1/fisiología , Actinas/metabolismo , Angiopoyetina 1/fisiología , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Permeabilidad Capilar , AMP Cíclico/fisiología , Citoesqueleto/metabolismo , Humanos , alfa Catenina/fisiología , beta Catenina/fisiología , Proteínas de Unión al GTP rap1/metabolismo
20.
Sci Rep ; 10(1): 13221, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32764635

RESUMEN

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).


Asunto(s)
Anticuerpos Monoclonales/inmunología , Integrinas/metabolismo , Proteínas de Unión al GTP rap1/fisiología , Animales , Línea Celular , Humanos , Integrinas/química , Integrinas/inmunología , Células Jurkat , Glicoproteínas de Membrana/química , Ratones , Péptidos/inmunología , Conformación Proteica , Dominios Proteicos , Ratas , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA