Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 117(50): 31993-32004, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33262282

RESUMEN

Effective cancer prevention requires the discovery and intervention of a factor critical to cancer development. Here we show that ovarian progesterone is a crucial endogenous factor inducing the development of primary tumors progressing to metastatic ovarian cancer in a mouse model of high-grade serous carcinoma (HGSC), the most common and deadliest ovarian cancer type. Blocking progesterone signaling by the pharmacologic inhibitor mifepristone or by genetic deletion of the progesterone receptor (PR) effectively suppressed HGSC development and its peritoneal metastases. Strikingly, mifepristone treatment profoundly improved mouse survival (∼18 human years). Hence, targeting progesterone/PR signaling could offer an effective chemopreventive strategy, particularly in high-risk populations of women carrying a deleterious mutation in the BRCA gene.


Asunto(s)
Proteína BRCA1/genética , Cistadenocarcinoma Seroso/prevención & control , Mifepristona/farmacología , Neoplasias Ováricas/prevención & control , Progesterona/antagonistas & inhibidores , Adulto , Animales , Mama/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/prevención & control , Cistadenocarcinoma Seroso/química , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patología , Modelos Animales de Enfermedad , Estradiol/administración & dosificación , Femenino , Humanos , Ratones , Persona de Mediana Edad , Mifepristona/uso terapéutico , Mutación , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neoplasias Experimentales/prevención & control , Neoplasias Ováricas/inducido químicamente , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Ovario/patología , Ovario/cirugía , Progesterona/administración & dosificación , Progesterona/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Salpingooforectomía , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
2.
Elife ; 72018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29360039

RESUMEN

Specific cell shapes are fundamental to the organization and function of multicellular organisms. Fibroblast Growth Factor (FGF) signaling induces the elongation of lens fiber cells during vertebrate lens development. Nonetheless, exactly how this extracellular FGF signal is transmitted to the cytoskeletal network has previously not been determined. Here, we show that the Crk family of adaptor proteins, Crk and Crkl, are required for mouse lens morphogenesis but not differentiation. Genetic ablation and epistasis experiments demonstrated that Crk and Crkl play overlapping roles downstream of FGF signaling in order to regulate lens fiber cell elongation. Upon FGF stimulation, Crk proteins were found to interact with Frs2, Shp2 and Grb2. The loss of Crk proteins was partially compensated for by the activation of Ras and Rac signaling. These results reveal that Crk proteins are important partners of the Frs2/Shp2/Grb2 complex in mediating FGF signaling, specifically promoting cell shape changes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Forma de la Célula , Factores de Crecimiento de Fibroblastos/metabolismo , Fibroblastos/fisiología , Cristalino/embriología , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-crk/metabolismo , Transducción de Señal , Animales , Fibroblastos/efectos de los fármacos , Proteína Adaptadora GRB2/metabolismo , Ratones , Morfogénesis , Unión Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo
3.
PLoS One ; 11(12): e0168501, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28002430

RESUMEN

Several genetic conditions can lead to left ventricular hypertrophy (LVH). Among them, hypertrophic cardiomyopathy (HCM), caused by mutations in sarcomere genes, is the most common inherited cardiac disease. Instead, RASopathies, a rare class of disorders characterized by neuro-cardio-facial-cutaneous abnormalities and sometimes presenting with LVH, are caused by mutations in the RAS-MAPK pathway. We report on a 62-years-old male who presented isolated severe obstructive LVH but did not carry the sarcomere mutation previously identified in his affected relatives. By exome sequencing, we detected a novel mutation in HRAS gene (NM_005343.2:p.Arg68Trp), present also in the proband's daughter, who showed mild LVH and severe intellectual disability. The cardiac phenotype was indistinguishable between family members carrying either mutation. In silico studies suggested that the mutated HRAS protein is constitutionally activated. Consistently, functional characterization in vitro confirmed elevated HRAS-GTP accumulation and downstream RAS-MAPK pathway activation that are known to drive cell proliferation in LVH. Our study emphasizes the role of RAS signaling in cardiac hypertrophy and highlights the complexity in differential diagnosis of RASopathies. In fact, the mild features of RASopathy and the recurrence of sarcomeric HCM in this family delayed the correct diagnosis until comprehensive genetic testing was performed.


Asunto(s)
Miosinas Cardíacas/genética , Hipertrofia Ventricular Izquierda/genética , Cadenas Pesadas de Miosina/genética , Proteínas ras/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Miosinas Cardíacas/química , Análisis Mutacional de ADN , Femenino , Genotipo , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hipertrofia Ventricular Izquierda/patología , Masculino , Persona de Mediana Edad , Simulación de Dinámica Molecular , Miocardio/patología , Cadenas Pesadas de Miosina/química , Linaje , Polimorfismo de Nucleótido Simple , Estructura Terciaria de Proteína , Proteínas ras/química , Proteínas ras/metabolismo
4.
Mol Ther Methods Clin Dev ; 3: 16056, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27606349

RESUMEN

To test the hypothesis that increased Rap1a activity specifically in retinal pigment epithelial cells resists choroidal neovascularization (CNV), self-complementary adeno-associated virus 2 (scAAV2) with RPE65-promoter-driven GFP vectors were generated and introduced subretinally into Rap1b-deficient mice. Six-week-old mice that received subretinal control (scAAV2-Con) or constitutively active Rap1a (scAAV2-CARap1a) showed strong GFP at the 5 × 10(8) viral particle/µl dose 5 weeks later without altering retinal morphology or function. Compared to scAAV2-Con- or phosphate-buffered saline (PBS)-injected, eyes injected with scAAV2-CARap1a had increased Rap1 in retinal pigment epithelial (RPE)/choroidal lysates and a significant reduction in CNV volume 7 days after laser, comparable to eyes that received intravitreal anti-VEGF versus IgG control. scAAV2-CARap1a-, but not anti-VEGF-, injected eyes had increased pan-cadherin in RPE/choroids. In cultured RPE cells, increased active Rap1a inhibited TNFα-induced disassociation of junctional pan-cadherin/ß-catenin complexes, increased transepithelial electrical resistance through an interaction of ß-catenin with phosphorylated scaffold protein, IQGAP1, and inhibited choroidal endothelial cell (CEC) transmigration of an RPE monolayer. This evidence shows that increased Rap1a activity specifically in RPE cells is sufficient to reduce CEC transmigration and CNV and involves IQGAP1-mediated protection of RPE junctional complexes.

5.
PLoS One ; 10(12): e0145689, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26714318

RESUMEN

BACKGROUND: Small GTPase Rap1 has been implicated in a number of basic cellular functions, including cell-cell and cell-matrix adhesion, proliferation and regulation of polarity. Evolutionarily conserved, Rap1 has been studied in model organisms: yeast, Drosophila and mice. Mouse in vivo studies implicate Rap1 in the control of multiple stem cell, leukocyte and vascular cell functions. In vitro, several Rap1 effectors and regulatory mechanisms have been proposed. In particular, Rap1 has been implicated in maintaining epithelial and endothelial cell junction integrity and linked with cerebral cavernous malformations. RATIONALE: How Rap1 signaling network controls mammalian development is not clear. As a first step in addressing this question, we present phenotypes of murine total and vascular-specific Rap1a, Rap1b and double Rap1a and Rap1b (Rap1) knockout (KO) mice. RESULTS AND CONCLUSIONS: The majority of total Rap1 KO mice die before E10.5, consistent with the critical role of Rap1 in epithelial morphogenesis. At that time point, about 50% of Tie2-double Rap1 KOs appear grossly normal and develop normal vasculature, while the remaining 50% suffer tissue degeneration and show vascular abnormalities, including hemorrhages and engorgement of perineural vessels, albeit with normal branchial arches. However, no Tie2-double Rap1 KO embryos are present at E15.5, with hemorrhages a likely cause of death. Therefore, at least one Rap1 allele is required for development prior to the formation of the vascular system; and in endothelium-for the life-supporting function of the vasculature.


Asunto(s)
Neovascularización Fisiológica , Proteínas de Unión al GTP rap/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Animales , Embrión de Mamíferos/fisiología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Técnicas de Inactivación de Genes , Hemorragia/enzimología , Proteína KRIT1 , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Fenotipo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rap/deficiencia , Proteínas de Unión al GTP rap/genética , Proteínas de Unión al GTP rap1/deficiencia , Proteínas de Unión al GTP rap1/genética
6.
Biochim Biophys Acta ; 1852(5): 778-91, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25545047

RESUMEN

Protective effects of prostacyclin (PC) or its stable analog beraprost against agonist-induced lung vascular inflammation have been associated with elevation of intracellular cAMP and Rac GTPase signaling which inhibited the RhoA GTPase-dependent pathway of endothelial barrier dysfunction. This study investigated a distinct mechanism of PC-stimulated lung vascular endothelial (EC) barrier recovery and resolution of LPS-induced inflammation mediated by small GTPase Rap1. Efficient barrier recovery was observed in LPS-challenged pulmonary EC after prostacyclin administration even after 15 h of initial inflammatory insult and was accompanied by the significant attenuation of p38 MAP kinase and NFκB signaling and decreased production of IL-8 and soluble ICAM1. These effects were reproduced in cells post-treated with 8CPT, a small molecule activator of Rap1-specific nucleotide exchange factor Epac. By contrast, pharmacologic Epac inhibitor, Rap1 knockdown, or knockdown of cell junction-associated Rap1 effector afadin attenuated EC recovery caused by PC or 8CPT post-treatment. The key role of Rap1 in lung barrier restoration was further confirmed in the murine model of LPS-induced acute lung injury. Lung injury was monitored by measurements of bronchoalveolar lavage protein content, cell count, and Evans blue extravasation and live imaging of vascular leak over 6 days using a fluorescent tracer. The data showed significant acceleration of lung recovery by PC and 8CPT post-treatment, which was abrogated in Rap1a(-/-) mice. These results suggest that post-treatment with PC triggers the Epac/Rap1/afadin-dependent mechanism of endothelial barrier restoration and downregulation of p38MAPK and NFκB inflammatory cascades, altogether leading to accelerated lung recovery.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Endotelio Vascular/efectos de los fármacos , Epoprostenol/farmacología , Proteínas de Unión al GTP rap1/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Animales , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Cultivadas , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Epoprostenol/análogos & derivados , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Immunoblotting , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-8/metabolismo , Lipopolisacáridos , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , FN-kappa B/metabolismo , Inhibidores de Agregación Plaquetaria/farmacología , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas de Unión al GTP rap1/genética
7.
J Biol Chem ; 289(25): 17689-98, 2014 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-24790104

RESUMEN

Rap1 is a Ras family GTPase with a well documented role in ERK/MAP kinase signaling and integrin activation. Stimulation of the G-protein-coupled receptor PAR-1 with thrombin in human 1321N1 glioblastoma cells led to a robust increase in Rap1 activation. This response was sustained for up to 6 h and mediated through RhoA and phospholipase D (PLD). Thrombin treatment also induced a 5-fold increase in cell adhesion to fibronectin, which was blocked by down-regulating PLD or Rap1A or by treatment with a ß1 integrin neutralizing antibody. In addition, thrombin treatment led to increases in phospho-focal adhesion kinase (tyrosine 397), ERK1/2 phosphorylation and cell proliferation, which were significantly inhibited in cells treated with ß1 integrin antibody or Rap1A siRNA. To assess the role of Rap1A in tumor formation in vivo, we compared growth of 1321N1 cells stably expressing control, Rap1A or Rap1B shRNA in a mouse xenograft model. Deletion of Rap1A, but not of Rap1B, reduced tumor mass by >70% relative to control. Similar observations were made with U373MG glioblastoma cells in which Rap1A was down-regulated. Collectively, these findings implicate a Rap1A/ß1 integrin pathway, activated downstream of G-protein-coupled receptor stimulation and RhoA, in glioblastoma cell proliferation. Moreover, our data demonstrate a critical role for Rap1A in glioblastoma tumor growth in vivo.


Asunto(s)
Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , Integrina beta1/metabolismo , Proteínas de Neoplasias/metabolismo , Trombina/metabolismo , Proteínas de Unión al GTP rap1/biosíntesis , Animales , Línea Celular Tumoral , Glioblastoma/genética , Xenoinjertos , Humanos , Integrina beta1/genética , Ratones , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Trombina/genética , Proteínas de Unión al GTP rap1/genética , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
8.
FASEB J ; 28(1): 265-74, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24043260

RESUMEN

Activation of Rap1 GTPase can improve the integrity of the barrier of the retina pigment epithelium (RPE) and reduce choroidal neovascularization (CNV). Inhibition of NADPH oxidase activation also reduces CNV. We hypothesize that Rap1 inhibits NADPH oxidase-generated ROS and thereby reduces CNV formation. Using a murine model of laser-induced CNV, we determined that reduced Rap1 activity in RPE/choroid occurred with CNV formation and that activation of Rap1 by 2'-O-Me-cAMP (8CPT)-reduced laser-induced CNV via inhibiting NADPH oxidase-generated ROS. In RPE, inhibition of Rap1 by Rap1 GTPase-activating protein (Rap1GAP) increased ROS generation, whereas activation of Rap1 by 8CPT reduced ROS by interfering with the assembly of NADPH oxidase membrane subunit p22phox with NOX4 or cytoplasmic subunit p47phox. Activation of NADPH oxidase with Rap1GAP reduced RPE barrier integrity via cadherin phosphorylation and facilitated choroidal EC migration across the RPE monolayer. Rap1GAP-induced ROS generation was inhibited by active Rap1a, but not Rap1b, and activation of Rap1a by 8CPT in Rap1b(-/-) mice reduced laser-induced CNV, in correlation with decreased ROS generation in RPE/choroid. These findings provide evidence that active Rap1 reduces CNV by interfering with the assembly of NADPH oxidase subunits and increasing the integrity of the RPE barrier.


Asunto(s)
Neovascularización Coroidal/metabolismo , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Animales , Ratones , Ratones Noqueados , Oxidación-Reducción
9.
PLoS One ; 8(9): e73070, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24039860

RESUMEN

Loss of barrier integrity precedes the development of pathologies such as metastasis, inflammatory disorders, and blood-retinal barrier breakdown present in neovascular age-related macular degeneration. Rap1 GTPase is involved in regulating both endothelial and epithelial cell junctions; the specific role of Rap1A vs. Rap1B isoforms is less clear. Compromise of retinal pigment epithelium barrier function is a contributing factor to the development of AMD. We utilized shRNA of Rap1 isoforms in cultured human retinal pigment epithelial cells, along with knockout mouse models to test the role of Rap1 on promoting RPE barrier properties, with emphasis on the dynamic junctional regulation that is triggered when the adhesion between cells is challenged. In vitro, Rap1A shRNA reduced steady-state barrier integrity, whereas Rap1B shRNA affected dynamic junctional responses. In a laser-induced choroidal neovascularization (CNV) model of macular degeneration, Rap1b(-/-) mice exhibited larger CNV volumes compared to wild-type or Rap1a(-/-) . In vivo, intravitreal injection of a cAMP analog (8CPT-2'-O-Me-cAMP) that is a known Rap1 activator significantly reduced laser-induced CNV volume, which correlated with the inhibition of CEC transmigration across 8CPT-2'O-Me-cAMP-treated RPE monolayers in vitro. Rap1 activation by 8CPT-2'-O-Me-cAMP treatment increased recruitment of junctional proteins and F-actin to cell-cell contacts, increasing both the linearity of junctions in vitro and in cells surrounding laser-induced lesions in vivo. We conclude that in vitro, Rap1A may be important for steady state barrier integrity, while Rap1B is involved more in dynamic junctional responses such as resistance to junctional disassembly induced by EGTA and reassembly of cell junctions following disruption. Furthermore, activation of Rap1 in vivo inhibited development of choroidal neovascular lesions in a laser-injury model. Our data suggest that targeting Rap1 isoforms in vivo with 8CPT-2'-O-Me-cAMP may be a viable pharmacological means to strengthen the RPE barrier against the pathological choroidal endothelial cell invasion that occurs in macular degeneration.


Asunto(s)
Barrera Hematorretinal/metabolismo , Neovascularización Coroidal/metabolismo , Degeneración Macular/metabolismo , Degeneración Macular/patología , Proteínas de Unión al GTP rap1/metabolismo , Animales , Neovascularización Coroidal/genética , AMP Cíclico/metabolismo , AMP Cíclico/farmacología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Activación Enzimática , Humanos , Uniones Intercelulares/metabolismo , Degeneración Macular/genética , Ratones , Ratones Noqueados , Migración Transendotelial y Transepitelial/efectos de los fármacos , Proteínas de Unión al GTP rap1/genética
10.
PLoS One ; 8(4): e60226, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23565202

RESUMEN

Tumor inflammation, the recruitment of myeloid lineage cells into the tumor microenvironment, promotes angiogenesis, immunosuppression and metastasis. CD11b+Gr1lo monocytic lineage cells and CD11b+Gr1hi granulocytic lineage cells are recruited from the circulation by tumor-derived chemoattractants, which stimulate PI3-kinase γ (PI3Kγ)-mediated integrin α4 activation and extravasation. We show here that PI3Kγ activates PLCγ, leading to RasGrp/CalDAG-GEF-I&II mediated, Rap1a-dependent activation of integrin α4ß1, extravasation of monocytes and granulocytes, and inflammation-associated tumor progression. Genetic depletion of PLCγ, CalDAG-GEFI or II, Rap1a, or the Rap1 effector RIAM was sufficient to prevent integrin α4 activation by chemoattractants or activated PI3Kγ (p110γCAAX), while activated Rap (RapV12) promoted constitutive integrin activation and cell adhesion that could only be blocked by inhibition of RIAM or integrin α4ß1. Similar to blockade of PI3Kγ or integrin α4ß1, blockade of Rap1a suppressed both the recruitment of monocytes and granulocytes to tumors and tumor progression. These results demonstrate critical roles for a PI3Kγ-Rap1a-dependent pathway in integrin activation during tumor inflammation and suggest novel avenues for cancer therapy.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Inflamación/metabolismo , Integrina alfa4beta1/metabolismo , Células Mieloides/metabolismo , Neoplasias/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Animales , Femenino , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Inflamación/genética , Integrina alfa4beta1/química , Ligandos , Ratones , Ratones Noqueados , Neoplasias/genética , Fosfolipasa C gamma/metabolismo , Unión Proteica , Conformación Proteica
11.
J Biol Chem ; 288(22): 15687-98, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23612979

RESUMEN

Many factors, including duration and intensity of the unfolded protein response (UPR), dictate whether cells will adapt to endoplasmic reticulum stress or undergo apoptosis. In tuberous sclerosis (TSC), elevation of mammalian target of rapamycin complex 1 (mTORC1) activity has been proposed to compound the induction of UPR transcription factors ATF4 and CHOP, suggesting that the UPR could be targeted to eradicate TSC1/2-null cells during patient therapy. Here we report that control of c-MYC translation by mTORC1 plays a key role in determining whether TSC2-null Elt3 rat leiomyoma cells apoptose in response to UPR induction by the proteasome inhibitor bortezomib. Although bortezomib induces eukaryotic initiating factor 2α phosphorylation, mTORC1 activity was also required for downstream induction of the UPR transcription factors ATF4 and CHOP by a mechanism involving increased expression of c-MYC. Although bortezomib-induced c-MYC transcription was resistant to rapamycin treatment, mTORC1 activity was required for efficient c-MYC translation. c-MYC subsequently bound to the ATF4 promoter, suggesting direct involvement of an mTORC1/c-MYC-driven signaling pathway in the activation of the UPR. Consistent with this notion, exogenously expressed c-MYC reversed the ability of rapamycin to prevent bortezomib-induced CHOP and ATF4 expression as well as apoptosis. These findings indicate that the induction of ATF4/CHOP expression occurs via mTORC1 regulation of c-MYC and that this signaling pathway is a major determinant in the ability of bortezomib to induce apoptosis.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ácidos Borónicos/farmacología , Biosíntesis de Proteínas/fisiología , Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Pirazinas/farmacología , Proteínas Supresoras de Tumor , Respuesta de Proteína Desplegada/efectos de los fármacos , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Animales , Apoptosis/fisiología , Bortezomib , Línea Celular Tumoral , Regulación de la Expresión Génica , Humanos , Inmunosupresores/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos , Proteínas/genética , Proteínas Proto-Oncogénicas c-myc/genética , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteína 2 del Complejo de la Esclerosis Tuberosa , Respuesta de Proteína Desplegada/fisiología
12.
J Immunol ; 189(8): 3800-4, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22984083

RESUMEN

Phagocyte NADPH oxidase plays a key role in pathogen clearance via reactive oxygen species (ROS) production. Defects in oxidase function result in chronic granulomatous disease with hallmark recurrent microbial infections and inflammation. The oxidase's role in the adaptive immune response is not well understood. Class II presentation of cytoplasmic and exogenous Ag to CD4(+) T cells was impaired in human B cells with reduced oxidase p40(phox) subunit expression. Naturally arising mutations, which compromise p40(phox) function in a chronic granulomatous disease patient, also perturbed class II Ag presentation and intracellular ROS production. Reconstitution of patient B cells with a wild-type, but not a mutant, p40(phox) allele restored exogenous Ag presentation and intracellular ROS generation. Remarkably, class II presentation of epitopes from membrane Ag was robust in p40(phox)-deficient B cells. These studies reveal a role for NADPH oxidase and p40(phox) in skewing epitope selection and T cell recognition of self Ag.


Asunto(s)
Presentación de Antígeno/inmunología , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , Antígenos HLA-DR/metabolismo , NADPH Oxidasas/fisiología , Presentación de Antígeno/genética , Subgrupos de Linfocitos B/enzimología , Línea Celular Transformada , Humanos , Líquido Intracelular/enzimología , Líquido Intracelular/inmunología , Líquido Intracelular/metabolismo , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Fosfoproteínas/biosíntesis , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
13.
Am J Physiol Renal Physiol ; 303(6): F831-44, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22791333

RESUMEN

Tuberous sclerosis complex (TSC), an inherited tumor predisposition syndrome associated with mutations in TSC1 or TSC2, affects ∼1 in 6,000 individuals. Eighty percent of TSC patients develop renal angiomyolipomas, and renal involvement is a major contributor to patient morbidity and mortality. Recent work has shown that mammalian target of rapamycin complex 1 (mTORC1) inhibition caused angiomyolipoma shrinkage but that this treatment may cause cytostatic not a cytotoxic effect. Endoplasmic reticulum (ER) stress can develop in TSC-associated cells due to mTORC1-driven protein translation. We hypothesized that renal angiomyolipoma cells experience ER stress that can be leveraged to result in targeted cytotoxicity. We used immortalized human angiomyolipoma cells stably transfected with empty vector or TSC2 (encoding tuberin). Using cell number quantification and cell death assays, we found that mTORC1 inhibition with RAD001 suppressed angiomyolipoma cell proliferation in a cytostatic manner. Angiomyolipoma cells exhibited enhanced sensitivity to proteasome inhibitor-induced ER stress compared with TSC2-rescued cells. After proteasome inhibition with MG-132, Western blot analyses showed greater induction of C/EBP-homologous protein (CHOP) and more poly (ADP-ribose) polymerase (PARP) and caspase-3 cleavage, supporting ER stress-induced apoptosis. Live cell numbers also were decreased and cell death increased by MG-132 in angiomyolipoma cells compared with TSC2 rescued. Intriguingly, while pretreatment of angiomyolipoma cells with RAD001 attenuated CHOP and BiP induction, apoptotic markers cleaved PARP and caspase-3 and eukaryotic translation initiation factor 2α phosphorylation were increased, along with evidence of increased autophagy. These results suggest that human angiomyolipoma cells are uniquely susceptible to agents that exacerbate ER stress and that additional synergy may be achievable with targeted combination therapy.


Asunto(s)
Angiomiolipoma/metabolismo , Estrés del Retículo Endoplásmico , Neoplasias Renales/metabolismo , Esclerosis Tuberosa/complicaciones , Proteínas Supresoras de Tumor/metabolismo , Angiomiolipoma/etiología , Angiomiolipoma/genética , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Caspasa 3/metabolismo , Línea Celular Tumoral , Factor 2 Eucariótico de Iniciación/metabolismo , Everolimus , Humanos , Inmunosupresores/farmacología , Neoplasias Renales/etiología , Neoplasias Renales/genética , Leupeptinas/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos , Fosforilación , Poli(ADP-Ribosa) Polimerasas/metabolismo , Inhibidores de Proteasoma/farmacología , Proteínas/metabolismo , Sirolimus/análogos & derivados , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Factor de Transcripción CHOP/metabolismo , Transfección , Esclerosis Tuberosa/genética , Esclerosis Tuberosa/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética
14.
Am J Physiol Cell Physiol ; 303(5): C499-511, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22621783

RESUMEN

Intracellular signaling by the second messenger cyclic AMP (cAMP) activates the Ras-related small GTPase Rap1 through the guanine exchange factor Epac. This activation leads to effector protein interactions, activation, and biological responses in the vasculature, including vasorelaxation. In vascular smooth muscle cells derived from human dermal arterioles (microVSM), Rap1 selectively regulates expression of G protein-coupled α(2C)-adrenoceptors (α(2C)-ARs) through JNK-c-jun nuclear signaling. The α(2C)-ARs are generally retained in the trans-Golgi compartment and mobilize to the cell surface and elicit vasoconstriction in response to cellular stress. The present study used human microVSM to examine the role of Rap1 in receptor localization. Complementary approaches included murine microVSM derived from tail arteries of C57BL6 mice that express functional α(2C)-ARs and mice deficient in Rap1A (Rap1A-null). In human microVSM, increasing intracellular cAMP by direct activation of adenylyl cyclase by forskolin (10 µM) or selectively activating Epac-Rap signaling by the cAMP analog 8-pCPT-2'-O-Me-cAMP (100 µM) activated RhoA, increased α(2C)-AR expression, and reorganized the actin cytoskeleton, increasing F-actin. The α(2C)-ARs mobilized from the perinuclear region to intracellular filamentous structures and to the plasma membrane. Similar results were obtained in murine wild-type microVSM, coupling Rap1-Rho-actin dynamics to receptor relocalization. This signaling was impaired in Rap1A-null murine microVSM and was rescued by delivery of constitutively active (CA) mutant of Rap1A. When tested in heterologous HEK293 cells, Rap1A-CA or Rho-kinase (ROCK-CA) caused translocation of functional α(2C)-ARs to the cell surface (~4- to 6-fold increase, respectively). Together, these studies support vascular bed-specific physiological role of Rap1 and suggest a role in vasoconstriction in microVSM.


Asunto(s)
AMP Cíclico/metabolismo , Miocitos del Músculo Liso/metabolismo , Transporte de Proteínas/fisiología , Receptores Adrenérgicos alfa 2/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Arteriolas/citología , Células Cultivadas , AMP Cíclico/genética , Regulación de la Expresión Génica/fisiología , Humanos , Ratones , Ratones Noqueados , Unión Proteica , Receptores Adrenérgicos alfa 2/genética , Proteínas de Unión al GTP rap1/genética , Proteína de Unión al GTP rhoA/genética
15.
J Cell Biochem ; 113(4): 1253-64, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22121046

RESUMEN

Constitutive activation of M-Ras has previously been reported to cause morphologic and growth transformation of murine cells, suggesting that M-Ras plays a role in tumorigenesis. Cell transformation by M-Ras correlated with weak activation of the Raf/MEK/ERK pathway, although contributions from other downstream effectors were suggested. Recent studies indicate that signaling events distinct from the Raf/MEK/ERK cascade are critical for human tumorigenesis. However, it is unknown what signaling events M-Ras triggers in human cells. Using constitutively active M-Ras (Q71L) containing additional mutations within its effector-binding loop, we found that M-Ras induces MEK/ERK-dependent and -independent Elk1 activation as well as phosphatidylinositol 3 kinase (PI3K)/Akt and JNK/cJun activation in human MCF-7 breast cancer cells. Among several human cell lines examined, M-Ras-induced MEK/ERK-independent Elk1 activation was only detected in MCF-7 cells, and correlated with Rlf/M-Ras interaction and Ral/JNK activation. Supporting a role for M-Ras signaling in breast cancer, EGF activated M-Ras and promoted its interaction with endogenous Rlf. In addition, constitutive activation of M-Ras induced estrogen-independent growth of MCF-7 cells that was dependent on PI3K/Akt, MEK/ERK, and JNK activation. Thus, our studies demonstrate that M-Ras signaling activity differs between human cells, highlighting the importance of defining Ras protein signaling within each cell type, especially when designing treatments for Ras-induced cancer. These findings also demonstrate that M-Ras activity may be important for progression of EGFR-dependent tumors.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , MAP Quinasa Quinasa 4/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas de Unión al GTP Monoméricas/fisiología , Proteínas de Unión al GTP ral/metabolismo , Línea Celular Tumoral , Activación Enzimática , Femenino , Humanos , Inmunoprecipitación , Transducción de Señal
16.
J Biol Chem ; 286(49): 42316-42324, 2011 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-22009749

RESUMEN

Phosphatases of the regenerating liver (PRL) play oncogenic roles in cancer development and metastasis. Although previous studies indicate that PRL-1 promotes cell growth and migration by activating both the ERK1/2 and RhoA pathways, the mechanism by which it activates these signaling events remains unclear. We have identified a PRL-1-binding peptide (Peptide 1) that shares high sequence identity with a conserved motif in the Src homology 3 (SH3) domain of p115 Rho GTPase-activating protein (GAP). p115 RhoGAP directly binds PRL-1 in vitro and in cells via its SH3 domain. Structural analyses of the PRL-1·Peptide 1 complex revealed a novel protein-protein interaction whereby a sequence motif within the PxxP ligand-binding site of the p115 RhoGAP SH3 domain occupies a folded groove within PRL-1. This prevents the canonical interaction between the SH3 domain of p115 RhoGAP and MEKK1 and results in activation of ERK1/2. Furthermore, PRL-1 binding activates RhoA signaling by inhibiting the catalytic activity of p115 RhoGAP. The results demonstrate that PRL-1 binding to p115 RhoGAP provides a coordinated mechanism underlying ERK1/2 and RhoA activation.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Regulación Enzimológica de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/química , Proteínas Inmediatas-Precoces/metabolismo , Proteínas de la Membrana/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Familia-src Quinasas/metabolismo , Secuencias de Aminoácidos , Animales , Fibroblastos/metabolismo , Células HEK293 , Humanos , Ligandos , Ratones , Unión Proteica , Factores de Intercambio de Guanina Nucleótido Rho , Transducción de Señal , Proteínas de Unión al GTP rho/metabolismo
17.
Am J Physiol Endocrinol Metab ; 301(6): E1072-80, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21828338

RESUMEN

Second-phase insulin release requires the sustained mobilization of insulin granules from internal storage pools to the cell surface for fusion with the plasma membrane. However, the detailed mechanisms underlying this process remain largely unknown. GTP-loading of the small GTPase Cdc42 is the first glucose-specific activation step in the process, although how glucose triggers Cdc42 activation is entirely unknown. In a directed candidate screen for guanine nucleotide exchange factors (GEFs), which directly activate small GTPases, Cool-1/ßPix was identified in pancreatic islet beta cells. In support of its role as the beta cell Cdc42 GEF, ßPix coimmunoprecipitated with Cdc42 in human islets and MIN6 beta cells in a glucose-dependent manner, peaking just prior to Cdc42 activation. Furthermore, RNAi-mediated ßPix reduction by 50% corresponded to full ablation of glucose-induced Cdc42 activation and significant attenuation of basal and glucose-stimulated insulin secretion. Of the two Cdc42 guanine nucleotide dissociation inhibitor (GDI) proteins identified in beta cells, ßPix competed selectively with caveolin-1 (Cav-1) but not RhoGDI in coimmunoprecipitation and GST-Cdc42-GDP interaction assays. However, a phospho-deficient Cav-1-Y14F mutant failed to compete with ßPix; Cav-1(Tyr14) is an established phosphorylation site for Src kinase. Taken together, these data support a new model, wherein glucose stimulates Cav-1 and induces its dissociation from Cdc42, possibly via Src kinase activation to phosphorylate Cav-1(Tyr14), to promote Cdc42-ßPix binding and Cdc42 activation, and to trigger downstream signaling and ultimately sustain insulin release.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/fisiología , Insulina/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Animales , Caveolina 1/metabolismo , Células Cultivadas , Técnicas de Silenciamiento del Gen , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , ARN Interferente Pequeño/farmacología , Factores de Intercambio de Guanina Nucleótido Rho , Estudios de Validación como Asunto , Proteína de Unión al GTP cdc42/genética
18.
Curr Drug Targets ; 12(8): 1223-31, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21561413

RESUMEN

mTOR exists in two distinct complexes. mTOR complex 1 (mTORC1) is potently inhibited by the immunosupressive macrolide rapamycin; whereas, mTORC2 is insensitive to this durg. These mTOR complexes play an integral role in the regulation of many cellular processes including protein synthesis, autophagy, lipid synthesis, mitochondrial metabolism/biogenesis, and cell cycle. Both mTOR complexes are important for maintaining cellular homeostasis and the growth of many types of cancer. Rapamycin and rapalogs have been effective in treating only a small number of these cancers, and other methods are being developed in order to address the short-comings of these drugs. The most direct of these approaches include ATP-competitive inhibitors of the mTOR kinase or dual inhibitors of both mTOR and PI3 kinase. However, other methods of inhibiting mTORC1 may prove clinically useful as well. These include amino acid depletion using asparaginase and inhibition of the Rheb GTPases with farnesyl transferase inhibitors or statins. Most excitingly, mTORC1 activation has been shown to cause and sensitize cells to DNA damage and ER stress. Many of the drugs currently used in the clinic for the treatment of cancer cause these types of stress, and existing drugs may be tailored to treat tumors with high mTORC1 activity.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Terapia Molecular Dirigida , Proteínas de Unión al GTP Monoméricas/fisiología , Neoplasias/tratamiento farmacológico , Neuropéptidos/fisiología , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/fisiología , Descubrimiento de Drogas , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias/metabolismo , Proteínas Quinasas/genética , Proteínas Quinasas/fisiología , Proteína Homóloga de Ras Enriquecida en el Cerebro , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología
19.
J Exp Med ; 207(9): 1923-38, 2010 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-20733035

RESUMEN

Rap1 GTPases control immune synapse formation and signaling in lymphocytes. However, the precise molecular mechanism by which Rap1 regulates natural killer (NK) cell activation is not known. Using Rap1a or Rap1b knockout mice, we identify Rap1b as the major isoform in NK cells. Its absence significantly impaired LFA1 polarization, spreading, and microtubule organizing center (MTOC) formation in NK cells. Neither Rap1 isoform was essential for NK cytotoxicity. However, absence of Rap1b impaired NKG2D, Ly49D, and NCR1-mediated cytokine and chemokine production. Upon activation, Rap1b colocalized with the scaffolding protein IQGAP1. This interaction facilitated sequential phosphorylation of B-Raf, C-Raf, and ERK1/2 and helped IQGAP1 to form a large signalosome in the perinuclear region. These results reveal a previously unrecognized role for Rap1b in NK cell signaling and effector functions.


Asunto(s)
Células Asesinas Naturales/inmunología , Transducción de Señal , Proteínas de Unión al GTP rap/inmunología , Proteínas Activadoras de ras GTPasa/inmunología , Animales , Movimiento Celular , Polaridad Celular , Células Cultivadas , Citotoxicidad Inmunológica , Células Asesinas Naturales/metabolismo , Antígeno-1 Asociado a Función de Linfocito/inmunología , Ratones , Ratones Noqueados , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Proteínas de Unión al GTP rap/deficiencia , Proteínas de Unión al GTP rap/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo
20.
Int J Biochem Cell Biol ; 42(6): 965-74, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20184965

RESUMEN

Atheroma formation and restenosis following percutaneous vascular intervention involve the growth and migration of vascular smooth muscle cells (SMCs) into neointimal lesions, in part due to changes in the extracellular matrix. While some clinical studies have suggested that, in comparison to non-diabetics, beta3 integrin inhibition in diabetic patients confers protection from restenosis, little is known regarding the role of beta3 integrin inhibition on SMC responses in this context. To understand the molecular mechanisms underlying integrin-mediated regulation of SMC function in diabetes, we examined SMC responses in diabetic mice deficient in integrin beta3 and observed that the integrin was required for enhanced proliferation, migration and extracellular regulated kinase (ERK) activation. Hyperglycemia-enhanced membrane recruitment and catalytic activity of PKCbeta in an integrin beta3-dependent manner. Hyperglycemia also promoted SMC filopodia formation and cell migration, both of which required alphaVbeta3, PKCbeta, and ERK activity. Furthermore, the integrin-kinase association was regulated by the alphaVbeta3 integrin ligand thrombospondin and the integrin modulator Rap1 under conditions of hyperglycemia. These results suggest that there are differences in SMC responses to vascular injury depending on the presence or absence of hyperglycemia and that SMC response under hyperglycemic conditions is largely mediated through beta3 integrin signaling.


Asunto(s)
Aterosclerosis/metabolismo , Diabetes Mellitus Experimental/metabolismo , Angiopatías Diabéticas , Integrina alfaVbeta3/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Endotelio Vascular/lesiones , Endotelio Vascular/patología , Humanos , Hiperglucemia , Ratones , Ratones Noqueados , Miocitos del Músculo Liso/patología , Proteína Quinasa C/metabolismo , Proteína Quinasa C beta , Transducción de Señal/efectos de los fármacos , Trombospondinas/farmacología , Cicatrización de Heridas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA