Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Oncogene ; 39(8): 1652-1664, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31740782

RESUMEN

Hepatocellular carcinoma (HCC) is the most common primary liver cancer, and the incidence of HCC is increasing. Recently, cancer immunotherapy has emerged as an efficient treatment against some cancers. Here we have used a mouse model of mutagen-induced HCC to explore the therapeutic usefulness of targeting the DNA-activated STING pathway in HCC. STING-deficient mice exhibited unaltered initial development of HCC, but had higher number of large tumors at late stages of disease. In the liver of STING-deficient HCC mice, we observed reduced levels of phospho-STAT1, autophagy, and cleaved caspase3. These responses were activated in the liver by treatment with a cyclic dinucleotide (CDN) STING agonist. Importantly, CDN treatment of mice after HCC development efficiently reduced tumor size. Initiation of CDN treatment at an even later stage of disease to allow HCC detection by MR scanning revealed that the majority of tumors regressed in response to CDN, but new tumors were also detected, which were unresponsive to CDN treatment. Overall, the modulation of the STING pathway affects the development of HCC, and holds promise for a use as a treatment of this disease, most likely in combination with other immunomodulatory treatments such as PD1 inhibitors or with standard of care.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Proteínas de la Membrana/metabolismo , Terapia Molecular Dirigida , Nucleotidiltransferasas/metabolismo , Animales , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Transformación Celular Neoplásica , Humanos , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Proteínas de la Membrana/agonistas , Ratones , Transducción de Señal/efectos de los fármacos , Carga Tumoral/efectos de los fármacos
2.
PLoS Pathog ; 14(4): e1006976, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29608601

RESUMEN

In recent years, there has been an increasing interest in immunomodulatory therapy as a means to treat various conditions, including infectious diseases. For instance, Toll-like receptor (TLR) agonists have been evaluated for treatment of genital herpes. However, although the TLR7 agonist imiquimod was shown to have antiviral activity in individual patients, no significant effects were observed in clinical trials, and the compound also exhibited significant side effects, including local inflammation. Cytosolic DNA is detected by the enzyme cyclic GMP-AMP (2'3'-cGAMP) synthase (cGAS) to stimulate antiviral pathways, mainly through induction of type I interferon (IFN)s. cGAS is activated upon DNA binding to produce the cyclic dinucleotide (CDN) 2'3'-cGAMP, which in turn binds and activates the adaptor protein Stimulator of interferon genes (STING), thus triggering type I IFN expression. In contrast to TLRs, STING is expressed broadly, including in epithelial cells. Here we report that natural and non-natural STING agonists strongly induce type I IFNs in human cells and in mice in vivo, without stimulating significant inflammatory gene expression. Systemic treatment with 2'3'-cGAMP reduced genital herpes simplex virus (HSV) 2 replication and improved the clinical outcome of infection. More importantly, local application of CDNs at the genital epithelial surface gave rise to local IFN activity, but only limited systemic responses, and this treatment conferred total protection against disease in both immunocompetent and immunocompromised mice. In direct comparison between CDNs and TLR agonists, only CDNs acted directly on epithelial cells, hence allowing a more rapid and IFN-focused immune response in the vaginal epithelium. Thus, specific activation of the STING pathway in the vagina evokes induction of the IFN system but limited inflammatory responses to allow control of HSV2 infections in vivo.


Asunto(s)
Antivirales/farmacología , Herpes Genital/prevención & control , Herpesvirus Humano 2/efectos de los fármacos , Interacciones Huésped-Patógeno/efectos de los fármacos , Proteínas de la Membrana/agonistas , Nucleótidos Cíclicos/farmacología , Animales , Células Cultivadas , Citosol/virología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/virología , Femenino , Herpes Genital/metabolismo , Herpes Genital/virología , Herpesvirus Humano 2/patogenicidad , Humanos , Interferón Tipo I/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Queratinocitos/virología , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Nucleotidiltransferasas/fisiología , Transducción de Señal
3.
Front Immunol ; 9: 687, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29696016

RESUMEN

The follicular (FO) versus marginal zone (MZ) B cell fate decision in the spleen depends upon BCR, BAFF, and Notch2 signaling. Whether or how Gi signaling affects this fate decision is unknown. Here, we show that direct contact with Notch ligand expressing stromal cells (OP9-Delta-like 1) cannot promote normal MZ B cell development when progenitor B cells lack Gαi proteins, or if Gi signaling is disabled. Consistent with faulty ADAM10-dependent Notch2 processing, Gαi-deficient transitional B cells had low ADAM10 membrane expression levels and reduced Notch2 target gene expression. Immunoblotting Gαi-deficient B cell lysates revealed a reduction in mature, processed ADAM10. Suggesting that Gαi signaling promotes ADAM10 membrane expression, stimulating normal transitional B cells with CXCL12 raised it, while inhibiting Gαi nucleotide exchange blocked its upregulation. Surprisingly, inhibiting Gαi nucleotide exchange in transitional B cells also impaired the upregulation of ADAM10 that occurs following antigen receptor crosslinking. These results indicate that Gαi signaling supports ADAM10 maturation and activity in transitional B cells, and ultimately Notch2 signaling to promote MZ B cell development.


Asunto(s)
Proteína ADAM10/fisiología , Secretasas de la Proteína Precursora del Amiloide/fisiología , Linfocitos B/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Proteínas de la Membrana/fisiología , Animales , Células Cultivadas , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Toxina del Pertussis/farmacología , Receptor Notch2/fisiología , Transducción de Señal , Bazo/citología
4.
J Med Chem ; 59(22): 10253-10267, 2016 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-27783523

RESUMEN

We describe novel STING-activating cyclic dinucleotides whose constituent nucleosides are adenosine and inosine and that vary by ribose substitution, internucleotide linkage position, and phosphate modification. In mammalian cells in vitro, some of these cAIMP analogs induce greater STING-dependent IRF and NF-κB pathway signaling than do the reference agonists for murine (DMXAA) or human (2',3'-cGAMP) STING. In human blood ex vivo, they induce type I interferons (IFNs) and proinflammatory cytokines: for the former, 3',3'-cAIMP (9; EC50 of 6.4 µM) and analogs 52-56 (EC50 of 0.4-4.7 µM), which contain one or two 2'-fluoro-2'-deoxyriboses and/or bis-phosphorothioate linkages, are more potent than 2',3'-cGAMP (EC50 of 19.6 µM). Interestingly, 9 induces type I IFNs more strongly than do its linkage isomers 2',3'-cAIMP (10), 3',2'-cAIMP (23), and 2',2'-cAIMP (27). Lastly, some of the cAIMP analogs are more resistant than 2',3'-cGAMP to enzymatic cleavage in vitro. We hope to exploit our findings to develop STING-targeted immunotherapies.


Asunto(s)
Adenosina/farmacología , IMP Cíclico/farmacología , Citocinas/metabolismo , Diseño de Fármacos , Interferón Tipo I/metabolismo , Adenosina/síntesis química , Adenosina/química , Animales , Línea Celular , IMP Cíclico/síntesis química , IMP Cíclico/química , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Estructura Molecular , Relación Estructura-Actividad
5.
Hypertension ; 68(6): 1365-1374, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27698068

RESUMEN

Hyperactivity of the renin-angiotensin-aldosterone system through the angiotensin II (Ang II)/Ang II type 1 receptor (AT1-R) axis constitutes a hallmark of hypertension. Recent findings indicate that only a subset of AT1-R signaling pathways is cardiodeleterious, and their selective inhibition by biased ligands promotes therapeutic benefit. To date, only synthetic biased ligands have been described, and whether natural renin-angiotensin-aldosterone system peptides exhibit functional selectivity at AT1-R remains unknown. In this study, we systematically determined efficacy and potency of Ang II, Ang III, Ang IV, and Ang-(1-7) in AT1-R-expressing HEK293T cells on the activation of cardiodeleterious G-proteins and cardioprotective ß-arrestin2. Ang III and Ang IV fully activate similar G-proteins than Ang II, the prototypical AT1-R agonist, despite weaker potency of Ang IV. Interestingly, Ang-(1-7) that binds AT1-R fails to promote G-protein activation but behaves as a competitive antagonist for Ang II/Gi and Ang II/Gq pathways. Conversely, all renin-angiotensin-aldosterone system peptides act as agonists on the AT1-R/ß-arrestin2 axis but display biased activities relative to Ang II as indicated by their differences in potency and AT1-R/ß-arrestin2 intracellular routing. Importantly, we reveal Ang-(1-7) a known Mas receptor-specific ligand, as an AT1-R-biased agonist, selectively promoting ß-arrestin activation while blocking the detrimental Ang II/AT1-R/Gq axis. This original pharmacological profile of Ang-(1-7) at AT1-R, similar to that of synthetic AT1-R-biased agonists, could, in part, contribute to its cardiovascular benefits. Accordingly, in vivo, Ang-(1-7) counteracts the phenylephrine-induced aorta contraction, which was blunted in AT1-R knockout mice. Collectively, these data suggest that Ang-(1-7) natural-biased agonism at AT1-R could fine-tune the physiology of the renin-angiotensin-aldosterone system.


Asunto(s)
Angiotensina II/farmacología , Angiotensina I/metabolismo , Cardiotónicos/metabolismo , Células HEK293/metabolismo , Fragmentos de Péptidos/metabolismo , Receptor de Angiotensina Tipo 2/metabolismo , Animales , Aorta Abdominal/efectos de los fármacos , Aorta Abdominal/fisiología , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Células HEK293/efectos de los fármacos , Humanos , Músculos , Fenilefrina/farmacología , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/fisiología , Sensibilidad y Especificidad , Transducción de Señal , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología , beta-Arrestinas/metabolismo
6.
Cell ; 166(4): 920-934, 2016 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-27499022

RESUMEN

Understanding how membrane nanoscale organization controls transmembrane receptors signaling activity remains a challenge. We studied interferon-γ receptor (IFN-γR) signaling in fibroblasts from homozygous patients with a T168N mutation in IFNGR2. By adding a neo-N-glycan on IFN-γR2 subunit, this mutation blocks IFN-γ activity by unknown mechanisms. We show that the lateral diffusion of IFN-γR2 is confined by sphingolipid/cholesterol nanodomains. In contrast, the IFN-γR2 T168N mutant diffusion is confined by distinct actin nanodomains where conformational changes required for Janus-activated tyrosine kinase/signal transducer and activator of transcription (JAK/STAT) activation by IFN-γ could not occur. Removing IFN-γR2 T168N-bound galectins restored lateral diffusion in lipid nanodomains and JAK/STAT signaling in patient cells, whereas adding galectins impaired these processes in control cells. These experiments prove the critical role of dynamic receptor interactions with actin and lipid nanodomains and reveal a new function for receptor glycosylation and galectins. Our study establishes the physiological relevance of membrane nanodomains in the control of transmembrane receptor signaling in vivo. VIDEO ABSTRACT.


Asunto(s)
Fibroblastos/metabolismo , Mutación Missense , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Transducción de Señal , Actinas/química , Actinas/metabolismo , Animales , Células COS , Membrana Celular/química , Membrana Celular/metabolismo , Chlorocebus aethiops , Difusión , Endocitosis , Activación Enzimática , Glicosilación , Humanos , Interferón gamma/metabolismo , Infecciones por Mycobacterium/genética , Infecciones por Mycobacterium/inmunología , Receptores de Interferón/química
7.
Front Pharmacol ; 6: 203, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26483685

RESUMEN

Cyclic adenosine 3',5'-monophosphate (cAMP) modulates a broad range of biological processes including the regulation of cardiac myocyte contractile function where it constitutes the main second messenger for ß-adrenergic receptors' signaling to fulfill positive chronotropic, inotropic and lusitropic effects. A growing number of studies pinpoint the role of spatial organization of the cAMP signaling as an essential mechanism to regulate cAMP outcomes in cardiac physiology. Here, we will briefly discuss the complexity of cAMP synthesis and degradation in the cardiac context, describe the way to detect it and review the main pharmacological arsenal to modulate its availability.

8.
J Immunol ; 195(5): 2090-102, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26232433

RESUMEN

Resistance to inhibitors of cholinesterase 8A (Ric-8A) is a highly evolutionarily conserved cytosolic protein initially identified in Caenorhabditis elegans, where it was assigned a regulatory role in asymmetric cell divisions. It functions as a guanine nucleotide exchange factor for Gαi, Gαq, and Gα12/13 and as a molecular chaperone required for the initial association of nascent Gα subunits with cellular membranes in embryonic stem cell lines. To test its role in hematopoiesis and B lymphocytes specifically, we generated ric8 (fl/fl) vav1-cre and ric8 (fl/fl) mb1-cre mice. The major hematopoietic cell lineages developed in the ric8 (fl/fl) vav1-cre mice, notwithstanding severe reduction in Gαi2/3, Gαq, and Gα13 proteins. B lymphocyte-specific loss of Ric-8A did not compromise bone marrow B lymphopoiesis, but splenic marginal zone B cell development failed, and B cells underpopulated lymphoid organs. The ric8 (fl/fl) mb1-cre B cells exhibited poor responses to chemokines, abnormal trafficking, improper in situ positioning, and loss of polarity components during B cell differentiation. The ric8 (fl/fl) mb1-cre mice had a severely disrupted lymphoid architecture and poor primary and secondary Ab responses. In B lymphocytes, Ric-8A is essential for normal Gα protein levels and is required for B cell differentiation, trafficking, and Ab responses.


Asunto(s)
Linfocitos B/inmunología , Subunidad alfa de la Proteína de Unión al GTP Gi2/inmunología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/inmunología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/inmunología , Factores de Intercambio de Guanina Nucleótido/inmunología , Inmunodeficiencia Combinada Grave/inmunología , Animales , Linfocitos B/metabolismo , Western Blotting , Calcio/inmunología , Calcio/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Células Cultivadas , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Centro Germinal/inmunología , Centro Germinal/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Inmunidad Humoral/genética , Inmunidad Humoral/inmunología , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Proteínas Proto-Oncogénicas c-vav/genética , Proteínas Proto-Oncogénicas c-vav/inmunología , Proteínas Proto-Oncogénicas c-vav/metabolismo , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/metabolismo , Imagen de Lapso de Tiempo
9.
PLoS Pathog ; 11(4): e1004827, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25884622

RESUMEN

Neutrophils form the first line of host defense against bacterial pathogens. They are rapidly mobilized to sites of infection where they help marshal host defenses and remove bacteria by phagocytosis. While splenic neutrophils promote marginal zone B cell antibody production in response to administered T cell independent antigens, whether neutrophils shape humoral immunity in other lymphoid organs is controversial. Here we investigate the neutrophil influx following the local injection of Staphylococcus aureus adjacent to the inguinal lymph node and determine neutrophil impact on the lymph node humoral response. Using intravital microscopy we show that local immunization or infection recruits neutrophils from the blood to lymph nodes in waves. The second wave occurs temporally with neutrophils mobilized from the bone marrow. Within lymph nodes neutrophils infiltrate the medulla and interfollicular areas, but avoid crossing follicle borders. In vivo neutrophils form transient and long-lived interactions with B cells and plasma cells, and their depletion augments production of antigen-specific IgG and IgM in the lymph node. In vitro activated neutrophils establish synapse- and nanotube-like interactions with B cells and reduce B cell IgM production in a TGF-ß1 dependent manner. Our data reveal that neutrophils mobilized from the bone marrow in response to a local bacterial challenge dampen the early humoral response in the lymph node.


Asunto(s)
Inmunidad Humoral/inmunología , Ganglios Linfáticos/inmunología , Infiltración Neutrófila/inmunología , Infecciones Estafilocócicas/inmunología , Traslado Adoptivo , Animales , Separación Celular , Ensayo de Inmunoadsorción Enzimática , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Staphylococcus aureus/inmunología
10.
Nat Chem Biol ; 11(4): 271-9, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25706338

RESUMEN

Hypersecretion of norepinephrine (NE) and angiotensin II (AngII) is a hallmark of major prevalent cardiovascular diseases that contribute to cardiac pathophysiology and morbidity. Herein, we explore whether heterodimerization of presynaptic AngII AT1 receptor (AT1-R) and NE α2C-adrenergic receptor (α2C-AR) could underlie their functional cross-talk to control NE secretion. Multiple bioluminescence resonance energy transfer and protein complementation assays allowed us to accurately probe the structures and functions of the α2C-AR-AT1-R dimer promoted by ligand binding to individual protomers. We found that dual agonist occupancy resulted in a conformation of the heterodimer different from that induced by active individual protomers and triggered atypical Gs-cAMP-PKA signaling. This specific pharmacological signaling unit was identified in vivo to promote not only NE hypersecretion in sympathetic neurons but also sympathetic hyperactivity in mice. Thus, we uncovered a new process by which GPCR heterodimerization creates an original functional pharmacological entity and that could constitute a promising new target in cardiovascular therapeutics.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Receptor de Angiotensina Tipo 1/agonistas , Transducción de Señal , Agonistas alfa-Adrenérgicos/química , Animales , Biofisica , Enfermedades Cardiovasculares/metabolismo , AMP Cíclico/metabolismo , Dimerización , Diseño de Fármacos , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Humanos , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Norepinefrina/química , Células PC12 , Fosforilación , Conformación Proteica , Ratas , Receptores Adrenérgicos alfa 2/química , Sistema Nervioso Simpático/efectos de los fármacos
11.
J Immunol ; 194(5): 2128-39, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25617475

RESUMEN

Chemokines engage B lymphocyte surface receptors, triggering heterotrimeric G protein Gαi subunit guanine nucleotide exchange. RGS proteins limit the duration that Gαi subunits remain GTP bound, and the loss of an individual RGS protein typically enhances chemokine receptor signaling. In this study, we show that B cells carrying a Gαi2 (G184S/G184S) mutation that disables all RGS protein/Gαi2 interactions exhibit an unexpectedly severe reduction in chemokine receptor signaling. The Gαi2 (G184S/G184S) B cells have markedly elevated basal calcium levels, but poor chemokine-induced increases, enhanced nonspecific migration, but extremely poor chemotaxis. In striking contrast, the Gαi2 (G184S/G184S) B cells exhibited enhanced sensitivity to sphingosine 1-phosphate (S1P). S1P elicited heightened intracellular calcium responses and enhanced S1P-triggered cell migration. Mice with the Gαi2 (G184S/G184S) mutation displayed excessive numbers of germinal center-like structures; abnormal serum Ig profiles; and aberrant B lymphocyte trafficking. These findings establish an essential role for RGS proteins in B cell chemoattractant signaling and for the proper position of B lymphocytes in lymphoid organs.


Asunto(s)
Subgrupos de Linfocitos B/metabolismo , Quimiotaxis de Leucocito/efectos de los fármacos , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Proteínas RGS/metabolismo , Bazo/metabolismo , Animales , Subgrupos de Linfocitos B/citología , Subgrupos de Linfocitos B/efectos de los fármacos , Subgrupos de Linfocitos B/inmunología , Sitios de Unión , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Calcio/inmunología , Calcio/metabolismo , Quimiocinas/farmacología , Femenino , Subunidad alfa de la Proteína de Unión al GTP Gi2/genética , Subunidad alfa de la Proteína de Unión al GTP Gi2/inmunología , Regulación de la Expresión Génica , Centro Germinal/citología , Centro Germinal/efectos de los fármacos , Centro Germinal/inmunología , Centro Germinal/metabolismo , Lisofosfolípidos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Cultivo Primario de Células , Unión Proteica , Proteínas RGS/genética , Proteínas RGS/inmunología , Transducción de Señal , Esfingosina/análogos & derivados , Esfingosina/farmacología , Bazo/citología , Bazo/efectos de los fármacos , Bazo/inmunología
12.
Mol Cell Biol ; 34(22): 4186-99, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25225330

RESUMEN

Both chemotaxis and phagocytosis depend upon actin-driven cell protrusions and cell membrane remodeling. While chemoattractant receptors rely upon canonical G-protein signaling to activate downstream effectors, whether such signaling pathways affect phagocytosis is contentious. Here, we report that Gαi nucleotide exchange and signaling helps macrophages coordinate the recognition, capture, and engulfment of zymosan bioparticles. We show that zymosan exposure recruits F-actin, Gαi proteins, and Elmo1 to phagocytic cups and early phagosomes. Zymosan triggered an increase in intracellular Ca(2+) that was partially sensitive to Gαi nucleotide exchange inhibition and expression of GTP-bound Gαi recruited Elmo1 to the plasma membrane. Reducing GDP-Gαi nucleotide exchange, decreasing Gαi expression, pharmacologically interrupting Gßγ signaling, or reducing Elmo1 expression all impaired phagocytosis, while favoring the duration that Gαi remained GTP bound promoted it. Our studies demonstrate that targeting heterotrimeric G-protein signaling offers opportunities to enhance or retard macrophage engulfment of phagocytic targets such as zymosan.


Asunto(s)
Subunidad alfa de la Proteína de Unión al GTP Gi2/inmunología , Macrófagos/citología , Fagocitosis , Transducción de Señal , Zimosan/inmunología , Actinas/análisis , Actinas/inmunología , Proteínas Adaptadoras Transductoras de Señales/análisis , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Calcio/análisis , Calcio/inmunología , Línea Celular , Subunidad alfa de la Proteína de Unión al GTP Gi2/análisis , Subunidad alfa de la Proteína de Unión al GTP Gi2/genética , Eliminación de Gen , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Fagosomas/genética , Fagosomas/inmunología , Fagosomas/microbiología , Fagosomas/ultraestructura , Saccharomyces cerevisiae/inmunología
13.
J Immunol ; 193(6): 3080-9, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25135833

RESUMEN

Coronaviruses (CoV) have recently emerged as potentially serious pathogens that can cause significant human morbidity and death. The severe acute respiratory syndrome (SARS)-CoV was identified as the etiologic agent of the 2002-2003 international SARS outbreak. Yet, how SARS evades innate immune responses to cause human disease remains poorly understood. In this study, we show that a protein encoded by SARS-CoV designated as open reading frame-9b (ORF-9b) localizes to mitochondria and causes mitochondrial elongation by triggering ubiquitination and proteasomal degradation of dynamin-like protein 1, a host protein involved in mitochondrial fission. Also, acting on mitochondria, ORF-9b targets the mitochondrial-associated adaptor molecule MAVS signalosome by usurping PCBP2 and the HECT domain E3 ligase AIP4 to trigger the degradation of MAVS, TRAF3, and TRAF 6. This severely limits host cell IFN responses. Reducing either PCBP2 or AIP4 expression substantially reversed the ORF-9b-mediated reduction of MAVS and the suppression of antiviral transcriptional responses. Finally, transient ORF-9b expression led to a strong induction of autophagy in cells. The induction of autophagy depended upon ATG5, a critical autophagy regulator, but the inhibition of MAVS signaling did not. These results indicate that SARS-CoV ORF-9b manipulates host cell mitochondria and mitochondrial function to help evade host innate immunity. This study has uncovered an important clue to the pathogenesis of SARS-CoV infection and illustrates the havoc that a small ORF can cause in cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Inmunidad Innata/genética , Mitocondrias/inmunología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , Proteínas Virales/inmunología , Autofagia/genética , Proteína 5 Relacionada con la Autofagia , Línea Celular , Dinaminas , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Proteínas Fluorescentes Verdes , Células HEK293 , Humanos , Evasión Inmune , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/genética , Mitocondrias/virología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Sistemas de Lectura Abierta/genética , Sistemas de Lectura Abierta/inmunología , Interferencia de ARN , ARN Interferente Pequeño , Proteínas de Unión al ARN/biosíntesis , Proteínas de Unión al ARN/genética , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Síndrome Respiratorio Agudo Grave/inmunología , Síndrome Respiratorio Agudo Grave/virología , Factor 3 Asociado a Receptor de TNF/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Ubiquitina-Proteína Ligasas/biosíntesis , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Proteínas Virales/genética
14.
Nat Commun ; 5: 4431, 2014 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-25028204

RESUMEN

Tumour suppressor PTEN is a phosphatase that negatively regulates the PI3K/AKT pathway. The ability to directly monitor PTEN conformation and function in a rapid, sensitive manner is a key step towards developing anti-cancer drugs aimed at enhancing or restoring PTEN-dependent pathways. Here we developed an intramolecular bioluminescence resonance energy transfer (BRET)-based biosensor, capable of detecting signal-dependent PTEN conformational changes in live cells. The biosensor retains intrinsic properties of PTEN, enabling structure-function and kinetic analyses. BRET shifts, indicating conformational change, were detected following mutations that disrupt intramolecular PTEN interactions, promoting plasma membrane targeting and also following physiological PTEN activation. Using the biosensor as a reporter, we uncovered PTEN activation by several G protein-coupled receptors, previously unknown as PTEN regulators. Trastuzumab, used to treat ERBB2-overexpressing breast cancers also elicited activation-associated PTEN conformational rearrangement. We propose the biosensor can be used to identify pathways regulating PTEN or molecules that enhance its anti-tumour activity.


Asunto(s)
Técnicas Biosensibles/métodos , Fosfohidrolasa PTEN/metabolismo , Línea Celular , Línea Celular Tumoral , Citometría de Flujo , Humanos , Immunoblotting , Inmunoprecipitación , Fosfohidrolasa PTEN/genética , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo
15.
PLoS One ; 9(6): e97957, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24911523

RESUMEN

The omega-3 (ω3) fatty acid docosahexaenoic acid (DHA) can suppress inflammation, specifically IL-1ß production through poorly understood molecular mechanisms. Here, we show that DHA reduces macrophage IL-1ß production by limiting inflammasome activation. Exposure to DHA reduced IL-1ß production by ligands that stimulate the NLRP3, AIM2, and NAIP5/NLRC4 inflammasomes. The inhibition required Free Fatty Acid Receptor (FFAR) 4 (also known as GPR120), a G-protein coupled receptor (GPR) known to bind DHA. The exposure of cells to DHA recruited the adapter protein ß-arrestin1/2 to FFAR4, but not to a related lipid receptor. DHA treatment reduced the initial inflammasome priming step by suppressing the nuclear translocation of NF-κB. DHA also reduced IL-1ß levels by enhancing autophagy in the cells. As a consequence macrophages derived from mice lacking the essential autophagy protein ATG7 were partially resistant to suppressive effects of DHA. Thus, DHA suppresses inflammasome activation by two distinct mechanisms, inhibiting the initial priming step and by augmenting autophagy, which limits inflammasome activity.


Asunto(s)
Autofagia/efectos de los fármacos , Ácidos Docosahexaenoicos/farmacología , Inflamasomas/metabolismo , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , FN-kappa B/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Arrestinas/metabolismo , Células de la Médula Ósea/citología , Calcio/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Interleucina-1beta/biosíntesis , Interleucina-1beta/metabolismo , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores Acoplados a Proteínas G/genética , beta-Arrestinas
16.
Cell Signal ; 26(6): 1269-82, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24583286

RESUMEN

Heterotrimeric guanine nucleotide-binding proteins (G proteins), which consist of three subunits α, ß, and γ, function as molecular switches to control downstream effector molecules activated by G protein-coupled receptors (GPCRs). The GTP/GDP binding status of Gα transmits information about the ligand binding state of the GPCR to intended signal transduction pathways. In immune cells heterotrimeric G proteins impact signal transduction pathways that directly, or indirectly, regulate cell migration, activation, survival, proliferation, and differentiation. The cells of the innate and adaptive immune system abundantly express chemoattractant receptors and lesser amounts of many other types of GPCRs. But heterotrimeric G-proteins not only function in classical GPCR signaling, but also in non-canonical signaling. In these pathways the guanine exchange factor (GEF) exerted by a GPCR in the canonical pathway is replaced or supplemented by another protein such as Ric-8A. In addition, other proteins such as AGS3-6 can compete with Gßγ for binding to GDP bound Gα. This competition can promote Gßγ signaling by freeing Gßγ from rapidly rebinding GDP bound Gα. The proteins that participate in these non-canonical signaling pathways will be briefly described and their role, or potential one, in cells of the immune system will be highlighted.


Asunto(s)
Proteínas de Unión al GTP Heterotriméricas/fisiología , Receptores Acoplados a Proteínas G/fisiología , Animales , Expresión Génica/inmunología , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/metabolismo , Inmunidad Celular , Inmunidad Innata , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal
17.
PLoS One ; 9(1): e86680, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24466196

RESUMEN

Resistance to inhibitors of cholinesterase (Ric)-8A is a guanine nucleotide exchange factor for Gαi, Gαq, and Gα12/13, which is implicated in cell signaling and as a molecular chaperone required for the initial association of nascent Gα subunits with cellular membranes. Ric-8A, Gαi subunits, and their regulators are localized at the midbody prior to abscission and linked to the final stages of cell division. Here, we identify a molecular mechanism by which Ric-8A affects cytokinesis and abscission by controlling Vps34 activity. We showed that Ric-8A protein expression is post-transcriptionally controlled during the cell cycle reaching its maximum levels at mitosis. A FRET biosensor created to measure conformational changes in Ric-8A by FLIM (Fluorescence Lifetime Imaging Microscopy) revealed that Ric-8A was in a close-state during mitosis and particularly so at cytokinesis. Lowering Ric-8A expression delayed the abscission time of dividing cells, which correlated with increased intercellular bridge length and multinucleation. During cytokinesis, Ric-8A co-localized with Vps34 at the midbody along with Gαi and LGN, where these proteins functioned to regulate Vps34 phosphatidylinositol 3-kinase activity.


Asunto(s)
Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Citocinesis/fisiología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Línea Celular Tumoral , Células HeLa , Humanos , Mitosis/fisiología , Proteínas Nucleares/metabolismo , Procesamiento Postranscripcional del ARN/fisiología
18.
EMBO J ; 30(13): 2557-68, 2011 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-21642958

RESUMEN

The tumour suppressor PTEN (phosphatase and tensin deleted on chromosome 10) regulates major cellular functions via lipid phosphatase-dependent and -independent mechanisms. Despite its fundamental pathophysiological importance, how PTEN's cellular activity is regulated has only been partially elucidated. We report that the scaffolding proteins ß-arrestins (ß-arrs) are important regulators of PTEN. Downstream of receptor-activated RhoA/ROCK signalling, ß-arrs activate the lipid phosphatase activity of PTEN to negatively regulate Akt and cell proliferation. In contrast, following wound-induced RhoA activation, ß-arrs inhibit the lipid phosphatase-independent anti-migratory effects of PTEN. ß-arrs can thus differentially control distinct functional outputs of PTEN important for cell proliferation and migration.


Asunto(s)
Arrestinas/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosfohidrolasa PTEN/fisiología , Animales , Arrestinas/antagonistas & inhibidores , Arrestinas/genética , Arrestinas/fisiología , Células COS , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Chlorocebus aethiops , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Ratones , Fosfohidrolasa PTEN/genética , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Unión Proteica/fisiología , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología , beta-Arrestinas
19.
Cell ; 143(7): 1149-60, 2010 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-21183077

RESUMEN

Following pilus-mediated adhesion to human brain endothelial cells, meningococcus (N. meningitidis), the bacterium causing cerebrospinal meningitis, initiates signaling cascades, which eventually result in the opening of intercellular junctions, allowing meningeal colonization. The signaling receptor activated by the pathogen remained unknown. We report that N. meningitidis specifically stimulates a biased ß2-adrenoceptor/ß-arrestin signaling pathway in endothelial cells, which ultimately traps ß-arrestin-interacting partners, such as the Src tyrosine kinase and junctional proteins, under bacterial colonies. Cytoskeletal reorganization mediated by ß-arrestin-activated Src stabilizes bacterial adhesion to endothelial cells, whereas ß-arrestin-dependent delocalization of junctional proteins results in anatomical gaps used by bacteria to penetrate into tissues. Activation of ß-adrenoceptor endocytosis with specific agonists prevents signaling events downstream of N. meningitidis adhesion and inhibits bacterial crossing of the endothelial barrier. The identification of the mechanism used for hijacking host cell signaling machineries opens perspectives for treatment and prevention of meningococcal infection.


Asunto(s)
Arrestinas/metabolismo , Encéfalo/microbiología , Células Endoteliales/microbiología , Infecciones Meningocócicas/metabolismo , Neisseria meningitidis/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal , Adhesión Bacteriana , Barrera Hematoencefálica , Línea Celular , Humanos , Infecciones Meningocócicas/microbiología , beta-Arrestinas
20.
Mol Cell ; 38(1): 78-88, 2010 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-20385091

RESUMEN

p53 downstream pathways control G1 and G2 cell-cycle arrest, DNA repair, or apoptosis. However, it is still not clear how cells differentiate the cell-biological outcome of p53 activation in response to different types of stresses. The p53/47 isoform lacks the first 39 amino acids of full-length p53 including the Mdm2 binding site and the first trans-activation domain, and tetramers including p53/47 exhibit altered activity and biochemical properties. Here we show that endoplasmic reticulum stress promotes PERK-dependent induction of p53/47 mRNA translation and p53/47 homo-oligomerization. p53/47 induces 14-3-3sigma and G2 arrest but does not affect G1 progression. This is contrary to p53FL, which promotes G1 arrest but has no effect on the G2. These results show a unique role for p53/47 in the p53 pathway and illustrate how a cellular stress leads to a defined cell-biological outcome through expression of a p53 isoform.


Asunto(s)
Retículo Endoplásmico/metabolismo , Fase G2/fisiología , Biosíntesis de Proteínas , Isoformas de Proteínas/metabolismo , ARN Mensajero/metabolismo , Estrés Fisiológico , Proteína p53 Supresora de Tumor/metabolismo , Proteínas 14-3-3 , Apoptosis/fisiología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Línea Celular , Exonucleasas/genética , Exonucleasas/metabolismo , Exorribonucleasas , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Isoformas de Proteínas/genética , ARN Mensajero/genética , Proteína p53 Supresora de Tumor/genética , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA