Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros












Intervalo de año de publicación
2.
Biol Chem ; 401(8): 901-902, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32436856
3.
FASEB J ; 33(2): 2599-2609, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30281335

RESUMEN

Neutrophil recruitment and plasma exudation are key elements in the immune response to injury or infection. Activated neutrophils stimulate opening of the endothelial barrier; however, the underlying mechanisms have remained largely unknown. In this study, we identified a pivotal role of the proinflammatory kallikrein-kinin system and consequent formation of bradykinin in neutrophil-evoked vascular leak. In mouse and hamster models of acute inflammation, inhibitors of bradykinin generation, and signaling markedly reduced plasma exudation in response to chemoattractant activation of neutrophils. The neutrophil-driven leak was likewise suppressed in mice deficient in either the bradykinin B2 receptor or factor XII (initiator of the kallikrein-kinin system). In human endothelial cell monolayers, material secreted from activated neutrophils induced cytoskeletal rearrangement, leading to paracellular gap formation in a bradykinin-dependent manner. As a mechanistic basis, we found that a neutrophil-derived heparin-binding protein (HBP/azurocidin) displaced the bradykinin precursor high-molecular-weight kininogen from endothelial cells, thereby enabling proteolytic processing of kininogen into bradykinin by neutrophil and plasma proteases. These data provide novel insight into the signaling pathway by which neutrophils open up the endothelial barrier and identify the kallikrein-kinin system as a target for therapeutic interventions in acute inflammatory reactions.-Kenne, E., Rasmuson, J., Renné, T., Vieira, M. L., Müller-Esterl, W., Herwald, H., Lindbom, L. Neutrophils engage the kallikrein-kinin system to open up the endothelial barrier in acute inflammation.


Asunto(s)
Permeabilidad de la Membrana Celular , Endotelio Vascular/fisiología , Inflamación/patología , Sistema Calicreína-Quinina/fisiología , Neutrófilos/metabolismo , Edema Pulmonar/patología , Animales , Bradiquinina/metabolismo , Endotelio Vascular/citología , Factor XII/metabolismo , Femenino , Humanos , Inflamación/metabolismo , Quininógeno de Alto Peso Molecular/metabolismo , Masculino , Mesocricetus , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila , Edema Pulmonar/etiología , Edema Pulmonar/metabolismo
4.
PLoS One ; 9(12): e116150, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25546391

RESUMEN

Holoprosencephaly is a common developmental disorder in humans characterised by incomplete brain hemisphere separation and midface anomalies. The etiology of holoprosencephaly is heterogeneous with environmental and genetic causes, but for a majority of holoprosencephaly cases the genes associated with the pathogenesis could not be identified so far. Here we report the generation of knockout mice for the ubiquitin E3 ligase NOSIP. The loss of NOSIP in mice causes holoprosencephaly and facial anomalies including cleft lip/palate, cyclopia and facial midline clefting. By a mass spectrometry based protein interaction screen we identified NOSIP as a novel interaction partner of protein phosphatase PP2A. NOSIP mediates the monoubiquitination of the PP2A catalytic subunit and the loss of NOSIP results in an increase in PP2A activity in craniofacial tissue in NOSIP knockout mice. We conclude, that NOSIP is a critical modulator of brain and craniofacial development in mice and a candidate gene for holoprosencephaly in humans.


Asunto(s)
Cara/embriología , Proteína Fosfatasa 2/metabolismo , Cráneo/embriología , Cráneo/enzimología , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Animales Recién Nacidos , Dominio Catalítico , Fisura del Paladar/embriología , Fisura del Paladar/enzimología , Cara/anomalías , Holoprosencefalia/embriología , Holoprosencefalia/enzimología , Holoprosencefalia/patología , Metilación , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Cráneo/anomalías , Ubiquitinación
5.
EMBO J ; 31(15): 3309-22, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22751148

RESUMEN

F-BAR proteins are multivalent adaptors that link plasma membrane and cytoskeleton and coordinate cellular processes such as membrane protrusion and migration. Yet, little is known about the function of F-BAR proteins in vivo. Here we report, that the F-BAR protein NOSTRIN is necessary for proper vascular development in zebrafish and postnatal retinal angiogenesis in mice. The loss of NOSTRIN impacts on the migration of endothelial tip cells and leads to a reduction of tip cell filopodia number and length. NOSTRIN forms a complex with the GTPase Rac1 and its exchange factor Sos1 and overexpression of NOSTRIN in cells induces Rac1 activation. Furthermore, NOSTRIN is required for fibroblast growth factor 2 dependent activation of Rac1 in primary endothelial cells and the angiogenic response to fibroblast growth factor 2 in the in vivo matrigel plug assay. We propose a novel regulatory circuit, in which NOSTRIN assembles a signalling complex containing FGFR1, Rac1 and Sos1 thereby facilitating the activation of Rac1 in endothelial cells during developmental angiogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Vasos Sanguíneos/embriología , Proteínas de Unión al ADN/fisiología , Factores de Crecimiento de Fibroblastos/metabolismo , Neovascularización Fisiológica/genética , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Animales Modificados Genéticamente , Animales Recién Nacidos , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/fisiología , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Embrión de Mamíferos , Embrión no Mamífero , Factores de Crecimiento de Fibroblastos/fisiología , Ratones , Ratones Noqueados , Modelos Biológicos , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología , Pez Cebra/embriología , Pez Cebra/genética
6.
PLoS One ; 7(5): e37485, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22629405

RESUMEN

Treatment of malignant glioma with chemotherapy is limited mostly because of delivery impediment related to the blood-brain tumor barrier (BTB). B1 receptors (B1R), inducible prototypical G-protein coupled receptors (GPCR) can regulate permeability of vessels including possibly that of brain tumors. Here, we determine the extent of BTB permeability induced by the natural and synthetic peptide B1R agonists, LysdesArg(9)BK (LDBK) and SarLys[dPhe(8)]desArg(9)BK (NG29), in syngeneic F98 glioma-implanted Fischer rats. Ten days after tumor inoculation, we detected the presence of B1R on tumor cells and associated vasculature. NG29 infusion increased brain distribution volume and uptake profiles of paramagnetic probes (Magnevist and Gadomer) at tumoral sites (T(1)-weighted imaging). These effects were blocked by B1R antagonist and non-selective cyclooxygenase inhibitors, but not by B2R antagonist and non-selective nitric oxide synthase inhibitors. Consistent with MRI data, systemic co-administration of NG29 improved brain tumor delivery of Carboplatin chemotherapy (ICP-Mass spectrometry). We also detected elevated B1R expression in clinical samples of high-grade glioma. Our results documented a novel GPCR-signaling mechanism for promoting transient BTB disruption, involving activation of B1R and ensuing production of COX metabolites. They also underlined the potential value of synthetic biostable B1R agonists as selective BTB modulators for local delivery of different sized-therapeutics at (peri)tumoral sites.


Asunto(s)
Bradiquinina/análogos & derivados , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Calidina/análogos & derivados , Receptor de Bradiquinina B1/agonistas , Adulto , Anciano , Animales , Transporte Biológico/efectos de los fármacos , Bradiquinina/farmacología , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/tratamiento farmacológico , Permeabilidad Capilar/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Glioma/irrigación sanguínea , Glioma/tratamiento farmacológico , Humanos , Calidina/farmacología , Masculino , Persona de Mediana Edad , Permeabilidad/efectos de los fármacos , Ratas , Ratas Endogámicas F344 , Receptor de Bradiquinina B1/metabolismo
7.
Immunity ; 34(2): 258-68, 2011 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-21349432

RESUMEN

Activated mast cells trigger edema in allergic and inflammatory disease. We report a paracrine mechanism by which mast cell-released heparin increases vascular permeability in vivo. Heparin activated the protease factor XII, which initiates bradykinin formation in plasma. Targeting factor XII or kinin B2 receptors abolished heparin-triggered leukocyte-endothelium adhesion and interfered with a mast cell-driven drop in blood pressure in rodents. Intravital laser scanning microscopy and tracer measurements showed heparin-driven fluid extravasation in mouse skin microvessels. Ablation of factor XII or kinin B2 receptors abolished heparin-induced skin edema and protected mice from allergen-activated mast cell-driven leakage. In contrast, heparin and activated mast cells induced excessive edema in mice deficient in the major inhibitor of factor XII, C1 esterase inhibitor. Allergen exposure triggered edema attacks in hereditary angioedema patients, lacking C1 esterase inhibitor. The data indicate that heparin-initiated bradykinin formation plays a fundamental role in mast cell-mediated diseases.


Asunto(s)
Bradiquinina/biosíntesis , Síndrome de Fuga Capilar/fisiopatología , Permeabilidad Capilar/fisiología , Heparina/fisiología , Mastocitos/metabolismo , Anafilaxis Cutánea Pasiva/fisiología , Animales , Bradiquinina/genética , Síndrome de Fuga Capilar/etiología , Adhesión Celular , Proteína Inhibidora del Complemento C1/fisiología , Edema/etiología , Edema/fisiopatología , Células Endoteliales/patología , Activación Enzimática , Factor XII/fisiología , Heparina/metabolismo , Hipotensión/etiología , Hipotensión/fisiopatología , Inmunoglobulina E/inmunología , Sistema Calicreína-Quinina/fisiología , Leucocitos/fisiología , Masculino , Ratones , Comunicación Paracrina/fisiología , Plasma , Ratas , Transducción de Señal/fisiología , Piel/irrigación sanguínea
8.
Microbiology (Reading) ; 156(Pt 12): 3660-3668, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20705662

RESUMEN

Since the late 1980s, a worldwide increase of severe Streptococcus pyogenes infections has been associated with strains of the M1 serotype, strains which all secrete the streptococcal inhibitor of complement-mediated lysis (SIC). Previous work has shown that SIC blocks complement-mediated haemolysis, inhibits the activity of antibacterial peptides and has affinity for the human plasma proteins clusterin and histidine-rich glycoprotein; the latter is a member of the cystatin protein family. The present work demonstrates that SIC binds to cystatin C, high-molecular-mass kininogen (HK) and low-molecular-mass kininogen, which are additional members of this protein family. The binding sites in HK are located in the cystatin-like domain D3 and the endothelial cell-binding domain D5. Immobilization of HK to cellular structures plays a central role in activation of the human contact system. SIC was found to inhibit the binding of HK to endothelial cells, and to reduce contact activation as measured by prolonged blood clotting time and impaired release of bradykinin. These results suggest that SIC modifies host defence systems, which may contribute to the virulence of S. pyogenes strains of the M1 serotype.


Asunto(s)
Proteínas Bacterianas/inmunología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/inmunología , Streptococcus pyogenes/patogenicidad , Proteínas Bacterianas/genética , Proteínas del Sistema Complemento , Cistatina C/inmunología , Interacciones Huésped-Patógeno , Humanos , Quininógeno de Alto Peso Molecular/inmunología , Unión Proteica , Infecciones Estreptocócicas/inmunología , Streptococcus pyogenes/genética , Virulencia
9.
J Cereb Blood Flow Metab ; 30(1): 130-9, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19773800

RESUMEN

Inflammatory mechanisms are known to contribute to the pathophysiology of traumatic brain injury (TBI). Since bradykinin is one of the first mediators activated during inflammation, we investigated the role of bradykinin and its receptors in posttraumatic secondary brain damage. We subjected wild-type (WT), B(1)-, and B(2)-receptor-knockout mice to controlled cortical impact (CCI) and analyzed tissue bradykinin as well as kinin receptor mRNA and protein expression up to 48 h thereafter. Brain edema, contusion volume, and functional outcome were assessed 24 h and 7 days after CCI. Tissue bradykinin was maximally increased 2 h after trauma (P<0.01 versus sham). Kinin B(1) receptor mRNA was upregulated up to four-fold 24 h after CCI. Immunohistochemistry showed that B(1) and B(2) receptors were expressed in the brain and were significantly upregulated in the traumatic penumbra 1 to 24 h after CCI. B(2)R(-/-) mice had significantly less brain edema (-51% versus WT, 24 h; P<0.001), smaller contusion volumes ( approximately 50% versus WT 24 h and 7 d after CCI; P<0.05), and better functional outcome 7 days after TBI as compared with WT mice (P<0.05). The present results show that bradykinin and its B(2) receptors play a causal role for brain edema formation and cell death after TBI.


Asunto(s)
Lesiones Encefálicas/patología , Receptor de Bradiquinina B1/fisiología , Receptor de Bradiquinina B2/fisiología , Animales , Bradiquinina/metabolismo , Edema Encefálico/patología , Contusiones/patología , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor de Bradiquinina B1/biosíntesis , Receptor de Bradiquinina B1/genética , Receptor de Bradiquinina B2/biosíntesis , Receptor de Bradiquinina B2/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Nat Cell Biol ; 11(7): 873-80, 2009 07.
Artículo en Inglés | MEDLINE | ID: mdl-19503073

RESUMEN

Epidermal growth factor-like domain 7 (EGFL7) is a secreted factor implicated in cellular responses such as cell migration and blood vessel formation; however the molecular mechanisms underlying the effects of EGFL7 are largely unknown. Here we have identified transmembrane receptors of the Notch family as EGFL7-binding molecules. Secreted EGFL7 binds to a region in Notch involved in ligand-mediated receptor activation, thus acting as an antagonist of Notch signalling. Expression of EGFL7 in neural stem cells (NSCs) in vitro decreased Notch-specific signalling and consequently, reduced proliferation and self-renewal of NSCs. Such altered Notch signalling caused a shift in the differentiation pattern of cultured NSCs towards an excess of neurons and oligodendrocytes. We identified neurons as a source of EGFL7 in the brain, suggesting that brain-derived EGFL7 acts as an endogenous antagonist of Notch signalling that regulates proliferation and differentiation of subventricular zone-derived adult NSCs.


Asunto(s)
Factores de Crecimiento Endotelial/fisiología , Neuronas/citología , Neuronas/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Células Madre/citología , Células Madre/metabolismo , Animales , Proteínas de Unión al Calcio , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Proliferación Celular , Células Cultivadas , Familia de Proteínas EGF , Factores de Crecimiento Endotelial/genética , Factores de Crecimiento Endotelial/metabolismo , Humanos , Ratones , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Técnicas del Sistema de Dos Híbridos
11.
J Immunol ; 182(12): 7906-15, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19494315

RESUMEN

The kinin-forming pathway is activated on endothelial cells and neutrophils when high-molecular weight kininogen (HK) is cleaved by plasma kallikrein liberating bradykinin, a potent mediator of inflammation. Kinins are released during inflammatory conditions such as vasculitis, associated with neutrophil influx around blood vessels. Some patients with vasculitis have elevated plasma levels of neutrophil-derived proteinase 3 (PR3) and anti-PR3 Abs. This study investigated if neutrophil-derived PR3 could induce activation of the kinin pathway. PR3 incubated with HK, or a synthetic peptide derived from HK, induced breakdown and release of a novel tridecapeptide termed PR3-kinin, NH(2)-MKRPPGFSPFRSS-COOH, consisting of bradykinin with two additional amino acids on each terminus. The reaction was specific and inhibited by anti-PR3 and alpha(1)-antitrypsin. Recombinant wild-type PR3 incubated with HK induced HK breakdown, whereas mutated PR3, lacking enzymatic activity, did not. PR3-kinin bound to and activated human kinin B(1) receptors, but did not bind to B(2) receptors, expressed by transfected HEK293 cells in vitro. In human plasma PR3-kinin was further processed to the B(2) receptor agonist bradykinin. PR3-kinin exerted a hypotensive effect in vivo through both B(1) and B(2) receptors as demonstrated using wild-type and B(1) overexpressing rats as well as wild-type and B(2) receptor knockout mice. Neutrophil extracts from vasculitis patients and healthy controls contained comparable amounts of PR3 and induced HK proteolysis, an effect that was abolished when PR3 was immunoadsorbed. Neutrophil-derived PR3 can proteolyze HK and liberate PR3-kinin, thereby initiating kallikrein-independent activation of the kinin pathway.


Asunto(s)
Cininas/metabolismo , Mieloblastina/metabolismo , Neutrófilos/enzimología , Adolescente , Anciano , Anciano de 80 o más Años , Animales , Bradiquinina/sangre , Niño , Humanos , Calicreínas/metabolismo , Quininógenos/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Mieloblastina/genética , Unión Proteica , Ratas
12.
Circ Res ; 105(1): 33-41, 2009 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-19478201

RESUMEN

Nitric oxide (NO) is an essential vasodilator. In vascular diseases, oxidative stress attenuates NO signaling by both chemical scavenging of free NO and oxidation and downregulation of its major intracellular receptor, the alphabeta heterodimeric heme-containing soluble guanylate cyclase (sGC). Oxidation can also induce loss of the heme of sGC, as well as the responsiveness of sGC to NO. sGC activators such as BAY 58-2667 bind to oxidized/heme-free sGC and reactivate the enzyme to exert disease-specific vasodilation. Here, we show that oxidation-induced downregulation of sGC protein extends to isolated blood vessels. Mechanistically, degradation was triggered through sGC ubiquitination and proteasomal degradation. The heme-binding site ligand BAY 58-2667 prevented sGC ubiquitination and stabilized both alpha and beta subunits. Collectively, our data establish oxidation-ubiquitination of sGC as a modulator of NO/cGMP signaling and point to a new mechanism of action for sGC activating vasodilators by stabilizing their receptor, oxidized/heme-free sGC.


Asunto(s)
Guanilato Ciclasa/metabolismo , Hemo/metabolismo , Óxido Nítrico/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Vasodilatadores/farmacología , Vasos Sanguíneos , Línea Celular , GMP Cíclico/metabolismo , Humanos , Oxidación-Reducción , Guanilil Ciclasa Soluble , Ubiquitinación
13.
Differentiation ; 77(4): 377-85, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19281786

RESUMEN

The V(2) vasopressin receptor gene contains an alternative splice site in exon-3, which leads to the generation of two splice variants (V(2a) and V(2b)) first identified in the kidney. The open reading frame of the alternatively spliced V(2b) transcript encodes a truncated receptor, showing the same amino acid sequence as the canonical V(2a) receptor up to the sixth transmembrane segment, but displaying a distinct sequence to the corresponding seventh transmembrane segment and C-terminal domain relative to the V(2a) receptor. Here, we demonstrate the postnatal expression of V(2a) and V(2b) variants in the rat cerebellum. Most importantly, we showed by in situ hybridization and immunocytochemistry that both V(2) splice variants were preferentially expressed in Purkinje cells, from early to late postnatal development. In addition, both variants were transiently expressed in the neuroblastic external granule cells and Bergmann fibers. These results indicate that the cellular distributions of both splice variants are developmentally regulated, and suggest that the transient expression of the V(2) receptor is involved in the mechanisms of cerebellar cytodifferentiation by AVP. Finally, transfected CHO-K1 expressing similar amounts of both V(2) splice variants, as that found in the cerebellum, showed a significant reduction in the surface expression of V(2a) receptors, suggesting that the differential expression of the V(2) splice variants regulates the vasopressin signaling in the cerebellum.


Asunto(s)
Cerebelo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Receptores de Vasopresinas/metabolismo , Animales , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Femenino , Variación Genética , Inmunohistoquímica , Hibridación in Situ , Isoformas de Proteínas/metabolismo , Células de Purkinje/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Vasopresinas/clasificación , Receptores de Vasopresinas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
J Immunol ; 182(4): 2063-73, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19201859

RESUMEN

Persistent mitochondrial hyperpolarization (MHP) and enhanced calcium fluxing underlie aberrant T cell activation and death pathway selection in systemic lupus erythematosus. Treatment with rapamycin, which effectively controls disease activity, normalizes CD3/CD28-induced calcium fluxing but fails to influence MHP, suggesting that altered calcium fluxing is downstream or independent of mitochondrial dysfunction. In this article, we show that activity of the mammalian target of rapamycin (mTOR), which is a sensor of the mitochondrial transmembrane potential, is increased in lupus T cells. Activation of mTOR was inducible by NO, a key trigger of MHP, which in turn enhanced the expression of HRES-1/Rab4, a small GTPase that regulates recycling of surface receptors through early endosomes. Expression of HRES-1/Rab4 was increased in CD4(+) lupus T cells, and in accordance with its dominant impact on the endocytic recycling of CD4, it was inversely correlated with diminished CD4 expression. HRES-1/Rab4 overexpression was also inversely correlated with diminished TCRzeta protein levels. Pull-down studies revealed a direct interaction of HRES-1/Rab4 with CD4 and TCRzeta. Importantly, the deficiency of the TCRzeta chain and of Lck and the compensatory up-regulation of FcepsilonRIgamma and Syk, which mediate enhanced calcium fluxing in lupus T cells, were reversed in patients treated with rapamcyin in vivo. Knockdown of HRES-1/Rab4 by small interfering RNA and inhibitors of lysosomal function augmented TCRzeta protein levels in vitro. The results suggest that activation of mTOR causes the loss of TCRzeta in lupus T cells through HRES-1/Rab4-dependent lysosomal degradation.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Lupus Eritematoso Sistémico/inmunología , Proteínas Quinasas/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas de Unión al GTP rab4/inmunología , Adolescente , Adulto , Western Blotting , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Femenino , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Humanos , Inmunosupresores/uso terapéutico , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/metabolismo , Microscopía Confocal , Persona de Mediana Edad , Óxido Nítrico/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Quinasas/metabolismo , ARN Interferente Pequeño , Receptores de Antígenos de Linfocitos T/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR , Transfección , Proteínas de Unión al GTP rab4/metabolismo , Proteínas de Unión al GTP rab5/biosíntesis
15.
Traffic ; 10(1): 26-34, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18980613

RESUMEN

We recently observed that a novel, shortened variant of eNOS trafficking inducer (NOSTRIN) is expressed in cirrhotic liver. This shortened variant (NOSTRINbeta) lacks the first 78 amino acids of full-length NOSTRIN (NOSTRINalpha) and thus a substantial part of its F-BAR domain. In contrast to NOSTRINalpha, NOSTRINbeta mainly localizes to the cell nucleus. In this study, we show that nuclear import of NOSTRINbeta depends on two nuclear localization signals (aa 32-36: KKRK and aa 57-61: KAKKK). Each of the sequences is independently functional, but both are required to sustain nuclear localization of NOSTRINbeta. Export of NOSTRINbeta from the nucleus is facilitated by a CRM1-dependent mechanism relying on the nuclear export sequence LELEKERIQL (aa 135-145). Unlike NOSTRINbeta, the full-length variant NOSTRINalpha was conspicuously absent from the nucleus. This is most likely because of the fact that its N-terminal F-BAR domain, which is truncated in NOSTRINbeta, facilitates association with cellular membranes. NOSTRINbeta directly binds to the 5'-regulatory region of the NOSTRIN gene (bp -200 to -1), and overexpression of NOSTRINbeta strongly decreases transcription of a reporter gene under control of this DNA region. Taken together, our results suggest that nuclear NOSTRINbeta may negatively regulate transcription of the NOSTRIN gene.


Asunto(s)
Empalme Alternativo/genética , Regulación de la Expresión Génica/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Transcripción Genética/genética , Transporte Activo de Núcleo Celular , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Aminoácidos , Línea Celular , Núcleo Celular/metabolismo , Proteínas de Unión al ADN , Genes Reporteros/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
16.
J Cell Physiol ; 216(1): 234-44, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18264983

RESUMEN

Bradykinin (BK) represents a pro-inflammatory mediator that partakes in many inflammatory diseases. The mechanism of action of BK is thought to be primarily mediated by specific cell surface membrane B2 receptors (B2Rs). Some evidence has suggested, however, the existence of an intracellular/nuclear B2R population. Whether these receptors are functional and contribute to BK signaling remains to be determined. In this study, by mean of Western blotting, 3D-confocal microscopy, receptor autoradiography and radioligand binding analysis, we showed that plasma membrane and highly purified nuclei from isolated rat hepatocytes contain specific B2R that bind BK. The results depicting B2R nuclear expression in isolated nuclear organelles were reproduced in situ on hepatic sections by immunogold labeling and transmission electron microscopy. Functional tests on single nuclei, by means of confocal microscopy and the calcium-sensitive probe fluo-4AM, showed that BK induces concentration-dependent transitory mobilization of nucleoplasmic calcium; these responses were blocked by B2R antagonist HOE 140, not by the B1R antagonist R954 and, were also found in wild-type C57/Bl6 mice, but not in B2R-KO mice. In isolated nuclei, BK elicited activation/phosphorylation of Akt, acetylation of histone H3 and ensuing pro-inflammatory iNOS gene induction as determined by Western blot and RT-PCR. ChIP assay confirmed binding of acetylated-histone H3 complexes, but not B2R, to promoter region of iNOS gene suggesting that B2R-mediated gene expression is bridged with accessory downstream effectors. This study discloses a previously undescribed mechanism in BK-induced transcriptional events, via intracrine B2R-mediated signaling, occurring in rat autologous hepatic cells.


Asunto(s)
Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Regulación de la Expresión Génica , Genes Inmediatos-Precoces , Receptor de Bradiquinina B2/metabolismo , Transducción de Señal/fisiología , Animales , Bradiquinina/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Quininógeno de Alto Peso Molecular/genética , Quininógeno de Alto Peso Molecular/metabolismo , Quininógeno de Bajo Peso Molecular/genética , Quininógeno de Bajo Peso Molecular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Receptor de Bradiquinina B2/genética , Activación Transcripcional , Vasodilatadores/metabolismo
17.
Am J Physiol Lung Cell Mol Physiol ; 294(3): L505-15, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18192590

RESUMEN

Ferritin is a protein principally known for its role in iron storage. We have previously shown that ferritin can bind high-molecular-weight kininogen (HK). Upon proteolytic cleavage by the protease kallikrein, HK releases the proinflammatory peptide bradykinin (BK) and other biologically active products, such as two-chain high-molecular-weight kininogen, HKa. At inflammatory sites, HK is oxidized, which renders it a poor substrate for kallikrein. However, oxidized HK remains a good substrate for elastase and tryptase, thereby providing an alternative cleavage mechanism for HK during inflammation. Here we report that ferritin can retard the cleavage of both native HK and oxidized HK by elastase and tryptase. Initial rates of cleavage were reduced 45-75% in the presence of ferritin. Ferritin is not a substrate for elastase or tryptase and does not interfere with the ability of either protease to digest a synthetic substrate, suggesting that ferritin may impede HK cleavage through direct interaction with HK. Immunoprecipitation and solid phase binding studies reveal that ferritin and HK bind directly with a Kd of 134 nM. To test whether ferritin regulates HK cleavage in vivo, we used THP-1 cells, a human monocyte/macrophage cell line that has been used to model pulmonary inflammatory cells. We observed that ferritin impedes the cleavage of HK by secretory proteases in stimulated macrophages. Furthermore, ferritin, HK, and elastase are all present in or on alveolar macrophages in a mouse model of pulmonary inflammation. Collectively, these results implicate ferritin in the modulation of HK cleavage at sites of inflammation.


Asunto(s)
Ferritinas/farmacología , Quininógeno de Alto Peso Molecular/metabolismo , Elastasa Pancreática/antagonistas & inhibidores , Elastasa Pancreática/metabolismo , Triptasas/antagonistas & inhibidores , Triptasas/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Ferritinas/metabolismo , Humanos , Inmunoprecipitación , Inflamación/fisiopatología , Macrófagos Alveolares/metabolismo , Ratones , Oxidación-Reducción , Peroxidasa/metabolismo , Unión Proteica , Succinimidas/química , Zinc/farmacología
18.
PLoS Pathog ; 3(11): e185, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18052532

RESUMEN

Although the concept that dendritic cells (DCs) recognize pathogens through the engagement of Toll-like receptors is widely accepted, we recently suggested that immature DCs might sense kinin-releasing strains of Trypanosoma cruzi through the triggering of G-protein-coupled bradykinin B2 receptors (B2R). Here we report that C57BL/6.B2R-/- mice infected intraperitoneally with T. cruzi display higher parasitemia and mortality rates as compared to B2R+/+ mice. qRT-PCR revealed a 5-fold increase in T. cruzi DNA (14 d post-infection [p.i.]) in B2R-/- heart, while spleen parasitism was negligible in both mice strains. Analysis of recall responses (14 d p.i.) showed high and comparable frequencies of IFN-gamma-producing CD4+ and CD8+ T cells in the spleen of B2R-/- and wild-type mice. However, production of IFN-gamma by effector T cells isolated from B2R-/- heart was significantly reduced as compared with wild-type mice. As the infection continued, wild-type mice presented IFN-gamma-producing (CD4+CD44+ and CD8+CD44+) T cells both in the spleen and heart while B2R-/- mice showed negligible frequencies of such activated T cells. Furthermore, the collapse of type-1 immune responses in B2R-/- mice was linked to upregulated secretion of IL-17 and TNF-alpha by antigen-responsive CD4+ T cells. In vitro analysis of tissue culture trypomastigote interaction with splenic CD11c+ DCs indicated that DC maturation (IL-12, CD40, and CD86) is controlled by the kinin/B2R pathway. Further, systemic injection of trypomastigotes induced IL-12 production by CD11c+ DCs isolated from B2R+/+ spleen, but not by DCs from B2R-/- mice. Notably, adoptive transfer of B2R+/+ CD11c+ DCs (intravenously) into B2R-/- mice rendered them resistant to acute challenge, rescued development of type-1 immunity, and repressed TH17 responses. Collectively, our results demonstrate that activation of B2R, a DC sensor of endogenous maturation signals, is critically required for development of acquired resistance to T. cruzi infection.


Asunto(s)
Enfermedad de Chagas/inmunología , Células Dendríticas/inmunología , Cininas/metabolismo , Receptor de Bradiquinina B2/inmunología , Células TH1/inmunología , Traslado Adoptivo , Animales , Enfermedad de Chagas/metabolismo , Células Dendríticas/metabolismo , Citometría de Flujo , Interferón gamma/biosíntesis , Interleucina-12/biosíntesis , Interleucina-17/biosíntesis , Cininas/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Mutantes , Reacción en Cadena de la Polimerasa , Receptor de Bradiquinina B2/metabolismo , Células TH1/metabolismo , Trypanosoma cruzi , Factor de Necrosis Tumoral alfa/biosíntesis
19.
Gastroenterology ; 132(7): 2533-41, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17570224

RESUMEN

BACKGROUND & AIMS: Increased intrahepatic resistance in cirrhosis is associated with reduced endothelial NO synthase (eNOS) activity and exacerbated by superimposed inflammation. NOSTRIN induces intracellular translocation of eNOS and reduces NO generation. Our aims were to quantify and compare hepatic expression of eNOS, NOSTRIN, NOSIP, and caveolin-1 in alcoholic cirrhosis with or without superimposed alcoholic hepatitis and in normal livers. METHODS: Biopsy specimens from 20 decompensated alcoholic cirrhotic patients with portal hypertension (10 with alcoholic hepatitis) and 6 normal livers were analyzed: real-time polymerase chain reaction for quantification of messenger RNA; Western blotting; and enzyme assays of eNOS in normal and diseased liver were performed. Localization and interaction of eNOS and NOSTRIN in liver was assessed by immunohistochemistry and co-immunoprecipitation. RESULTS: eNOS mRNA was significantly increased and eNOS activity decreased in alcoholic hepatitis patients, despite no differences in eNOS protein expression among the patients. Patients with alcoholic hepatitis had significantly higher hepatic levels of NOSTRIN and caveolin-1 mRNA compared with cirrhosis alone or normal biopsy specimens. A NOSTRIN splice variant, not present in normal tissue, was detected on mRNA and protein levels in all alcoholic patients. Coimmunoprecipitation demonstrated association among NOSTRIN, eNOS, and caveolin-1. CONCLUSIONS: An increase in mRNA and protein of NOSTRIN and its shortened variant in alcoholic hepatitis may partly account for the paradox of increased mRNA levels and normal protein expression but decreased enzymatic activity of eNOS in diseased liver. Such intracellular regulators of NO production may be important in the development of increased intrahepatic resistance in alcoholic hepatitis patients.


Asunto(s)
Expresión Génica , Variación Genética , Hepatitis Alcohólica/genética , Hepatitis Alcohólica/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hígado/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Células CHO , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caveolina 1/genética , Caveolina 1/metabolismo , Línea Celular Tumoral , Cricetinae , Cricetulus , Proteínas de Unión al ADN , Femenino , Humanos , Cirrosis Hepática/metabolismo , Masculino , Persona de Mediana Edad , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Mensajero/metabolismo , Distribución Tisular , Ubiquitina-Proteína Ligasas
20.
J Biol Chem ; 282(18): 13656-63, 2007 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-17329248

RESUMEN

Phosphodiesterase type 2A (PDE2A) hydrolyzes cyclic nucleotides cAMP and cGMP, thus efficiently controlling cNMP-dependent signaling pathways. PDE2A is composed of an amino-terminal region, two regulatory GAF domains, and a catalytic domain. Cyclic nucleotide hydrolysis is known to be activated by cGMP binding to GAF-B; however, other mechanisms may operate to fine-tune local cyclic nucleotide levels. In a yeast two-hybrid screening we identified XAP2, a crucial component of the aryl hydrocarbon receptor (AhR) complex, as a major PDE2A-interacting protein. We mapped the XAP2 binding site to the GAF-B domain of PDE2A. PDE assays with purified proteins showed that XAP2 binding does not change the enzymatic activity of PDE2A. To analyze whether PDE2A could affect the function of XAP2, we studied nuclear translocation of AhR, i.e. the master transcription factor controlling the expression of multiple detoxification genes. Notably, regulation of AhR target gene expression is initiated by tetrachlorodibenzodioxin (TCDD) binding to AhR and by a poorly understood cAMP-dependent pathway followed by the translocation of AhR from the cytosol into the nucleus. Binding of PDE2A to XAP2 inhibited TCDD- and cAMP-induced nuclear translocation of AhR in Hepa1c1c7 hepatocytes. Furthermore, PDE2A attenuated TCDD-induced transcription in reporter gene assays. We conclude that XAP2 targets PDE2A to the AhR complex, thereby restricting AhR mobility, possibly by a local reduction of cAMP levels. Our results provide first insights into the elusive cAMP-dependent regulation of AhR.


Asunto(s)
Proteínas Portadoras/metabolismo , Núcleo Celular/metabolismo , Chaperonas Moleculares/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Proteínas/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/fisiología , Animales , Células COS , Proteínas Portadoras/genética , Núcleo Celular/genética , Chlorocebus aethiops , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2 , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular , Chaperonas Moleculares/genética , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Mapeo Peptídico , Hidrolasas Diéster Fosfóricas/genética , Dibenzodioxinas Policloradas/farmacología , Estructura Terciaria de Proteína/genética , Proteínas/genética , Receptores de Hidrocarburo de Aril/genética , Teratógenos/farmacología , Transcripción Genética/efectos de los fármacos , Transcripción Genética/fisiología , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...