Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Pathol ; 248(1): 88-102, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30632166

RESUMEN

Ischemia/reperfusion (I/R) injury is a severe inflammatory insult associated with numerous pathologies, such as myocardial infarction, stroke and acute kidney injury. I/R injury is characterized by a rapid influx of activated neutrophils secreting toxic free radical species and degrading enzymes that can irreversibly damage the tissue, thus impairing organ functions. Significant efforts have been invested in identifying therapeutic targets to suppress neutrophil recruitment and activation post-I/R injury. In this context, pharmacological targeting of neutrophil elastase (NE) has shown promising anti-inflammatory efficacy in a number of experimental and clinical settings of I/R injury and is considered a plausible clinical strategy for organ care. However, the mechanisms of action of NE, and hence its inhibitors, in this process are not fully understood. Here we conducted a comprehensive analysis of the impact of NE genetic deletion on neutrophil infiltration in four murine models of I/R injury as induced in the heart, kidneys, intestine and cremaster muscle. In all models, neutrophil migration into ischemic regions was significantly suppressed in NE-/- mice as compared with wild-type controls. Analysis of inflamed cremaster muscle and mesenteric microvessels by intravital and confocal microscopy revealed a selective entrapment of neutrophils within venular walls, most notably at the level of the venular basement membrane (BM) following NE deletion/pharmacological blockade. This effect was associated with the suppression of NE-mediated remodeling of the low matrix protein expressing regions within the venular BM used by transmigrating neutrophils as exit portals. Furthermore, whilst NE deficiency led to reduced neutrophil activation and vascular leakage, levels of monocytes and prohealing M2 macrophages were reduced in tissues of NE-/- mice subjected to I/R. Collectively our results identify a vital and non-redundant role for NE in supporting neutrophil breaching of the venular BM post-I/R injury but also suggest a protective role for NE in promoting tissue repair. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Elastasa de Leucocito/fisiología , Neutrófilos/fisiología , Daño por Reperfusión/enzimología , Migración Transendotelial y Transepitelial/fisiología , Remodelación Vascular/fisiología , Animales , Membrana Basal/enzimología , Membrana Basal/patología , Membrana Basal/fisiopatología , Modelos Animales de Enfermedad , Eliminación de Gen , Riñón/irrigación sanguínea , Riñón/patología , Elastasa de Leucocito/deficiencia , Elastasa de Leucocito/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Infiltración Neutrófila/fisiología , Neutrófilos/enzimología , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Vénulas/enzimología , Vénulas/patología , Vénulas/fisiopatología
2.
Am J Physiol Heart Circ Physiol ; 316(5): H1065-H1075, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30681366

RESUMEN

The Notch ligand delta-like ligand 4 (Dll4), upregulated by VEGF, is a key regulator of vessel morphogenesis and function, controlling tip and stalk cell selection during sprouting angiogenesis. Inhibition of Dll4 results in hypersprouting, nonfunctional, poorly perfused vessels, suggesting a role for Dll4 in the formation of mature, reactive, functional vessels, with low permeability and able to restrict fluid and solute exchange. We tested the hypothesis that Dll4 controls transvascular fluid exchange. A recombinant protein expressing only the extracellular portion of Dll4 [soluble Dll4 (sDll4)] induced Notch signaling in endothelial cells (ECs), resulting in increased expression of vascular-endothelial cadherin, but not the tight junctional protein zonula occludens 1, at intercellular junctions. sDll4 decreased the permeability of FITC-labeled albumin across EC monolayers, and this effect was abrogated by coculture with the γ-secretase inhibitor N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester. One of the known molecular effectors responsible for strengthening EC-EC contacts is PKA, so we tested the effect of modulation of PKA on the sDll4-mediated reduction of permeability. Inhibition of PKA reversed the sDll4-mediated reduction in permeability and reduced expression of the Notch target gene Hey1. Knockdown of PKA reduced sDLL4-mediated vascular-endothelial cadherin junctional expression. sDll4 also caused a significant decrease in the hydraulic conductivity of rat mesenteric microvessels in vivo. This reduction was abolished upon coperfusion with the PKA inhibitor H89 dihydrochloride. These results indicate that Dll4 signaling through Notch activation acts through a cAMP/PKA pathway upon intercellular adherens junctions, but not tight junctions, to regulate endothelial barrier function. NEW & NOTEWORTHY Notch signaling reduces vascular permeability through stimulation of cAMP-dependent protein kinase A.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/farmacología , Proteínas de Unión al Calcio/farmacología , Permeabilidad Capilar/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Mesenterio/irrigación sanguínea , Receptores Notch/metabolismo , Sistemas de Mensajero Secundario/efectos de los fármacos , Uniones Adherentes/efectos de los fármacos , Uniones Adherentes/enzimología , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Masculino , Inhibidores de Proteínas Quinasas/farmacología , Ratas Wistar , Vénulas/efectos de los fármacos , Vénulas/enzimología
4.
Circ Res ; 110(8): 1057-66, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22415018

RESUMEN

RATIONALE: Intravascular neutrophil recruitment and activation are key pathogenic factors that contribute to vascular injury. Intravenous immunoglobulin (IVIG) has been shown to have a beneficial effect in systemic inflammatory disorders; however, the mechanisms underlying IVIG's inhibitory effect on neutrophil recruitment and activation are not understood. OBJECTIVE: We studied the mechanisms by which IVIG exerts protection from neutrophil-mediated acute vascular injury. METHODS AND RESULTS: We examined neutrophil behavior in response to IVIG in vivo, using real-time intravital microscopy. We found that an antibody that blocks both FcγRIII and its inhibitory receptor counterpart, FcγRIIB, abrogated the inhibitory effect of IVIG on leukocyte recruitment and heterotypic red blood cell (RBC) interactions with adherent leukocytes in wild-type mice. In the context of sickle cell disease, the blockade of both FcγRIIB and III abrogated the protective effect of IVIG on acute vaso-occlusive crisis caused by neutrophil recruitment and activation. Analysis of FcγRIIB- and FcγRIII-deficient mice revealed the predominant expression of FcγRIII on circulating neutrophils. FcγRIII mediated IVIG-triggered inhibition of leukocyte recruitment, circulating RBC capture, and enhanced Mac-1 activity, whereas FcγRIIB was dispensable. In addition, FcγRIII-induced IVIG anti-inflammatory activity in neutrophils was mediated by recruitment of Src homology 2 (SH2)-containing tyrosine phosphatase-1 (SHP-1). Indeed, the protective effect of IVIG on leukocyte recruitment and activation was abrogated in SHP-1-mutant mice. CONCLUSIONS: FcγRIII, a classic activating receptor, has an unexpected inhibitory role on neutrophil adhesion and activation via recruitment of SHP-1 in response to IVIG. Our results identify SHP-1 as a therapeutic target in neutrophil-mediated vascular injury.


Asunto(s)
Anemia de Células Falciformes/tratamiento farmacológico , Células Endoteliales/efectos de los fármacos , Inmunoglobulinas Intravenosas/farmacología , Músculo Liso/irrigación sanguínea , Activación Neutrófila/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Receptores de IgG/metabolismo , Enfermedades Vasculares/prevención & control , Anemia de Células Falciformes/enzimología , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/inmunología , Anemia de Células Falciformes/patología , Animales , Constricción Patológica , Células Endoteliales/enzimología , Células Endoteliales/inmunología , Células Endoteliales/patología , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Antígeno de Macrófago-1/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía por Video , Neutrófilos/enzimología , Neutrófilos/inmunología , Receptores de IgG/deficiencia , Receptores de IgG/genética , Factores de Tiempo , Enfermedades Vasculares/enzimología , Enfermedades Vasculares/genética , Enfermedades Vasculares/inmunología , Enfermedades Vasculares/patología , Vénulas/efectos de los fármacos , Vénulas/enzimología , Vénulas/inmunología
5.
Am J Physiol Heart Circ Physiol ; 305(10): H1484-93, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24043249

RESUMEN

Exogenously applied caveolin-1 scaffolding domain (CAV) has been shown to inhibit inflammatory mediator-induced nitric oxide (NO) production and NO-mediated increases in microvessel permeability. However, the effect of CAV on endothelial basal NO that prevents leukocyte adhesion remains unknown. This study aims to investigate the roles of exogenously applied CAV in endothelial basal NO production, leukocyte adhesion, and adhesion-induced changes in microvessel permeability. Experiments were conducted in individually perfused rat mesenteric venules. Microvessel permeability was determined by measuring hydraulic conductivity (Lp). NO was quantified with fluorescence imaging in DAF-2-loaded vessels. Perfusing venules with CAV inhibited basal NO production without affecting basal Lp. Resuming blood flow in CAV-perfused vessels significantly increased leukocyte adhesion. The firmly adherent leukocytes altered neither basal Lp nor adherens junction integrity. Increases in Lp occurred only upon formyl-Met-Leu-Phe application that induces release of reactive oxygen species from the adherent leukocytes. The application of NO synthase inhibitor showed similar results to CAV, and NO donor abolished CAV-mediated leukocyte adhesion. Immunofluorescence staining showed increases in binding of ICAM-1 to an adhesion-blocking antibody concurrent with a Src-dependent ICAM-1 phosphorylation following CAV perfusion. Pre-perfusing vessels with anti-ICAM-1 blocking antibody or a Src kinase inhibitor attenuated CAV-induced leukocyte adhesion. These results indicate that the application of CAV, in addition to preventing excessive NO-mediated permeability increases, also causes reduction of basal NO and promotes ICAM-1-mediated leukocyte adhesion through Src activation-mediated ICAM-1 phosphorylation. CAV-induced leukocyte adhesion was uncoupled from leukocyte oxidative burst and microvessel barrier function, unless in the presence of a secondary stimulation.


Asunto(s)
Caveolina 1/farmacología , Adhesión Celular/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Molécula 1 de Adhesión Intercelular/metabolismo , Leucocitos/efectos de los fármacos , Mesenterio/irrigación sanguínea , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Fragmentos de Péptidos/farmacología , Animales , Proteína con Homeodominio Antennapedia/farmacología , Permeabilidad Capilar/efectos de los fármacos , Endotelio Vascular/enzimología , Endotelio Vascular/inmunología , Activación Enzimática , Femenino , Técnica del Anticuerpo Fluorescente , Leucocitos/inmunología , Leucocitos/metabolismo , Microscopía Confocal , Microscopía Fluorescente , N-Formilmetionina Leucil-Fenilalanina/farmacología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo , Vénulas/efectos de los fármacos , Vénulas/enzimología , Vénulas/inmunología , Familia-src Quinasas/metabolismo
6.
Am J Physiol Heart Circ Physiol ; 305(4): H521-32, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23771693

RESUMEN

Previously we have shown that, unlike wild-type mice (WT), heme oxygenase-1 knockout (HO-1-/-) mice developed nitrate tolerance and were not protected from inflammation caused by ischemia-reperfusion (I/R) when preconditioned with a H2S donor. We hypothesized that stimulation (with BAY 41-2272) or activation (with BAY 60-2770) of soluble guanylate cyclase (sGC) would precondition HO-1-/- mice against an inflammatory effect of I/R and increase arterial nitrate responses. Intravital fluorescence microscopy was used to visualize leukocyte rolling and adhesion to postcapillary venules of the small intestine in anesthetized mice. Relaxation to ACh and BAY compounds was measured on superior mesenteric arteries isolated after I/R protocols. Preconditioning with either BAY compound 10 min (early phase) or 24 h (late phase) before I/R reduced postischemic leukocyte rolling and adhesion to sham control levels and increased superior mesenteric artery responses to ACh, sodium nitroprusside, and BAY 41-2272 in WT and HO-1-/- mice. Late-phase preconditioning with BAY 60-2770 was maintained in HO-1-/- and endothelial nitric oxide synthase knockout mice pretreated with an inhibitor (dl-propargylglycine) of enzymatically produced H2S. Pretreatment with BAY compounds also prevented the I/R increase in small intestinal TNF-α. We speculate that increasing sGC activity and related PKG acts downstream to H2S and disrupts signaling processes triggered by I/R in part by maintaining low cellular Ca²âº. In addition, BAY preconditioning did not increase sGC levels, yet increased the response to agents that act on reduced heme-containing sGC. Collectively these actions would contribute to increased nitrate sensitivity and vascular function.


Asunto(s)
Benzoatos/farmacología , Compuestos de Bifenilo/farmacología , Activadores de Enzimas/farmacología , Hemo-Oxigenasa 1/deficiencia , Hidrocarburos Fluorados/farmacología , Inflamación/prevención & control , Intestino Delgado/irrigación sanguínea , Isquemia/tratamiento farmacológico , Proteínas de la Membrana/deficiencia , Oclusión Vascular Mesentérica/tratamiento farmacológico , Pirazoles/farmacología , Piridinas/farmacología , Receptores Citoplasmáticos y Nucleares/agonistas , Daño por Reperfusión/prevención & control , Enfermedades Vasculares/tratamiento farmacológico , Animales , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Activación Enzimática , Guanilato Ciclasa/metabolismo , Hemo-Oxigenasa 1/genética , Sulfuro de Hidrógeno/metabolismo , Inflamación/enzimología , Inflamación/genética , Inflamación/fisiopatología , Mediadores de Inflamación/metabolismo , Isquemia/enzimología , Isquemia/genética , Isquemia/fisiopatología , Rodamiento de Leucocito/efectos de los fármacos , Proteínas de la Membrana/genética , Arteria Mesentérica Superior/efectos de los fármacos , Arteria Mesentérica Superior/enzimología , Arteria Mesentérica Superior/cirugía , Isquemia Mesentérica , Oclusión Vascular Mesentérica/enzimología , Oclusión Vascular Mesentérica/genética , Oclusión Vascular Mesentérica/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Daño por Reperfusión/enzimología , Daño por Reperfusión/genética , Daño por Reperfusión/fisiopatología , Transducción de Señal/efectos de los fármacos , Guanilil Ciclasa Soluble , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo , Enfermedades Vasculares/enzimología , Enfermedades Vasculares/genética , Enfermedades Vasculares/fisiopatología , Vasodilatación/efectos de los fármacos , Vénulas/efectos de los fármacos , Vénulas/enzimología
7.
Am J Physiol Heart Circ Physiol ; 301(3): H888-94, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21666111

RESUMEN

We recently demonstrated that preconditioning with an exogenous hydrogen sulfide donor (NaHS-PC) 24 h before ischemia and reperfusion (I/R) causes postcapillary venules to shift to an anti-inflammatory phenotype in C57BL/6J wild-type (WT) mice such that these vessels fail to support increases in postischemic leukocyte rolling (LR) and leukocyte adhesion (LA). The objective of the present study was to determine whether heme oxygenase-1 (HO-1) is a mediator of these anti-inflammatory effects noted during I/R in mice preconditioned with NaHS. Intravital fluorescence microscopy was used to visualize LR and LA in single postcapillary venules of the murine small intestine. I/R induced marked increases in LR and LA, effects that were prevented by NaHS-PC. Treatment with the HO inhibitor tin protoporphyrin IX, but not the inactive protoporphyrin CuPPIX, just before reperfusion prevented the anti-inflammatory effects of antecedent NaHS. The anti-inflammatory effects of NaHS-PC were mimicked by preconditioning with hemin, an agent that induces HO-1 expression. We then evaluated the effect of NaHS as a preconditioning stimulus in mice that were genetically deficient in HO-1 (HO-1(-/-) on an H129 background with appropriate WT strain controls). NaHS-PC was ineffective in HO-1(-/-) mice. Our work indicates that HO-1 serves as an effector of the anti-inflammatory effects of NaHS-PC during I/R 24 h later.


Asunto(s)
Antiinflamatorios/farmacología , Células Endoteliales/efectos de los fármacos , Hemo-Oxigenasa 1/metabolismo , Sulfuro de Hidrógeno/metabolismo , Intestino Delgado/irrigación sanguínea , Proteínas de la Membrana/metabolismo , Daño por Reperfusión/prevención & control , Sulfuros/farmacología , Análisis de Varianza , Animales , Antiinflamatorios/metabolismo , Adhesión Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Células Endoteliales/enzimología , Células Endoteliales/inmunología , Inhibidores Enzimáticos/farmacología , Hemo-Oxigenasa 1/antagonistas & inhibidores , Hemo-Oxigenasa 1/deficiencia , Hemo-Oxigenasa 1/genética , Rodamiento de Leucocito/efectos de los fármacos , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Metaloporfirinas/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Fenotipo , Protoporfirinas/farmacología , Daño por Reperfusión/enzimología , Daño por Reperfusión/genética , Daño por Reperfusión/inmunología , Sulfuros/metabolismo , Factores de Tiempo , Vénulas/efectos de los fármacos , Vénulas/enzimología , Vénulas/inmunología
8.
Microvasc Res ; 81(1): 108-16, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20923679

RESUMEN

Venous hypertension is associated with microvascular inflammation, restructuring, and apoptosis, but the cellular and molecular mechanisms underlying these events remain uncertain. In the present study, we tested the hypothesis that elevated venous pressure and reduction of shear stress induce elevated enzymatic activity. This activity in turn may affect endothelial surface receptors and promote their dysfunction. Using a rodent model for venous hypertension using acute venular occlusion, microzymographic techniques for enzyme detection, and immunohistochemistry for receptor labeling, we found increased activity of the matrix metalloproteases (MMPs) -1, -8, and -9 and tissue inhibitors of metalloproteases (TIMPs) -1 and -2 in both high- and low-pressure regions. In this short time frame, we also observed that elevated venule pressure led to two different fates for the vascular endothelial growth factor receptor-2 (VEGFR2); in higher-pressure upstream regions, some animals exhibited higher VEGFR2 expression, while others displayed lower levels upstream compared to their downstream counterparts with lower pressure. VEGFR2 expression was, on average, more pronounced upon application of MMP inhibitor, suggesting possible cleavage of the receptor by activated enzymes in this model. We conclude that venous pressure elevation increases enzymatic activity which may contribute to inflammation and endothelial dysfunction associated with this disease by influencing critical surface receptors.


Asunto(s)
Endotelio Vascular/enzimología , Endotelio Vascular/fisiopatología , Metaloproteinasas de la Matriz/metabolismo , Oclusión Vascular Mesentérica/enzimología , Venas Mesentéricas/enzimología , Animales , Biocatálisis/efectos de los fármacos , Dipéptidos/farmacología , Células Endoteliales/enzimología , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Hipertensión/enzimología , Hipertensión/metabolismo , Hipertensión/fisiopatología , Leucocitos/enzimología , Masculino , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Metaloproteinasa 8 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz , Oclusión Vascular Mesentérica/metabolismo , Oclusión Vascular Mesentérica/fisiopatología , Venas Mesentéricas/metabolismo , Venas Mesentéricas/fisiopatología , Ratas , Ratas Wistar , Reperfusión , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Vénulas/enzimología , Vénulas/metabolismo , Vénulas/fisiopatología
9.
J Immunol ; 182(9): 5461-8, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19380794

RESUMEN

High endothelial venules (HEVs) are specialized blood vessels of secondary lymphoid organs composed of endothelial cells with a characteristic cuboidal morphology. Lymphocytes selectively adhere to and migrate across HEVs to initiate immune responses. In this study, we established a novel transgenic mouse line expressing Cre recombinase under the transcriptional control of the gene encoding HEV-expressed sulfotransferase, N-acetylglucosamine-6-O-sulfotransferase 2 (GlcNAc6ST-2), using bacterial artificial chromosome recombineering. Crossing these transgenic mice with the ROSA26 reporter strain, which expresses lacZ following Cre-mediated recombination, and staining the resulting progeny with 5-bromo-4-chloro-5-indolyl-beta-D-galactoside indicated that Cre recombinase was specifically expressed in mAb MECA79-reactive HEVs in secondary lymphoid organs but not in any other blood vessels of the transgenic mice. The expression of Cre recombinase correlated with a developmental switch, from immature, mAb MECA367-reactive HEVs to mature, mAb MECA79-reactive HEVs in neonatal lymph nodes. In addition to the HEVs, Cre recombinase was also strongly expressed in the colonic villi, which recapitulated the intrinsic expression of GlcNAc6ST-2 as confirmed in GlcNAc6ST-2(GFP/GFP) knock-in mice and by RT-PCR. Furthermore, treatment with an antimicrobial agent revealed that the colonic expression of Cre recombinase in the transgenic mice was regulated by commensal bacteria in the colon. In addition, Cre recombinase was expressed in a small subset of cells in the brain, testis, stomach, small intestine, and lung. In view of the restricted expression of Cre recombinase, this transgenic mouse line should be useful for elucidating tissue-specific gene functions using the Cre/loxP system.


Asunto(s)
Endotelio Linfático/inmunología , Endotelio Linfático/metabolismo , Técnicas de Sustitución del Gen/métodos , Sulfotransferasas/genética , Sulfotransferasas/metabolismo , Vénulas/inmunología , Vénulas/metabolismo , Animales , Endotelio Linfático/enzimología , Femenino , Regulación del Desarrollo de la Expresión Génica , Integrasas/biosíntesis , Integrasas/genética , Integrasas/metabolismo , Mucosa Intestinal/enzimología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Recombinación Genética/inmunología , Sulfotransferasas/biosíntesis , Vénulas/enzimología , Carbohidrato Sulfotransferasas
10.
Am J Physiol Regul Integr Comp Physiol ; 297(4): R968-77, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19625688

RESUMEN

It has been suggested that nitric oxide (NO) stimulates the cyclooxygenase (COX)-dependent mechanisms in the ocular vasculature; however, the importance of the pathway in regulating retinal circulation in vivo remains to be elucidated. Therefore, we investigated the role of COX-dependent mechanisms in NO-induced vasodilation of retinal blood vessels in thiobutabarbital-anesthetized rats with and without neuronal blockade (tetrodotoxin treatment). Fundus images were captured with a digital camera that was equipped with a special objective lens. The retinal vascular response was assessed by measuring changes in diameter of the retinal blood vessel. The localization of COX and soluble guanylyl cyclase in rat retina was examined using immunohistochemistry. The NO donors (sodium nitroprusside and NOR3) increased the diameter of the retinal blood vessels but decreased systemic blood pressure in a dose-dependent manner. Treatment of rats with indomethacin, a nonselective COX inhibitor, or SC-560, a selective COX-1 inhibitor, markedly attenuated the vasodilation of retinal arterioles, but not the depressor response, to the NO donors. However, both the vascular responses to NO donors were unaffected by the selective COX-2 inhibitors NS-398 and nimesulide. Indomethacin did not change the retinal vascular and depressor responses to hydralazine, 8-(4-chlorophenylthio)-guanosine-3', 5'-cyclic monophosphate (a membrane-permeable cGMP analog) and 8-(4-chlorophenylthio)-adenosine-3', 5'-cyclic monophosphate (a membrane-permeable cAMP analog). Treatment with SQ 22536, an adenylyl cyclase inhibitor, but not ODQ, a soluble guanylyl cyclase inhibitor, significantly attenuated the NOR3-induced vasodilation of retinal arterioles. The COX-1 immunoreactivity was found in retinal blood vessels. The retinal blood vessel was faintly stained for soluble guanylyl cyclase, although the apparent immunoreactivities on mesenteric and choroidal blood vessels were observed. These results suggest that NO exerts a substantial part of its dilatory effect via a mechanism that involves COX-1-dependent pathway in rat retinal vasculature.


Asunto(s)
Ciclooxigenasa 1/metabolismo , Proteínas de la Membrana/metabolismo , Óxido Nítrico/metabolismo , Vasos Retinianos/enzimología , Vasodilatación , Inhibidores de Adenilato Ciclasa , Adenilil Ciclasas/metabolismo , Anestésicos Locales/administración & dosificación , Animales , Arteriolas/enzimología , Presión Sanguínea , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/administración & dosificación , Inhibidores de la Ciclooxigenasa/administración & dosificación , Guanilato Ciclasa/antagonistas & inhibidores , Guanilato Ciclasa/metabolismo , Frecuencia Cardíaca , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Donantes de Óxido Nítrico/administración & dosificación , Ratas , Ratas Wistar , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/metabolismo , Vasos Retinianos/efectos de los fármacos , Guanilil Ciclasa Soluble , Factores de Tiempo , Vasodilatación/efectos de los fármacos , Vasodilatadores/administración & dosificación , Vénulas/enzimología
11.
J Vasc Res ; 46(5): 397-405, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19155631

RESUMEN

BACKGROUND: It has been shown that increases in intraluminal flow elicit dilation in venules, but the mediation of response is not yet clarified. We hypothesized that - in addition to nitric oxide (NO) and dilator prostaglandins (PGI(2)/ PGE(2)) - thromboxane A(2) (TxA(2)) contributes to the mediation of flow-induced responses of venules. METHODS AND RESULTS: Isolated rat gracilis muscle venules (259 +/- 11 microm at 10 mm Hg) dilated as a function of intraluminal flow, which was augmented in the presence of the TxA(2) receptor antagonist SQ 29,548 or the TxA(2) synthase inhibitor ozagrel. In the presence of SQ 29,548, indomethacin or Nomega-nitro-L-arginine methyl-ester decreased flow-induced dilations, whereas in their simultaneous presence dilations were abolished. The selective cyclooxygenase (COX) 1 inhibitor SC 560 reduced, whereas the selective COX-2 inhibitor NS 398 enhanced flow-induced dilations. Immunohistochemistry showed that both COX-1 and COX-2 are present in the wall of venules. CONCLUSION: In skeletal muscle venules, increases in intraluminal flow elicit production of constrictor TxA(2), in addition to the dilator NO and PGI(2)/PGE(2), with an overall effect of limited dilation. These mediators are likely to have important roles in the multiple feedback regulation of wall shear stress in venules during changes in blood flow velocity and/or viscosity.


Asunto(s)
Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Músculo Esquelético/irrigación sanguínea , Flujo Sanguíneo Regional/fisiología , Tromboxano A2/metabolismo , Animales , Velocidad del Flujo Sanguíneo/fisiología , Viscosidad Sanguínea/fisiología , Compuestos Bicíclicos Heterocíclicos con Puentes , Dinoprostona/metabolismo , Inhibidores Enzimáticos/farmacología , Epoprostenol/metabolismo , Ácidos Grasos Insaturados , Hidrazinas/farmacología , Masculino , Metacrilatos/farmacología , Músculo Esquelético/fisiología , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/metabolismo , Prostaglandina H2/metabolismo , Ratas , Ratas Wistar , Estrés Mecánico , Vénulas/enzimología
12.
J Leukoc Biol ; 80(6): 1491-9, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16997858

RESUMEN

During inflammation, leukocytes roll along the wall of postcapillary venules scanning the surface for immobilized CXCL1, a chemokine that triggers firm adhesion by activating CXCR2 on the neutrophil. PI-3K are signaling molecules important in cellular processes, ranging from cellular differentiation to leukocyte migration. PI-3Kgamma can be activated directly by the betagamma dimer of heterotrimeric G proteins coupled to CXCR2. Here, we used in vivo and ex vivo intravital microscopy models to test the role of PI-3Kgamma in leukocyte arrest. PI-3Kgamma null mice showed an 80% decrease in CXCL1-induced leukocyte adhesion in venules of the exteriorized mouse cremaster muscle. In wild-type mice, rolling leukocytes showed rapid and sustained adhesion, but in PI-3Kgamma(-/-) mice, adhesion was not triggered at all or was transient, suggesting that absence of PI-3Kgamma interferes with integrin bond strengthening. Wild-type mice reconstituted with PI-3Kgamma null bone marrow showed a 50% decrease in CXCL1-induced leukocyte adhesion. In a blood-perfused micro-flow chamber, leukocytes from PI-3Kgamma(-/-) mice showed a defect in adhesion on a P-selectin/ICAM-1/CXCL1 substrate, indicating that leukocyte PI-3Kgamma was required for adhesion. The adhesion defect in PI-3Kgamma(-/-) mice was as severe as that in mice lacking LFA-1, the major integrin responsible for neutrophil adhesion. We conclude that the gamma isoform of PI-3K must be functional in leukocytes to allow efficient adhesion from rolling in response to chemokine stimulation.


Asunto(s)
Quimiocinas CXC/metabolismo , Rodamiento de Leucocito/fisiología , Leucocitos/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Quimiocina CXCL1 , Quimiocinas CXC/farmacología , Fosfatidilinositol 3-Quinasa Clase Ib , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Isoenzimas/deficiencia , Isoenzimas/metabolismo , Rodamiento de Leucocito/efectos de los fármacos , Leucocitos/citología , Ratones , Ratones Noqueados , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/citología , Músculo Esquelético/enzimología , Selectina-P/metabolismo , Fosfatidilinositol 3-Quinasas/deficiencia , Vénulas/citología , Vénulas/enzimología
13.
Br J Pharmacol ; 116(6): 2710-4, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8590994

RESUMEN

1. The effects of aminoguanidine on neutrophil adherence to venules and on the diameter of arterioles in the mesenteric vascular bed of the pentobarbitone-anaesthetized rat have been compared with those of the nitric oxide synthase inhibitor, NG-nitro-L-arginine methyl ester (L-NAME). 2. Administration of L-NAME (1-10 mg kg-1, i.v.) caused a dose-dependent increase in leukocyte adherence and a reduction in leukocyte rolling velocity in postcapillary venules of the rat mesentery over 1 h. 3. Likewise, aminoguanidine (10-100 mg kg-1, i.v.) dose-dependently increased leukocyte adherence and decreased leukocyte rolling velocity over 1 h. 4. Both L-NAME and aminoguanidine caused a dose-dependent reduction in mesenteric arteriolar diameter and an increase in systemic arterial blood pressure. 5. The effects of aminoguanidine (50 mg kg-1, i.v.) on leukocyte adherence, arteriolar diameter and on blood pressure were significantly reversed by pretreatment with L-arginine (300 mg kg-1, i.v.). 6. These findings indicate that, like L-NAME, aminoguanidine can acutely promote leukocyte adherence to the mesenteric venular wall and reduce arteriolar diameter. Moreover, these acute effects were reversed by L-arginine, suggesting they are mediated through inhibition of constitutive NO synthase.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Guanidinas/farmacología , Venas Mesentéricas/efectos de los fármacos , Venas Mesentéricas/enzimología , Neutrófilos/efectos de los fármacos , Óxido Nítrico Sintasa/antagonistas & inhibidores , Animales , Arginina/análogos & derivados , Arginina/farmacología , Arteriolas/anatomía & histología , Arteriolas/efectos de los fármacos , Arteriolas/enzimología , Presión Sanguínea/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Masculino , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/enzimología , NG-Nitroarginina Metil Éster , Neutrófilos/citología , Neutrófilos/fisiología , Ratas , Ratas Wistar , Vénulas/anatomía & histología , Vénulas/efectos de los fármacos , Vénulas/enzimología
14.
Placenta ; 21(7): 621-7, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10985964

RESUMEN

Aminopeptidase A (AP-A EC 3.4.11.7), which is a membrane-bound zinc metalloprotease, is present in the placenta. AP-A selectively hydrolyzes N-terminal glutamyl and aspartyl residues and cleaves angiotensin II to form angiotensin III. To determine the role of placental aminopeptidase A under physiological and pathological conditions, we evaluated its immunolocalization and enzymatic activities in the placenta. AP-A was localized in the basal zone of the syncytiotrophoblast, in the membranes of the cytotrophoblast, and in fetal arterioles and venules within the stem villi. AP-A activity in the microsomal fraction of placental villi seemed to be remained essentially constant throughout gestation. The renin-angiotensin system is considered to be accelerated in pre-eclampsia. This AP-A activity was higher in pre-eclampsia (2.86+/-0.30 nmol beta NA/mg protein/h) than in uncomplicated pregnancy from 28 to 41 weeks of gestation (2.08+/-0.18 nmol beta NA/mg protein/h). Angiotensin II evoked AP-A activity in first trimester trophoblast, and Losartan and PD 123177 in combination significantly inhibited this induction of AP-A activity. The results of immunohistochemical evaluation and enzymatic activity suggested that placental aminopeptidase A may play a role as a component of the barrier of angiotensin II between mother and fetus.


Asunto(s)
Aminopeptidasas/metabolismo , Angiotensina II/metabolismo , Placenta/enzimología , Adulto , Aminopeptidasas/análisis , Angiotensina III/metabolismo , Arteriolas/enzimología , Células Cultivadas , Femenino , Feto/irrigación sanguínea , Edad Gestacional , Glutamil Aminopeptidasa , Humanos , Preeclampsia/enzimología , Embarazo , Sistema Renina-Angiotensina/fisiología , Distribución Tisular , Trofoblastos/enzimología , Vénulas/enzimología
15.
J Appl Physiol (1985) ; 87(3): 873-81, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10484552

RESUMEN

Although the effects of ischemia-reperfusion have received considerable attention, few studies have directly evaluated the microcirculatory response to systemic hypoxia. The overall objective of this study was to assess the effect of environmental hypoxia on adhesive interactions of circulating leukocytes with rat mesenteric venules by using intravital microscopy. Experiments were designed to 1) characterize the adhesive interactions of circulating leukocytes to venules during acute hypoxia produced by a reduction in inspired PO(2), 2) evaluate the role of nitric oxide in these adhesive interactions, 3) determine whether the effect of hypoxia on leukocyte adhesive interactions differs between acclimatized and nonacclimatized rats, and 4) assess whether compensatory changes in nitric oxide formation contribute to this difference. The results showed that acute hypoxia promotes leukocyte-endothelial adherence in mesenteric venules of nonacclimatized rats. The mechanism of this response is consistent with depletion of nitric oxide within the microcirculation. In contrast, no leukocyte-endothelial adherence occurred during hypoxia in rats acclimatized to hypobaric hypoxia. The results are consistent with increased nitric oxide formation due to expression of inducible nitric oxide synthase during the acclimatization period. Further studies are needed to establish the cause of nitric oxide depletion during acute hypoxia as well as to define the compensatory responses that attenuate hypoxia-induced leukocyte-endothelial adherence in the microvasculature of acclimatized rats.


Asunto(s)
Aclimatación/fisiología , Hipoxia de la Célula/fisiología , Leucocitos/fisiología , Venas Mesentéricas/fisiología , Animales , Análisis de los Gases de la Sangre , Adhesión Celular/fisiología , Leucocitos/enzimología , Masculino , Venas Mesentéricas/citología , Venas Mesentéricas/enzimología , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa de Tipo II , Ratas , Ratas Sprague-Dawley , Vénulas/citología , Vénulas/enzimología , Vénulas/fisiología
16.
Brain Res ; 792(2): 353-7, 1998 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-9593993

RESUMEN

The goal of this study was to examine the effect of lipopoly saccharide on the permeability of the blood-brain barrier and reactivity of cerebral arterioles. We examined the pial microcirculation in rats using intravital fluorescence microscopy. Permeability of the blood-brain barrier (clearance of fluorescent-labeled dextran; molecular weight 10,000 Da; FITC-dextran-10K) and diameter of pial arterioles were measured in the absence and presence of topical application of vehicle (saline) or lipopolysaccharide (200 ng/ml). During superfusion with vehicle, clearance of FITC-dextran-10K from pial vessels was minimal, and diameter of pial arterioles remained constant. Topical application of lipopolysaccharide (200 ng/ml) produced an increase in clearance of FITC-dextran-10K and dilated pial arterioles. To determine whether lipopolysaccharide-induced changes in permeability of the blood-brain barrier and dilatation of cerebral arterioles was related to the synthesis/release of inducible nitric oxide, we examined the effects of aminoguanidine (0.5 mM). Aminoguanidine inhibited lipopolysaccharide-induced increases in permeability of the blood-brain barrier and dilatation of cerebral arterioles. The findings of the present study suggest that lipopolysaccharide increases permeability of the blood-brain barrier and diameter of pial arterioles via the activation of inducible nitric oxide synthase.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Circulación Cerebrovascular/efectos de los fármacos , Dextranos/farmacocinética , Fluoresceína-5-Isotiocianato/análogos & derivados , Lipopolisacáridos/farmacología , Óxido Nítrico Sintasa/fisiología , Animales , Arteriolas/efectos de los fármacos , Arteriolas/enzimología , Infecciones Bacterianas/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/enzimología , Inhibidores Enzimáticos/farmacología , Fluoresceína-5-Isotiocianato/farmacocinética , Guanidinas/farmacología , Masculino , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II , Piamadre/irrigación sanguínea , Piamadre/enzimología , Ratas , Ratas Endogámicas WF , Vénulas/efectos de los fármacos , Vénulas/enzimología
17.
Brain Res Bull ; 58(3): 271-8, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12128152

RESUMEN

This study was designed to test the hypothesis that the sphingomyelin-ceramide signaling pathway may be important in proinflammatory-like responses in the intact brain. Effects of neutral sphingomyelinase (N-SMase), ceramide analogs, phosphorylcholine and ceramide metabolites were studied on rat brain cerebral (cortical) venule lumen sizes, leukocyte rolling, velocity and endothelial cell wall adhesion, microvessel permeability, microvessel rupture and focal hemorrhages using in vivo high resolution TV microscopy. Perivascular and close intra-arterial administration of N-SMase, C(2)-, C(8)-, and C(16)-ceramide, but not either phosphorylcholine, C(6)-ceramide, nervonic (C(24):1) ceramide, lignoceric (C(24):0) ceramide, C(8)-ceramide-1-phosphate, glucosylceramide or 1-0-acylceramide, resulted in potent, concentration-dependent constriction (and spasm) of cortical venules, followed by increased leukocyte rolling, decreased leukocyte velocities, increased leukocyte-endothelial wall adhesion, increased venular wall permeability, postcapillary venule rupture and, often, micro-hemorrhaging at high concentrations; angiotensin II, serotonin and PGF(2alpha) didn't demonstrate these characteristics. Pretreatment with either one of three different antioxidants, including inhibitors of NF-kappaB activation, or two different Ca(2+) channel blockers either prevented or attenuated the adverse venular effects of N-SMase and the ceramides. Likewise, pretreatment with either a PKCalpha-beta antagonist or a MAP kinase antagonist also attenuated the adverse venular effects. These results suggest that N-SMase and several ceramides can result in potent venular cerebrovasospasm, leukocyte-endothelial chemoattraction, and microvessel wall permeability changes in the intact rat brain. These proinflammatory-like actions suggest that N-SMase and ceramides could produce brain-vascular damage by reperfusion injury triggering lipid peroxidation, release of reactive oxygen species and activation of diverse signaling pathways: PKCalpha-beta isozymes, MAP kinase and NF-kappaB.


Asunto(s)
Ceramidas/metabolismo , Venas Cerebrales/enzimología , Encefalitis/enzimología , Endotelio Vascular/enzimología , Leucocitos/enzimología , Esfingomielina Fosfodiesterasa/metabolismo , Vénulas/enzimología , Angiotensina II/metabolismo , Angiotensina II/farmacología , Animales , Lesiones Encefálicas/enzimología , Lesiones Encefálicas/fisiopatología , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/fisiología , Comunicación Celular/efectos de los fármacos , Comunicación Celular/fisiología , Ceramidas/farmacología , Venas Cerebrales/efectos de los fármacos , Venas Cerebrales/fisiopatología , Dinoprost/metabolismo , Dinoprost/farmacología , Relación Dosis-Respuesta a Droga , Encefalitis/fisiopatología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiopatología , Leucocitos/efectos de los fármacos , Masculino , Ratas , Ratas Wistar , Serotonina/metabolismo , Serotonina/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Esfingomielina Fosfodiesterasa/farmacología , Esfingomielinas/metabolismo , Accidente Cerebrovascular/enzimología , Accidente Cerebrovascular/fisiopatología , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología , Vénulas/efectos de los fármacos , Vénulas/fisiopatología
18.
Eur J Gastroenterol Hepatol ; 11(10): 1113-9, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10524640

RESUMEN

OBJECTIVES: The distribution of the intestinal vascular lesions and their relation with the fibrinolysis process are poorly known in Crohn's disease (CD). The mediators of the plasminogen activator system, namely urokinase-type plasminogen activator (u-PA), tissue-type plasminogen activator (t-PA) and plasminogen activator inhibitor type-1 (PAI-1), are a key complex involved in fibrinolysis. The aims of this study were: (1) to further define vascular lesions and their distribution in the intestine; and (2) to study concomitantly the qualitative in situ expression and the levels of u-PA, t-PA and PAI-1 in the ileum of patients with CD. PATIENTS AND METHODS: Histological, immunohistochemical and ultrastructural studies of vascular lesions in the resected ileum of 27 patients with CD were performed and compared with 36 control patients. Levels of u-PA, t-PA and PAI-1 measured by ELISA methods were compared in healthy and inflamed ileal tissues of 17 patients with CD. RESULTS: Acute vascular lesions involving mainly serosal venules and capillaries were present in 63% of patients with CD vs 3/36 controls and were associated with PAI-1 expression. They were prominent on the mesenteric border beneath macroscopically normal mucosa. In contrast, chronic vascular lesions were present in all layers beneath mucosal ulcerations, where a significant increase of PAI-1 levels was found. CONCLUSIONS: These results suggest that vascular involvement associated with abnormalities of PAI-1 expression is an early and widespread event in CD. Their prominence on the mesenteric border might explain the characteristic location of CD ulceration along the mesenteric margin.


Asunto(s)
Enfermedad de Crohn/patología , Íleon/enzimología , Íleon/patología , Inflamación/patología , Activadores Plasminogénicos/metabolismo , Adolescente , Adulto , Anciano , Biopsia , Capilares/enzimología , Capilares/patología , Capilares/ultraestructura , Niño , Enfermedad de Crohn/enzimología , Femenino , Humanos , Íleon/irrigación sanguínea , Inmunohistoquímica , Inflamación/enzimología , Masculino , Persona de Mediana Edad , Inhibidor 1 de Activador Plasminogénico/biosíntesis , Activador de Tejido Plasminógeno/biosíntesis , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis , Vénulas/enzimología , Vénulas/patología , Vénulas/ultraestructura
19.
Anat Embryol (Berl) ; 199(1): 29-34, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9924932

RESUMEN

Pial microvessels have several important blood-brain barrier (BBB) characteristics in common with cerebral microvessels, despite lacking their astrocytic ensheathment. We have therefore determined whether they have the same distribution of two enzymes, gamma-glutamyl transpeptidase (GGTP) and alkaline phosphatase, both of which are known to be astrocyte-dependent. GGTP was absent from all rat pial microvessels but strongly present in brain cortical capillaries. Alkaline phosphatase was heterogeneously expressed in pial microvessels, including capillaries, but strongly positive in brain cortical capillaries. Diffusible, inductive factors produced by astrocytes could account for these differences in enzyme distribution between the two vessel types. Furthermore, differences in expression between the two markers may reflect their differing sensitivities to the astrocytic factors. Caution is urged in the common usage of the pial microvessel as a model system in BBB studies.


Asunto(s)
Fosfatasa Alcalina/biosíntesis , Capilares/enzimología , Corteza Cerebral/irrigación sanguínea , Piamadre/irrigación sanguínea , gamma-Glutamiltransferasa/biosíntesis , Animales , Arteriolas/enzimología , Corteza Cerebral/enzimología , Endotelio Vascular/enzimología , Endotelio Vascular/ultraestructura , Histocitoquímica , Microscopía Electrónica , Pericitos/enzimología , Pericitos/ultraestructura , Piamadre/enzimología , Ratas , Ratas Sprague-Dawley , Vénulas/enzimología
20.
J Histochem Cytochem ; 60(5): 397-407, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22260995

RESUMEN

Two members of the N-acetylglucosamine-6-O-sulfotransferase (GlcNAc6ST) family, GlcNAc6ST-1 and GlcNAc6ST-2, function in the biosynthesis of 6-sulfo sialyl Lewis X-capped glycoproteins expressed on high endothelial venules (HEVs) in secondary lymphoid organs. Thus, both enzymes play a critical role in L-selectin-expressing lymphocyte homing. Human GlcNAc6ST-1 is encoded by a 1593-bp open reading frame exhibiting two 5' in-frame methionine codons spaced 141 bp apart. Both resemble the consensus sequence for translation initiation. Thus, it has been hypothesized that both long and short forms of GlcNAc6ST-1 may be present, although endogenous expression of either form has not been confirmed in humans. Here, the authors developed an antibody recognizing amino acid residues between the first two human GlcNAc6ST-1 methionines. This antibody specifically recognizes the long form of the enzyme, a finding validated by Western blot analysis and immunofluorescence cytochemistry of HeLa cells misexpressing long and/or short forms of human GlcNAc6ST-1. Using this antibody, the authors carried out immunofluorescence histochemistry of human lymph node tissue sections and found endogenous expression of the long form of the enzyme in human tissue, predominantly in the trans-Golgi network of endothelial cells that form HEVs.


Asunto(s)
Endotelio Vascular/enzimología , Sulfotransferasas/metabolismo , Vénulas/enzimología , Secuencia de Aminoácidos , Anticuerpos , Secuencia de Bases , Western Blotting , Endotelio Vascular/ultraestructura , Técnica del Anticuerpo Fluorescente , Aparato de Golgi/enzimología , Células HeLa , Humanos , Ganglios Linfáticos/irrigación sanguínea , Datos de Secuencia Molecular , Sulfotransferasas/inmunología , Vénulas/ultraestructura , Carbohidrato Sulfotransferasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA