Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Am J Physiol Heart Circ Physiol ; 315(4): H978-H988, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30028198

RESUMEN

Neonatal asphyxia leads to cerebrovascular disease and neurological complications via a mechanism that may involve oxidative stress. Carbon monoxide (CO) is an antioxidant messenger produced via a heme oxygenase (HO)-catalyzed reaction. Cortical astrocytes are the major cells in the brain that express constitutive HO-2 isoform. We tested the hypothesis that CO, produced by astrocytes, has cerebroprotective properties during neonatal asphyxia. We developed a survival model of prolonged asphyxia in newborn pigs that combines insults of severe hypoxia, hypercapnia, and acidosis while avoiding extreme hypotension and cerebral blood flow reduction. During the 60-min asphyxia, CO production by brain and astrocytes was continuously elevated. Excessive formation of reactive oxygen species during asphyxia/reventilation was potentiated by the HO inhibitor tin protoporphyrin, suggesting that endogenous CO has antioxidant effects. Cerebral vascular outcomes tested 24 and 48 h after asphyxia demonstrated the sustained impairment of cerebral vascular responses to astrocyte- and endothelium-specific vasodilators. Postasphyxia cerebral vascular dysfunction was aggravated in newborn pigs pretreated with tin protoporphyrin to inhibit brain HO/CO. The CO donor CO-releasing molecule-A1 (CORM-A1) reduced brain oxidative stress during asphyxia/reventilation and prevented postasphyxia cerebrovascular dysfunction. The antioxidant and antiapoptotic effects of HO/CO and CORM-A1 were confirmed in primary cultures of astrocytes from the neonatal pig brain exposed to glutamate excitotoxicity. Overall, prolonged neonatal asphyxia leads to neurovascular injury via an oxidative stress-mediated mechanism that is counteracted by an astrocyte-based constitutive antioxidant HO/CO system. We propose that gaseous CO or CO donors can be used as novel approaches for prevention of neonatal brain injury caused by prolonged asphyxia. NEW & NOTEWORTHY Asphyxia in newborn infants may lead to lifelong neurological disabilities. Using the model of prolonged asphyxia in newborn piglets, we propose novel antioxidant therapy based on systemic administration of low doses of a carbon monoxide donor that prevent loss of cerebral blood flow regulation and may improve the neurological outcome of asphyxia.


Asunto(s)
Arteriolas/efectos de los fármacos , Asfixia Neonatal/tratamiento farmacológico , Astrocitos/efectos de los fármacos , Boranos/farmacología , Dióxido de Carbono/metabolismo , Carbonatos/farmacología , Circulación Cerebrovascular/efectos de los fármacos , Trastornos Cerebrovasculares/prevención & control , Fármacos Neuroprotectores/farmacología , Piamadre/irrigación sanguínea , Animales , Animales Recién Nacidos , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Arteriolas/metabolismo , Arteriolas/fisiopatología , Asfixia Neonatal/complicaciones , Asfixia Neonatal/metabolismo , Asfixia Neonatal/fisiopatología , Astrocitos/metabolismo , Astrocitos/patología , Velocidad del Flujo Sanguíneo , Células Cultivadas , Trastornos Cerebrovasculares/etiología , Trastornos Cerebrovasculares/metabolismo , Trastornos Cerebrovasculares/fisiopatología , Modelos Animales de Enfermedad , Femenino , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1/metabolismo , Masculino , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Sus scrofa , Factores de Tiempo , Vasodilatación/efectos de los fármacos
2.
Am J Physiol Heart Circ Physiol ; 311(5): H1202-H1213, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27591217

RESUMEN

Epileptic seizures in neonates cause cerebrovascular injury and impairment of cerebral blood flow (CBF) regulation. In the bicuculline model of seizures in newborn pigs, we tested the hypothesis that selective head cooling prevents deleterious effects of seizures on cerebral vascular functions. Preventive or therapeutic ictal head cooling was achieved by placing two head ice packs during the preictal and/or ictal states, respectively, for the ∼2-h period of seizures. Head cooling lowered the brain and core temperatures to 25.6 ± 0.3 and 33.5 ± 0.1°C, respectively. Head cooling had no anticonvulsant effects, as it did not affect the bicuculline-evoked electroencephalogram parameters, including amplitude, duration, spectral power, and spike frequency distribution. Acute and long-term cerebral vascular effects of seizures in the normothermic and head-cooled groups were tested during the immediate (2-4 h) and delayed (48 h) postictal periods. Seizure-induced cerebral vascular injury during the immediate postictal period was detected as terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling-positive staining of cerebral arterioles and a surge of brain-derived circulating endothelial cells in peripheral blood in the normothermic group, but not in the head-cooled groups. During the delayed postictal period, endothelium-dependent cerebral vasodilator responses were greatly reduced in the normothermic group, indicating impaired CBF regulation. Preventive or therapeutic ictal head cooling mitigated the endothelial injury and greatly reduced loss of postictal cerebral vasodilator functions. Overall, head cooling during seizures is a clinically relevant approach to protecting the neonatal brain by preventing cerebrovascular injury and the loss of the endothelium-dependent control of CBF without reducing epileptiform activity.


Asunto(s)
Arteriolas/fisiopatología , Arterias Cerebrales/fisiopatología , Circulación Cerebrovascular , Trastornos Cerebrovasculares/fisiopatología , Cabeza , Hipotermia Inducida/métodos , Convulsiones/fisiopatología , Vasodilatación/fisiología , Animales , Animales Recién Nacidos , Bicuculina/toxicidad , Trastornos Cerebrovasculares/etiología , Trastornos Cerebrovasculares/prevención & control , Convulsivantes/toxicidad , Electroencefalografía , Células Endoteliales/citología , Femenino , Etiquetado Corte-Fin in Situ , Masculino , Convulsiones/inducido químicamente , Convulsiones/complicaciones , Porcinos
3.
Am J Physiol Cell Physiol ; 304(11): C1105-15, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23576575

RESUMEN

In cerebral microvascular endothelial cells (CMVEC) of newborn pigs, glutamate at excitotoxic concentrations (mM) causes apoptosis mediated by reactive oxygen species (ROS). Carbon monoxide (CO) produced by CMVEC or delivered by a CO-releasing molecule, CORM-A1, has antioxidant properties. We tested the hypothesis that CORM-A1 prevents cerebrovascular endothelial barrier dysfunction caused by glutamate excitotoxicity. First, we identified the glutamate receptors (GluRs) and enzymatic sources of ROS involved in the mechanism of endothelial apoptosis. In glutamate-exposed CMVEC, ROS formation and apoptosis were blocked by rotenone, 2-thenoyltrifluoroacetone (TTFA), and antimycin, indicating that mitochondrial complexes I, II, and III are the major sources of oxidative stress. Agonists of ionotropic GluRs (iGluRs) N-methyl-D-aspartate (NMDA), cis-ACPD, AMPA, and kainate increased ROS production and apoptosis, whereas iGluR antagonists exhibited antiapoptotic properties, suggesting that iGluRs mediate glutamate-induced endothelial apoptosis. The functional consequences of endothelial injury were tested in the model of blood-brain barrier (BBB) composed of CMVEC monolayer on semipermeable membranes. Glutamate and iGluR agonists reduced transendothelial electrical resistance and increased endothelial paracellular permeability to 3-kDa dextran. CORM-A1 exhibited potent antioxidant and antiapoptotic properties in CMVEC and completely prevented BBB dysfunction caused by glutamate and iGluR agonists. Overall, the endothelial component of the BBB is a cellular target for excitotoxic glutamate that, via a mechanism involving a iGluR-mediated activation of mitochondrial ROS production and apoptosis, leads to BBB opening that may be prevented by the antioxidant and antiapoptotic actions of CORMs. Antioxidant CORMs therapy may help preserve BBB functional integrity in neonatal cerebrovascular disease.


Asunto(s)
Apoptosis/fisiología , Barrera Hematoencefálica/metabolismo , Boranos/metabolismo , Carbonatos/metabolismo , Células Endoteliales/metabolismo , Estrés Oxidativo/fisiología , Receptores Ionotrópicos de Glutamato/metabolismo , Animales , Barrera Hematoencefálica/patología , Encéfalo/metabolismo , Encéfalo/patología , Fragmentación del ADN , Células Endoteliales/patología , Aminoácidos Excitadores/metabolismo , Aminoácidos Excitadores/toxicidad , Femenino , Ácido Glutámico/metabolismo , Ácido Glutámico/toxicidad , Masculino , Especies Reactivas de Oxígeno/metabolismo , Porcinos
4.
Am J Physiol Heart Circ Physiol ; 302(11): H2257-66, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22467311

RESUMEN

In newborn pigs, vasodilation of pial arterioles in response to glutamate is mediated via carbon monoxide (CO), a gaseous messenger endogenously produced from heme degradation by a heme oxygenase (HO)-catalyzed reaction. We addressed the hypothesis that ionotropic glutamate receptors (iGluRs), including N-methyl-D-aspartic acid (NMDA)- and 2-amino-3-(5-methyl-3-oxo-1,2-oxazol-4-yl) propanoic acid (AMPA)/kainate-type receptors, expressed in cortical astrocytes mediate glutamate-induced astrocyte HO activation that leads to cerebral vasodilation. Acute vasoactive effects of topical iGluR agonists were determined by intravital microscopy using closed cranial windows in anesthetized newborn pigs. iGluR agonists, including NMDA, (±)1-aminocyclopentane-cis-1,3-dicarboxylic acid (cis-ACPD), AMPA, and kainate, produced pial arteriolar dilation. Topical L-2-aminoadipic acid, a gliotoxin that selectively disrupts glia limitans, reduced vasodilation caused by iGluR agonists, but not by hypercapnia, bradykinin, or sodium nitroprusside. In freshly isolated and cultured cortical astrocytes constitutively expressing HO-2, iGluR agonists NMDA, cis-ACPD, AMPA, and kainate rapidly increased CO production two- to threefold. Astrocytes overexpressing inducible HO-1 had high baseline CO but were less sensitive to glutamate stimulation of CO production when compared with HO-2-expressing astrocytes. Glutamate-induced astrocyte HO-2-mediated CO production was inhibited by either the NMDA receptor antagonist (R)-3C4HPG or the AMPA/kainate receptor antagonist DNQX. Accordingly, either antagonist abolished pial arteriolar dilation in response to glutamate, NMDA, and AMPA, indicating functional interaction among various subtypes of astrocytic iGluRs in response to glutamate stimulation. Overall, these data indicate that the astrocyte component of the neurovascular unit is responsible for the vasodilation response of pial arterioles to topically applied glutamate via iGluRs that are functionally linked to activation of constitutive HO in newborn piglets.


Asunto(s)
Arteriolas/efectos de los fármacos , Astrocitos/metabolismo , Monóxido de Carbono/metabolismo , Circulación Cerebrovascular/fisiología , Glutamatos/farmacología , Receptores de Glutamato/metabolismo , Vasodilatación/efectos de los fármacos , Animales , Animales Recién Nacidos , Arteriolas/fisiología , Astrocitos/citología , Astrocitos/efectos de los fármacos , Células Cultivadas , Femenino , Hemo Oxigenasa (Desciclizante)/metabolismo , Técnicas In Vitro , Ácido Kaínico/farmacología , Masculino , Modelos Animales , N-Metilaspartato/farmacología , Receptores de Glutamato/efectos de los fármacos , Porcinos , Vasodilatación/fisiología , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/farmacología
5.
J Thromb Thrombolysis ; 33(3): 218-29, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22238031

RESUMEN

Insulin inhibits platelet aggregation through nitric oxide synthesis by stimulating platelet insulin activated nitric oxide synthase. Impaired platelet insulin activated nitric oxide synthase in acute myocardial infarction (AMI) patients had been reported and thus our aim was to identify and isolate the factors impairing insulin activated nitric oxide in acute myocardial infarction patients' plasma and study its effect on platelets aggregation in vitro. The insulin activated nitric oxide synthase inhibitor was identified as a protein and was purified from the plasma of AMI subjects using DEAE cellulose and Sephadex G-50 column, molecular weight determined by SDS-PAGE, nitric oxide quantified by methaemoglobin method, inhibitor protein quantified in plasma by immunoblot and ELISA, platelet aggregation studies done using an aggregometer, thromboxane-A2 in the platelets determined by radioimmunoassay, (125)I-insulin radioligand binding studies done using normal subject platelets. The purified nitric oxide synthase inhibitor protein was ~66 kDa, concentration in AMI subjects' plasma varied from 114 to 9,090 µM and was undetected in normal subjects' plasma. The inhibitor protein competes with insulin for insulin receptor binding sites. The Incubation of the normal subject PRP with 5.0 µM inhibitor for 30 min followed by 0.4 µM ADP addition caused platelet aggregation in vitro, 130 µM aspirin or 400 µU insulin/ml addition was able to abrogate 0.4 µM ADP induced platelet aggregation even in the presence of 5.0 µM inhibitor. A potent inhibitory protein against insulin activated nitric oxide synthase in platelets appears in circulation of AMI subjects impairing nitric oxide production, potentiating ADP induced platelet aggregation and increasing the thromboxane-A2 level in platelets.


Asunto(s)
Proteínas Sanguíneas/aislamiento & purificación , Proteínas Sanguíneas/metabolismo , Insulina/metabolismo , Infarto del Miocardio/sangre , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Adulto , Anciano , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Insulina/farmacología , Masculino , Persona de Mediana Edad , Infarto del Miocardio/metabolismo , Unión Proteica/fisiología
6.
Am J Physiol Cell Physiol ; 300(2): C256-65, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21123734

RESUMEN

We investigated the role of reactive oxygen species (ROS) in promoting cell survival during oxidative stress induced by the inflammatory mediator tumor necrosis factor-α (TNF-α) in cerebral microvascular endothelial cells (CMVEC) from newborn piglets. Nox4 is the major isoform of NADPH oxidase responsible for TNF-α-induced oxidative stress and apoptosis in CMVEC. We present novel data that Nox4 NADPH oxidase-derived ROS also initiate a cell survival mechanism by increasing production of a gaseous antioxidant mediator carbon monoxide (CO) by constitutive heme oxygenase-2 (HO-2). TNF-α rapidly enhanced endogenous CO production in a superoxide- and NADPH oxidase-dependent manner in CMVEC with innate, but not with small interfering RNA (siRNA)-downregulated Nox4 activity. CORM-A1, a CO-releasing compound, inhibited Nox4-mediated ROS production and enhanced cell survival in TNF-α-challenged CMVEC. The ROS-induced CO-mediated survival mechanism requires functional interactions between the protein kinase B/Akt and extracellular signal-related kinase (ERK)/p38 MAPK signaling pathways activated by TNF-α. In Akt siRNA-transfected CMVEC and in cells with pharmacologically inhibited Akt, Erk1/2, and p38 mitogen-activated protein kinase (MAPK) activities, CORM-A1 was no longer capable of blocking Nox4 activation and apoptosis caused by TNF-α. Overall, Nox4 NADPH oxidase-derived ROS initiate both death and survival pathways in TNF-α-challenged CMVEC. The ROS-dependent cell survival pathway is mediated by an endogenous antioxidant CO, which inhibits Nox4 activation via a mechanism that includes Akt, ERK1/2, and p38 MAPK signaling pathways. The ability of CO to inhibit TNF-α-induced ERK1/2 and p38 MAPK activities in an Akt-dependent manner appears to be the key element in ROS-dependent survival of endothelial cells during TNF-α-mediated brain inflammatory disease.


Asunto(s)
Apoptosis , Encéfalo/metabolismo , Monóxido de Carbono/metabolismo , Células Endoteliales/metabolismo , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Animales Recién Nacidos , Boranos/administración & dosificación , Encéfalo/efectos de los fármacos , Carbonatos/administración & dosificación , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hemo Oxigenasa (Desciclizante)/metabolismo , Humanos , Isoenzimas/metabolismo , Microvasos/efectos de los fármacos , Microvasos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Porcinos/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
Am J Physiol Heart Circ Physiol ; 301(2): H428-33, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21572018

RESUMEN

Glutamate-stimulated, astrocyte-derived carbon monoxide (CO) causes cerebral arteriole dilation by activating smooth muscle cell large-conductance Ca(2+)-activated K(+) channels. Here, we examined the hypothesis that glutamate activates heme oxygenase (HO)-2 and CO production via the intracellular Ca(2+) concentration ([Ca(2+)](i))/Ca(2+)-calmodulin signaling pathway in newborn pig astrocytes. The major findings are: 1) glutamate stimulated Ca(2+) transients and increased steady-state [Ca(2+)](i) in cerebral cortical astrocytes in primary culture, 2) in astrocytes permeabilized with ionomycin, elevation of [Ca(2+)](i) concentration-dependently increased CO production, 3) glutamate did not affect CO production at any [Ca(2+)](i) when the [Ca(2+)](i) was held constant, 4) thapsigargin, a sarco/endoplasmic reticulum Ca(2+)-ATPase blocker, decreased basal CO production and blocked glutamate-induced increases in CO, and 5) calmidazolium, a calmodulin inhibitor, blocked CO production induced by glutamate and by [Ca(2+)](i) elevation. Taken together, our data are consistent with the hypothesis that glutamate elevates [Ca(2+)](i) in astrocytes, leading to Ca(2+)- and calmodulin-dependent HO-2 activation, and CO production.


Asunto(s)
Astrocitos/metabolismo , Señalización del Calcio , Monóxido de Carbono/metabolismo , Ácido Glutámico/metabolismo , Análisis de Varianza , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Calmodulina/antagonistas & inhibidores , Calmodulina/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Hemo Oxigenasa (Desciclizante)/metabolismo , Imidazoles/farmacología , Ionomicina/farmacología , Ionóforos/farmacología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/antagonistas & inhibidores , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Porcinos , Tapsigargina/farmacología , Factores de Tiempo , Regulación hacia Arriba
8.
Am J Physiol Heart Circ Physiol ; 300(2): H440-7, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21131483

RESUMEN

Hydrogen sulfide (H2S) is a gaseous signaling molecule that appears to be involved in numerous biological processes, including regulation of blood pressure and vascular tone. The present study is designed to address the hypothesis that H2S is a functionally significant, endogenous dilator in the newborn cerebrovascular circulation. In vivo experiments were conducted using newborn pigs with surgically implanted, closed, cranial windows. Topical application of H2S concentration-dependently (10(-6) to 2×10(-4) M) dilated pial arterioles. This dilation was blocked by glibenclamide (10(-6) M). L-cysteine, the substrate of the H2S-producing enzymes cystathionine γ-lyase (CSE) and cystathionine ß-synthase (CBS), also dilated pial arterioles. The dilation to L-cysteine was blocked by the CSE inhibitor d,l-propargylglycine (PPG, 10 mM) but was unaffected by the CBS inhibitor amino-oxyacetate (AOA, 1 mM). Western blots detected CSE, but not CBS, in cerebral microvessels, whereas CBS is detected in brain parenchyma. Immunohistological CSE expression is predominantly vascular while CBS is expressed mainly in neurons and astrocytes. L-cysteine (5 mM) increased H2S concentration in cerebrospinal fluid (CSF), measured by GC-MS, from 561±205 to 2,783±818 nM before but not during treatment with PPG (1,030±70 to 622±78 nM). Dilation to hypercapnia was inhibited by PPG but not AOA. Hypercapnia increased CSF H2S concentration from 763±243 to 4,337±1789 nM before but not during PPG treatment (357±178 vs. 425±217 nM). These data show that H2S is a dilator of the newborn cerebral circulation and that endogenous CSE can produce sufficient H2S to decrease vascular tone. H2S appears to be a physiologically significant dilator in the cerebral circulation.


Asunto(s)
Animales Recién Nacidos/fisiología , Capilares/efectos de los fármacos , Circulación Cerebrovascular/efectos de los fármacos , Sulfuro de Hidrógeno/metabolismo , Sulfuro de Hidrógeno/farmacología , Músculo Liso Vascular/efectos de los fármacos , Animales , Arteriolas/efectos de los fármacos , Arteriolas/fisiología , Química Encefálica/fisiología , Cisteína/farmacología , Gliburida/farmacología , Hipercapnia/fisiopatología , Hipoxia/fisiopatología , Inmunohistoquímica , Isoproterenol/farmacología , Canales KATP/antagonistas & inhibidores , Tono Muscular/efectos de los fármacos , Nitroprusiato/farmacología , Porcinos , Vasodilatación/efectos de los fármacos
9.
Am J Physiol Gastrointest Liver Physiol ; 299(1): G186-95, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20378826

RESUMEN

Recent studies showed that c-Src and phosphatidylinositol 3 (PI3) kinase mediate the oxidative stress-induced disruption of tight junctions in Caco-2 cell monolayers. The present study evaluated the roles of PI3 kinase and Src kinase in the oxidative stress-induced activation of focal adhesion kinase (FAK) and acceleration of cell migration. Oxidative stress, induced by xanthine and xanthine oxidase system, rapidly increased phosphorylation of FAK on Y397, Y925, and Y577 in the detergent-insoluble and soluble fractions and increased its tyrosine kinase activity. The PI3 kinase inhibitors, wortmannin and LY294002, and the Src kinase inhibitor, 4-amino-5[chlorophyll]-7-[t-butyl]pyrazolo[3-4-d]pyrimidine, attenuated tyrosine phosphorylation of FAK. Oxidative stress induced phosphorylation of c-Src on Y418 by a PI3 kinase-dependent mechanism, whereas oxidative stress-induced activation of PI3 kinase was independent of Src kinase activity. Hydrogen peroxide accelerated Caco-2 cell migration in a concentration-dependent manner. Promotion of cell migration by hydrogen peroxide was attenuated by LY294002 and PP2. Reduced expression of FAK by siRNA attenuated hydrogen peroxide-induced acceleration of cell migration. The expression of constitutively active c-Src(Y527F) enhanced cell migration, whereas the expression of dominant negative c-Src(K296R/Y528F) attenuated hydrogen peroxide-induced stimulation of cell migration. Oxidative stress-induced activation of c-Src and FAK was associated with a rapid increase in the tyrosine phosphorylation and the levels of paxillin and p130(CAS) in actin-rich, detergent-insoluble fractions. This study shows that oxidative stress activates FAK and accelerates cell migration in an intestinal epithelium by a PI3 kinase- and Src kinase-dependent mechanism.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Peróxido de Hidrógeno/farmacología , Mucosa Intestinal/efectos de los fármacos , Oxidantes/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Células CACO-2 , Pollos , Proteína Sustrato Asociada a CrK/metabolismo , Relación Dosis-Respuesta a Droga , Activación Enzimática , Quinasa 1 de Adhesión Focal/genética , Humanos , Mucosa Intestinal/enzimología , Ratones , Mutación , Estrés Oxidativo/efectos de los fármacos , Paxillin/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Interferencia de ARN , Factores de Tiempo , Tirosina , Vinculina/metabolismo , Xantina/metabolismo , Xantina Oxidasa/metabolismo
10.
Am J Physiol Heart Circ Physiol ; 298(6): H1687-98, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20363895

RESUMEN

Circulating endothelial cells (CECs) are nonhematopoetic mononuclear cells in peripheral blood that are dislodged from injured vessels during cardiovascular disease, systemic vascular disease, and inflammation. Their occurrence during cerebrovascular insults has not been previously described. Epileptic seizures cause the long-term loss of cerebrovascular endothelial dilator function. We hypothesized that seizures cause endothelial sloughing from cerebral vessels and the appearance of brain-derived CECs (BCECs), possible early indicators of cerebral vascular damage. Epileptic seizures were induced by bicuculline in newborn pigs; venous blood was then sampled during a 4-h period. CECs were identified in the fraction of peripheral blood mononuclear cells by the expression of endothelial antigens (CD146, CD31, and endothelial nitric oxide synthase) and by Ulex europeaus lectin binding. In control animals, few CECs were detected. Seizures caused a time-dependent increase in CECs 2-4 h after seizure onset. Seizure-induced CECs coexpress glucose transporter-1, a blood-brain barrier-specific glucose transporter, indicating that these cells originate in the brain vasculature and are thus BCECs. Seizure-induced BCECs cultured in EC media exhibited low proliferative potential and abnormal cell contacts. BCEC appearance during seizures was blocked by a CO-releasing molecule (CORM-A1) or cobalt protoporphyrin (heme oxygenase-1 inducer), which prevented apoptosis in cerebral arterioles and the loss of cerebral vascular endothelial function during the late postictal period. These findings suggest that seizure-induced BCECs are injured ECs dislodged from cerebral microvessels during seizures. The correlation between the appearance of BCECs in peripheral blood, apoptosis in cerebral vessels, and the loss of postictal cerebral vascular function suggests that BCECs are early indicators of late cerebral vascular damage.


Asunto(s)
Arterias Cerebrales/patología , Trastornos Cerebrovasculares/sangre , Trastornos Cerebrovasculares/diagnóstico , Endotelio Vascular/patología , Epilepsia/sangre , Microvasos/patología , Animales , Apoptosis/fisiología , Bicuculina/efectos adversos , Antígeno CD146/metabolismo , Comunicación Celular/fisiología , Proliferación Celular , Células Cultivadas , Trastornos Cerebrovasculares/etiología , Convulsivantes/efectos adversos , Modelos Animales de Enfermedad , Endotelio Vascular/inmunología , Epilepsia/inducido químicamente , Epilepsia/complicaciones , Femenino , Inmunofenotipificación , Masculino , Valor Predictivo de las Pruebas , Porcinos
11.
Biochem J ; 393(Pt 1): 69-77, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16134968

RESUMEN

The MAPK (mitogen-activated protein kinase) pathway is a major intracellular signalling pathway involved in EGF (epithelial growth factor) receptor-mediated cell growth and differentiation. A novel function of MAPK activity in the mechanism of EGF-mediated protection of TJs (tight junctions) from H2O2 was examined in Caco-2 cell monolayers. EGF-mediated prevention of H2O2-induced increase in paracellular permeability was associated with the prevention of H2O2-induced Tyr-phosphorylation, Thr-dephosphorylation and cellular redistribution of occludin and ZO-1 (zonula occludin-1). EGF also prevented H2O2-induced disruption of the actin cytoskeleton and the dissociation of occludin and ZO-1 from the actin-rich detergent-insoluble fractions. MEK (MAPK/ERK kinase, where ERK stands for extracellular signal related kinase) inhibitors, PD98059 and U0126, completely blocked these protective effects of EGF on TJs. EGF rapidly increased the levels of phosphorylated MEK (p-MEK) in detergent-soluble fractions and phosphorylated ERK (p-ERK) in detergent-insoluble fractions. p-ERK was colocalized and co-immunoprecipitated with occludin. GST (glutathione S-transferase) pull-down assay showed that the C-terminal tail of occludin binds to p-ERK in Caco-2 cell extracts. Pair-wise binding studies using recombinant proteins demonstrated that ERK1 directly interacts with the C-terminal tail of occludin. Therefore the present study shows that ERK interacts with the C-terminal region of occludin and mediates the prevention of H2O2-induced disruption of TJs by EGF.


Asunto(s)
Factor de Crecimiento Epidérmico/farmacología , Peróxido de Hidrógeno/farmacología , Proteínas de la Membrana/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Células CACO-2 , Humanos , Inulina/metabolismo , Ocludina , Estrés Oxidativo , Fosfoproteínas/metabolismo , Unión Proteica , Proteína de la Zonula Occludens-1
12.
Physiol Rep ; 3(3)2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25804265

RESUMEN

Neonatal seizures have been associated with cerebrovascular endothelial injury and neurological disabilities. In a piglet model, the long-term loss of endothelial regulation of cerebral blood flow coincides with the surge of brain-derived circulating endothelial cells (BCECs) in blood. We hypothesized that BCECs could serve as a noninvasive biomarker of cerebrovascular injury in neonates with seizures. In a prospective pilot feasibility study, we enrolled newborn infants with confirmed diagnoses of perinatal asphyxia and intraventricular hemorrhage (IVH); both are commonly associated with seizures. Infants without clinical evidence of cerebrovascular injuries were representative of the control group. BCECs were detected in the CD45-negative fraction of peripheral blood mononuclear cells by coexpression of CD31 (common endothelial antigen) and GLUT1 (blood-brain barrier antigen) via automated flow cytometry method. In Infants with asphyxia (n = 12) and those with IVH grade III/IV (n = 5), the BCEC levels were 9.9 ± 0.9% and 19.0 ± 2.0%, respectively. These levels were significantly higher than the control group (n = 27), 0.9 ± 0.2%, P < 0.001. BCECs in infants with cerebrovascular insults with documented clinical seizures (n = 10; 16.8 ± 1.3%) were significantly higher than infants with cerebrovascular insults with subclinical or no seizures (n = 7; 9.5 ± 1.2%); P < 0.001. BCEC levels decreased with seizure control. BCECs levels were elevated in infants with seizures caused by severe IVH and perinatal asphyxia. We suggest that monitoring BCEC levels in peripheral blood can potentially offer a biological marker that reflects cerebrovascular insult and recovery. Further studies with a larger number of patients are required to support these findings.

13.
J Cereb Blood Flow Metab ; 32(6): 1024-34, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22354150

RESUMEN

Postictal cerebrovascular dysfunction is an adverse effect of seizures in newborn piglets. The brain heme oxygenase (HO) provides protection against cerebrovascular dysfunction. We investigated the contribution of reactive oxygen species (ROS) to seizure-induced vascular damage and the mechanism of HO vasoprotection. In a bicuculline model of seizures, we addressed the hypotheses: (1) seizures increase brain ROS; (2) ROS contribute to cerebral vascular dysfunction; (3) ROS initiate a vasoprotective mechanisms by activating endogenous HO; and (4) HO products have antioxidant properties. As assessed by dihydroethidium oxidation (ox-DHE), seizures increased ROS in cerebral vessels and cortical astrocytes; ox-DHE elevation was prevented by tiron and apocynin. An HO inhibitor, tin protoporphyrin, potentiated, whereas an HO-1 inducer, cobalt protoporphyrin, blocked seizure-induced increase in DHE oxidation. Heme oxygenase products carbon monoxide (CO) (CORM-A1) and bilirubin attenuated ox-DHE elevation during seizures. Antioxidants tiron and bilirubin prevented the loss of postictal cerebrovascular dilations to bradykinin, glutamate, and sodium nitroprusside. Tiron and apocynin abrogated activation of the brain HO during seizures. Overall, these data suggest that long-term adverse cerebrovascular effects of seizures are attributed to oxidative stress. On the other hand, seizure-induced ROS are required for activation of the endogenous antioxidant HO/CO/bilirubin system that alleviates oxidative stress-induced loss of postictal cerebrovascular function in piglets.


Asunto(s)
Antioxidantes/metabolismo , Bilirrubina/metabolismo , Monóxido de Carbono/metabolismo , Circulación Cerebrovascular , Hemo Oxigenasa (Desciclizante)/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Convulsiones/metabolismo , Animales , Astrocitos/metabolismo , Bradiquinina/metabolismo , Inhibidores Enzimáticos/farmacología , Ácido Glutámico/metabolismo , Metaloporfirinas/farmacología , Estrés Oxidativo , Protoporfirinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Convulsiones/fisiopatología , Porcinos
14.
Am J Physiol Cell Physiol ; 296(3): C422-32, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19118162

RESUMEN

Inflammatory brain disease may damage cerebral vascular endothelium leading to cerebral blood flow dysregulation. The proinflammatory cytokine TNF-alpha causes oxidative stress and apoptosis in cerebral microvascular endothelial cells (CMVEC) from newborn pigs. We investigated contribution of major cellular sources of reactive oxygen species to endothelial inflammatory response. Nitric oxide synthase and xanthine oxidase inhibitors (N(omega)-nitro-l-arginine and allopurinol) had no effect, while mitochondrial electron transport inhibitors (CCCP, 2-thenoyltrifluoroacetone, and rotenone) attenuated TNF-alpha-induced superoxide (O(2)(*-)) and apoptosis. NADPH oxidase inhibitors (diphenylene iodonium and apocynin) greatly reduced TNF-alpha-evoked O(2)(*-) generation and apoptosis. TNF-alpha rapidly increased NADPH oxidase activity in CMVEC. Nox4, the cell-specific catalytic subunit of NADPH oxidase, is highly expressed in CMVEC, contributes to basal O(2)(*-) production, and accounts for a burst of oxidative stress in response to TNF-alpha. Nox4 small interfering RNA, but not Nox2, knockdown prevented oxidative stress and apoptosis caused by TNF-alpha in CMVEC. Nox4 is colocalized with HO-2, the constitutive isoform of heme oxygenase (HO), which is critical for endothelial protection against TNF-alpha toxicity. The products of HO activity, bilirubin and carbon monoxide (CO, as a CO-releasing molecule, CORM-A1), inhibited Nox4-generated O(2)(*-) and apoptosis caused by TNF-alpha stimulation. We conclude that Nox4 is the primary source of inflammation- and TNF-alpha-induced oxidative stress leading to apoptosis in brain endothelial cells. The ability of CO and bilirubin to combat TNF-alpha-induced oxidative stress by inhibiting Nox4 activity and/or by O(2)(*-) scavenging, taken together with close intracellular compartmentalization of HO-2 and Nox4 in cerebral vascular endothelium, may contribute to HO-2 cytoprotection against inflammatory cerebrovascular disease.


Asunto(s)
Apoptosis , Corteza Cerebral/irrigación sanguínea , Células Endoteliales/enzimología , NADPH Oxidasas/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Bilirrubina/metabolismo , Monóxido de Carbono/metabolismo , Células Cultivadas , Citoprotección , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Inhibidores Enzimáticos/farmacología , Técnica del Anticuerpo Fluorescente , Depuradores de Radicales Libres/farmacología , Hemo Oxigenasa (Desciclizante)/metabolismo , Inflamación/metabolismo , Inflamación/patología , Microvasos/enzimología , Microvasos/patología , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/genética , Estrés Oxidativo/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Porcinos
15.
Am J Physiol Cell Physiol ; 290(5): C1399-410, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16371440

RESUMEN

In cerebral circulation, epileptic seizures associated with excessive release of the excitatory neurotransmitter glutamate cause endothelial injury. Heme oxygenase (HO), which metabolizes heme to a vasodilator, carbon monoxide (CO), and antioxidants, biliverdin/bilirubin, is highly expressed in cerebral microvessels as a constitutive isoform, HO-2, whereas the inducible form, HO-1, is not detectable. Using cerebral vascular endothelial cells from newborn pigs and HO-2-knockout mice, we addressed the hypotheses that 1) glutamate induces oxidative stress-related endothelial death by apoptosis, and 2) HO-1 and HO-2 are protective against glutamate cytotoxicity. In cerebral endothelial cells, glutamate (0.1-2.0 mM) increased formation of reactive oxygen species, including superoxide radicals, and induced major keystone events of apoptosis, such as NF-kappaB nuclear translocation, caspase-3 activation, DNA fragmentation, and cell detachment. Glutamate-induced apoptosis was greatly exacerbated in HO-2 gene-deleted murine cerebrovascular endothelial cells and in porcine cells with pharmacologically inhibited HO-2 activity. Glutamate toxicity was prevented by superoxide dismutase, suggesting apoptotic changes are oxidative stress related. When HO-1 was pharmacologically upregulated by cobalt protoporphyrin, apoptotic effects of glutamate in cerebral endothelial cells were completely prevented. Glutamate-induced reactive oxygen species production and apoptosis were blocked by a CO-releasing compound, CORM-A1 (50 microM), and by bilirubin (1 microM), consistent with the antioxidant and cytoprotective roles of the end products of HO activity. We conclude that both HO-1 and HO-2 have anti-apoptotic effects against oxidative stress-related glutamate toxicity in cerebral vascular endothelium. Although HO-1, when induced, provides powerful protection, HO-2 is an essential endogenous anti-apoptotic factor against glutamate toxicity in the cerebral vascular endothelium.


Asunto(s)
Encéfalo/irrigación sanguínea , Citoprotección/fisiología , Células Endoteliales/fisiología , Ácido Glutámico/administración & dosificación , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1/metabolismo , Estrés Oxidativo/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Encéfalo/citología , Encéfalo/fisiología , Células Cultivadas , Citoprotección/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo/efectos de los fármacos , Porcinos
16.
Am J Physiol Heart Circ Physiol ; 291(6): H2897-904, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16891404

RESUMEN

Astrocytes can act as intermediaries between neurons and cerebral arterioles to regulate vascular tone in response to neuronal activity. Release of glutamate from presynaptic neurons increases blood flow to match metabolic demands. CO is a gasotransmitter that can be related to neural function and blood flow regulation in the brain. The present study addresses the hypothesis that glutamatergic stimulation promotes perivascular astrocyte CO production and pial arteriolar dilation in the newborn brain. Experiments used anesthetized newborn pigs with closed cranial windows, piglet astrocytes, and cerebrovascular endothelial cells in primary culture and immunocytochemical visualization of astrocytic markers. Pial arterioles and arteries of newborn pigs are ensheathed by astrocytes visualized by glial fibrillary acidic protein staining. Treatment (2 h) of astrocytes in culture with L-2-alpha-aminoadipic acid (L-AAA), followed by 14 h in toxin free medium, dose-dependently increased cell detachment, suggesting injury. Conversely, 16 h of continuous exposure to L-AAA caused no decrease in endothelial cell attachment. In vivo, topical L-AAA (2 mM, 5 h) disrupted the cortical glia limitans histologically. Such treatment also eliminated pial arteriolar dilation to the astrocyte-dependent dilator ADP and to glutamate but not to isoproterenol or CO. Glutamate stimulated CO production by the brain surface that also was abolished following L-AAA. In contrast, tetrodotoxin blocked dilation to N-methyl-D-aspartate but not to glutamate, isoproterenol, or CO or the glutamate-induced increase in CO. The concurrent loss of CO production and pial arteriolar dilation to glutamate following astrocyte injury suggests astrocytes may employ CO as a gasotransmitter for glutamatergic cerebrovascular dilation.


Asunto(s)
Arteriolas/fisiología , Astrocitos/metabolismo , Monóxido de Carbono/metabolismo , Ácido Glutámico/fisiología , Ácido 2-Aminoadípico/farmacología , Anestésicos Locales/farmacología , Animales , Animales Recién Nacidos , Astrocitos/citología , Astrocitos/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Antagonistas de Aminoácidos Excitadores/farmacología , Piamadre/irrigación sanguínea , Receptores de Glutamato/efectos de los fármacos , Receptores de Glutamato/fisiología , Porcinos , Tetrodotoxina/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología
17.
Am J Physiol Cell Physiol ; 291(5): C897-908, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16822952

RESUMEN

Tumor necrosis factor-alpha (TNF-alpha) causes oxidative stress and apoptosis in a variety of cell types. Heme oxygenase (HO) degrades heme to bilirubin, an antioxidant, and carbon monoxide (CO), a cell cycle modulator, and a vasodilator. Newborn pig cerebral microvascular endothelial cells (CMVEC) highly express constitutive HO-2. We investigated the role of HO-2 in protection against TNF-alpha-induced apoptosis in cerebral vascular endothelium. In CMVEC from mice and newborn pigs, 15 ng/ml TNF-alpha alone, or with 10 microg/ml cycloheximide (CHX) caused apoptosis detected by nuclear translocation of p65 NF-kappaB, caspase-3 activation, DNA fragmentation, cell-cell contact destabilization, and cell detachment. TNF-alpha did not induce HO-1 expression in CMVEC. CMVEC from HO-2 knockout mice showed greater sensitivity to apoptosis caused by serum deprivation and TNF-alpha than did wild-type mice. TNF-alpha increased reactive oxygen species generation, including hydrogen peroxide and superoxide radicals, as detected by dihydrorhodamine-123 and dihydroethidium. The TNF-alpha response was inhibited by superoxide dismutase and catalase suggesting apoptosis is oxidative stress related. Inhibition of endogenous HO-2 in newborn pig CMVEC increased oxidative stress and exaggerated apoptosis caused by serum deprivation and TNF-alpha. In HO-1-overexpressing CMVEC (HO-1 selective induction by cobalt portophyrin), TNF-alpha did not cause apoptosis. A CO-releasing compound, CORM-A1, and bilirubin blocked TNF-alpha-induced reactive oxygen species accumulation and apoptosis consistent with the antioxidant and antiapoptotic roles of the end products of HO activity. We conclude that HO-2 is critical for protection of cerebrovascular endothelium against apoptotic changes induced by oxidative stress and cytokine-mediated inflammation.


Asunto(s)
Apoptosis/efectos de los fármacos , Encéfalo/citología , Encéfalo/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Hemo Oxigenasa (Desciclizante)/metabolismo , Estrés Oxidativo/efectos de los fármacos , Factor de Necrosis Tumoral alfa/toxicidad , Animales , Animales Recién Nacidos , Bilirrubina/farmacología , Monóxido de Carbono/farmacología , Células Cultivadas , Células Endoteliales/enzimología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Expresión Génica/efectos de los fármacos , Hemo Oxigenasa (Desciclizante)/antagonistas & inhibidores , Hemo-Oxigenasa 1/metabolismo , Isoenzimas/metabolismo , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Transporte de Proteínas/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno/metabolismo , Porcinos , Regulación hacia Arriba/efectos de los fármacos
18.
J Biol Chem ; 278(49): 49239-45, 2003 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-14500730

RESUMEN

A recent study (Nusrat, A., Chen, J. A., Foley, C. S., Liang, T. W., Tom, J., Cromwell, M., Quan, C., and Mrsny, R. J. (2000) J. Biol. Chem. 275, 29816-29822) suggested that phosphatidylinositol 3-kinase (PI 3-kinase) may interact with occludin; however, there exists no evidence of direct interaction of PI 3-kinase with the tight junctions. Activation of PI 3-kinase by oxidative stress and its role in disruption of tight junctions was examined in Caco-2 cell monolayer. The oxidative stress-induced decrease in electrical resistance, increase in inulin permeability, and redistribution of occludin and ZO-1 were reduced by a PI 3-kinase inhibitor, LY294002. Oxidative stress-induced tyrosine phosphorylation and dissociation from the actin cytoskeleton of occludin and ZO-1 were reduced by LY294002. The regulatory subunit of PI 3-kinase, p85, and the PI 3-kinase activity were co-immunoprecipitated with occludin, which were rapidly increased by oxidative stress. Oxidative stress resulted in increased translocation of p85 from the intracellular compartment into the intercellular junctions. Pair-wise glutathione S-transferase pull-down assay showed that glutathione S-transferase-occludin (C-terminal tail) binds to recombinant p85. This study shows that oxidative stress increases the association of PI 3-kinase with the occludin, and that PI 3-kinase activity is involved in oxidative stress-induced disruption of tight junction.


Asunto(s)
Estrés Oxidativo , Fosfatidilinositol 3-Quinasas/fisiología , Uniones Estrechas/fisiología , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Microscopía Fluorescente , Morfolinas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación
19.
Biochem J ; 368(Pt 2): 471-81, 2002 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-12169098

RESUMEN

The oxidative-stress-induced alteration in paracellular junctional complexes was analysed in Caco-2 cell monolayer. Oxidative stress induced a rapid increase in tyrosine phosphorylation of occludin, zonula occludens (ZO)-1, E-cadherin and beta-catenin. An oxidative-stress-induced decrease in transepithelial electrical resistance was associated with a redistribution of occludin-ZO-1 and E-cadherin-beta-catenin complexes from the intercellular junctions. Genistein, a tyrosine kinase inhibitor, prevented the oxidative-stress-induced decrease in resistance and redistribution of protein complexes. Occludin, ZO-1, E-cadherin and beta-catenin in the Triton-insoluble cytoskeletal fraction were reduced by oxidative stress, which was prevented by genistein. Oxidative stress also reduced the co-immunoprecipitation of ZO-1 with occludin, which was prevented by genistein. Co-immunoprecipitation of beta-catenin with E-cadherin was unaffected by oxidative stress or genistein. ZO-1, E-cadherin and beta-catenin in the plasma membrane or membrane-cytoskeleton were either slightly reduced or unaffected by oxidative stress or genistein. These results show that oxidative stress induces tyrosine phosphorylation and cellular redistribution of occludin-ZO-1 and E-cadherin-beta-catenin complexes by a tyrosine-kinase-dependent mechanism.


Asunto(s)
Cadherinas/metabolismo , Citoesqueleto/metabolismo , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Tirosina/metabolismo , Células CACO-2/efectos de los fármacos , Células CACO-2/metabolismo , Membrana Celular/metabolismo , Proteínas del Citoesqueleto/efectos de los fármacos , Proteínas del Citoesqueleto/metabolismo , Inhibidores Enzimáticos/farmacología , Genisteína/farmacología , Humanos , Uniones Intercelulares/metabolismo , Ocludina , Estrés Oxidativo , Fosforilación/efectos de los fármacos , Proteínas Tirosina Quinasas/efectos de los fármacos , Proteínas Tirosina Quinasas/metabolismo , Transactivadores/efectos de los fármacos , Transactivadores/metabolismo , Xantina/farmacología , Xantina Oxidasa/farmacología , Proteína de la Zonula Occludens-1 , beta Catenina
20.
J Biol Chem ; 278(14): 11916-24, 2003 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-12547828

RESUMEN

The activity of Src kinases appears to play a role in both assembly and disassembly of tight junction. However, the role of a specific isoform of Src kinase in regulation of tight junction is not known. In the present study the role of c-Src in regulation of epithelial tight junction was investigated in Caco-2 cell monolayers. Oxidative stress (xanthine oxidase + xanthine) induced an activation and membrane translocation of c-Src. The oxidative stress-induced decrease in transepithelial electrical resistance, increase in inulin permeability, and redistribution of occludin and ZO-1 from the intercellular junctions were prevented by PP2. The rates of oxidative stress-induced activation of c-Src, tyrosine phosphorylation of ZO-1 and beta-catenin, decrease in resistance, increase in permeability to inulin, and redistribution of occludin and ZO-1 were significantly greater in cells transfected with wild type c-Src, whereas it was low in cells transfected with kinase-inactive c-SrcK297R mutant, when compared with those in empty vector-transfected cells. The rates of recovery of resistance, increase in barrier to inulin, and reorganization of occludin and ZO-1 into the intercellular junctions during the calcium-induced reassembly of tight junction were much greater in Caco-2 cells transfected with c-SrcK297R as compared with those in cells transfected with empty vector or wild type c-Src. These results show that the dominant-negative expression of kinase-inactive c-Src delays the oxidative stress-induced disruption of tight junction and accelerates calcium-induced assembly of tight junction in Caco-2 cells and demonstrate that oxidative stress-induced disruption of tight junction is mediated by the activation of c-Src.


Asunto(s)
Calcio/metabolismo , Estrés Oxidativo/fisiología , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Uniones Estrechas/enzimología , Animales , Proteína Tirosina Quinasa CSK , Células CACO-2 , Permeabilidad de la Membrana Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , Regulación Enzimológica de la Expresión Génica , Humanos , Inulina/farmacocinética , Proteínas de la Membrana/metabolismo , Ocludina , Fosfoproteínas/metabolismo , Fosforilación , Transactivadores/metabolismo , Tirosina/metabolismo , Proteína de la Zonula Occludens-1 , beta Catenina , Familia-src Quinasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA