Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 181
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 86(1): E7-E12, 2000 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-10625313

RESUMEN

Long-term nitroglycerin (NTG) treatment has been shown to be associated with cross-tolerance to endothelium-dependent vasodilators. It may involve increased production of reactive oxygen species (such as superoxide, O(2)(.-)) that rapidly inactivate the nitric oxide (NO) released from the endothelial cells. It remains to be elucidated, however, whether long-term treatment with NTG alters the activity and expression of the endothelial NO synthase (NOS III) and whether this enzyme can contribute to O(2)(.-) formation. We studied the influence of long-term NTG treatment on the expression of NOS III as assessed by RNase protection assay and Western blot. Tolerance was measured ex vivo in organ chamber experiments with rat aortic rings. O(2)(.-) and NO formation were quantified using lucigenin- and Cypridina luciferin analog-enhanced chemiluminescence as well as electron spin resonance (ESR) spectroscopy. Treatment of Wistar rats with NTG (Alzet osmotic minipumps, NTG concentration 10 microg x kg(-1) x min(-1)) for 3 days caused marked tolerance, cross-tolerance to the endothelium-dependent vasodilator acetylcholine, and a significant increase in O(2)(.-)-induced chemiluminescence. Tolerance was associated with a significant increase in NOS III mRNA to 236+/-28% and NOS III protein to 239+/-17%. In control vessels, the NOS inhibitor N(G)-nitro-L-arginine (L-NNA) increased the O(2)(.-)-mediated chemiluminescence, indicating that basal production of endothelium-derived NO depresses the baseline chemiluminescence signal. In the setting of tolerance, however, L-NNA decreased steady-state O(2)(.-) levels, indicating the involvement of NOS III in O(2)(.-) formation. Likewise, A23187-induced, NOS III-mediated O(2)(.-) production was more pronounced in tolerant than in control vessels. Vascular NO bioavailability as assessed with ESR spectroscopy using iron-thiocarbamate as a trap for NO was significantly reduced in tolerant vessels. Pretreatment of tolerant tissue in vitro with the protein kinase C (PKC) inhibitors reduced basal and stimulated NOS III-mediated O(2)(.-) production and partially reversed vascular tolerance. These findings suggest that NTG treatment increases the expression of a dysfunctional NOS III gene, leading to increased formation of O(2)(.-) and decreased vascular NO bioavailability. Normalization of NOS III-mediated O(2)(. -) production and improvement of tolerance with PKC inhibition suggests an important role for PKC isoforms in mediating vascular dysfunction caused by long-term NTG treatment.


Asunto(s)
Endotelio Vascular/metabolismo , Óxido Nítrico Sintasa/genética , Óxido Nítrico/metabolismo , Nitroglicerina/farmacología , Superóxidos/metabolismo , Acetilcolina/farmacología , Alcaloides , Animales , Arginina/farmacología , Benzofenantridinas , Disponibilidad Biológica , Calcimicina/farmacología , Carbazoles/farmacología , Clonación Molecular , Endotelio Vascular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Indoles/farmacología , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa de Tipo III , Fenantridinas/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/fisiología , ARN Mensajero/biosíntesis , Ratas , Ratas Wistar , Tiempo , Vasodilatación/efectos de los fármacos
2.
Circ Res ; 88(2): E14-22, 2001 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-11157681

RESUMEN

Incubation of endothelial cells in vitro with high concentrations of glucose activates protein kinase C (PKC) and increases nitric oxide synthase (NOS III) gene expression as well as superoxide production. The underlying mechanisms remain unknown. To address this issue in an in vivo model, diabetes was induced with streptozotocin in rats. Streptozotocin treatment led to endothelial dysfunction and increased vascular superoxide production, as assessed by lucigenin- and coelenterazine-derived chemiluminescence. The bioavailability of vascular nitric oxide (as measured by electron spin resonance) was reduced in diabetic aortas, although expression of endothelial NOS III (mRNA and protein) was markedly increased. NOS inhibition with N:(G)-nitro-L-arginine increased superoxide levels in control vessels but reduced them in diabetic vessels, identifying NOS as a superoxide source. Similarly, we found an activation of the NADPH oxidase and a 7-fold increase in gp91(phox) mRNA in diabetic vessels. In vitro PKC inhibition with chelerythrine reduced vascular superoxide in diabetic vessels, whereas it had no effect on superoxide levels in normal vessels. In vivo PKC inhibition with N:-benzoyl-staurosporine did not affect glucose levels in diabetic rats but prevented NOS III gene upregulation and NOS-mediated superoxide production, thereby restoring vascular nitric oxide bioavailability and endothelial function. The reduction of superoxide in vitro by chelerythrine and the normalization of NOS III gene expression and reduction of superoxide in vivo by N:-benzoyl-staurosporine point to a decisive role of PKC in mediating these phenomena and suggest a therapeutic potential of PKC inhibitors in the prevention or treatment of vascular complications of diabetes mellitus. The full text of this article is available at http://www.circresaha.org.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Endotelio Vascular/metabolismo , Superóxidos/metabolismo , Enfermedades Vasculares/metabolismo , Animales , Aorta , Glucemia/efectos de los fármacos , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/fisiopatología , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiopatología , Inhibidores Enzimáticos/farmacología , Técnicas In Vitro , Mediciones Luminiscentes , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , NADPH Oxidasa 2 , NADPH Oxidasas/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo III , Estrés Oxidativo/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Estreptozocina , Regulación hacia Arriba/efectos de los fármacos , Enfermedades Vasculares/etiología
3.
Biochim Biophys Acta ; 793(3): 338-45, 1984 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-6370311

RESUMEN

Five different vascular beds of the cat were tested for their ability to metabolize tritium-labeled or unlabeled prostacyclin by 15-hydroxyprostaglandin dehydrogenase in vivo. Thin-layer radiochromatography with two different solvent systems was used to characterize the radioactive metabolites formed. Following the infusion of unlabeled prostacyclin, plasma levels of 6-ketoprostaglandin F1 alpha and 6,15-diketo-13,14-dihydroprostaglandin F1 alpha immunoreactivities were monitored using previously described radioimmunoassays. No important metabolism was found in the head and the hindlimb. Plasma extracts from the vascular bed of the lung (and the heart) contained only small amounts of 6,15-diketo-13,14- dihydroprostaglandin F1 alpha-like material. prostacyclin was extensively metabolized in the liver (probably by beta- and omega-oxidation), but no material similar to 6,15-diketo-13,14-dihydroprostaglandin F1 alpha was found. In renal venous blood the predominating material showed properties similar to those of 6,15-diketo-13,14-dihydroprostaglandin F1 alpha with both analytical methods. In other experiments aortic blood was collected during and after infusions of tritium-labeled prostacyclin into the postcava . Two consistent peaks were demonstrated in plasma extracts of this blood, one co-chromatographing with 6-ketoprostaglandin F1 alpha and the other one with 6,15-diketo-13,14-dihydroprostaglandin F1 alpha. In nephrectomized animals the formation of the 6,15-diketo-13,14-dihydroprostaglandin F1 alpha-like material was not significantly impaired. Similarly, no significant differences in the formation and elimination of 6,15-diketo-13,14-dihydroprostaglandin F1 alpha immunoreactivity and 6-ketoprostaglandin F1 alpha immunoreactivity were found after nephrectomy. It is concluded that, although the kidneys have a considerable capacity to metabolize prostacyclin by 15-hydroxyprostaglandin dehydrogenase, they seem to be of little importance for the overall conversion of prostacyclin via this pathway in the total circulation.


Asunto(s)
Epoprostenol/metabolismo , Riñón/metabolismo , Animales , Gatos , Femenino , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Riñón/enzimología , Masculino , Prostaglandinas F/biosíntesis , Radioinmunoensayo
4.
Biochim Biophys Acta ; 712(3): 684-91, 1982 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-6751407

RESUMEN

Using previously described radioimmunoassays for 6-ketoprostaglandin F1 alpha and 6,15-diketo-13,14-dihydroprostaglandin F1 alpha, plasma levels of these two products of degradation of prostacyclin (prostaglandin I2) were monitored in cats and rabbits. It was shown that both exogenous prostaglandin I2 and 6-ketoprostaglandin F1 alpha could be rapidly converted to 6,15-diketo-13,14-dihydroprostaglandin F1 alpha immunoreactivity, 6-ketoprostaglandin F1 alpha to an even somewhat larger extent. This difference in conversion could be explained by competing mechanisms which could delay or hinder the access of prostaglandin I2 to the sites of metabolism by 15-hydroxyprostaglandin dehydrogenase. The 6-ketoprostaglandin F1 alpha and 6,15-diketo-13,14-dihydroprostaglandin F1 alpha immunoreactivities were further characterized in two different thin-layer chromatographic systems and were shown to co-chromatograph exclusively with their respective standards. Similar results were obtained when tritium-labeled prostaglandin I2 or 6-ketoprostaglandin F1 alpha were infused into cats. Extracts of plasma samples taken at different time intervals after the infusion were submitted to thin-layer chromatography. The radioscans of the chromatograms showed two consistent peaks, one co-chromatographing with authentic 6-ketoprostaglandin F1 alpha and the other one with 6,15-diketo-13,14-dihydroprostaglandin F1 alpha. After infusion of 6-ketoprostaglandin F1 alpha the conversion to the 6,15-diketo-13,14-dihydro-prostaglandin F1 alpha-like product occurred somewhat faster than after infusion of prostaglandin I2. We conclude that, under in vivo conditions in the two species investigated, 6-ketoprostaglandin F1 alpha can be rapidly and effectively metabolized by 15-hydroxyprostaglandin dehydrogenase.


Asunto(s)
6-Cetoprostaglandina F1 alfa/análogos & derivados , 6-Cetoprostaglandina F1 alfa/metabolismo , Epoprostenol/metabolismo , Prostaglandinas/metabolismo , 6-Cetoprostaglandina F1 alfa/sangre , Animales , Gatos , Cromatografía en Capa Delgada , Cinética , Conejos , Radioinmunoensayo
5.
J Am Coll Cardiol ; 32(4): 964-9, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9768718

RESUMEN

OBJECTIVES: The expression and localization of inducible nitric oxide (NO) synthase (NOS II) was evaluated as a source of NO which has been shown to affect muscle contraction. BACKGROUND: Advanced stages of chronic heart failure are associated with systemic activation of cytokines which have been shown to stimulate the expression of NOS II in various cell types, including myocytes. We hypothesized that systemic cytokine activation could lead to expression of NOS II in skeletal muscle of patients with chronic heart failure. METHODS: Skeletal muscle specimens were obtained by percutaneous needle biopsy in six normal volunteers and eight patients with heart failure (New York Heart Association class III). Electron microscopy immunocytochemistry (immunogold labeling) with specific anti-NOS antibodies was utilized to elucidate the intracellular localization of NOS II and neuronal NO synthase (NOS I) in myocytes of skeletal muscle. Reverse transcriptase, competitive polymerase chain reaction (PCR) was applied to quantify NOS II mRNA in skeletal muscle. RESULTS: Inducible nitric oxide synthase was readily expressed in the cytosol of skeletal muscle myocytes; NOS I expression was sparse. Polymerase chain reaction results indicated that NOS II gene expression is increased in patients with chronic heart failure. CONCLUSIONS: Inducible NO synthase is expressed in human skeletal muscle and its gene expression is increased in patients with severe heart failure. Given the experimental evidence that NO can attenuate contractile performance of skeletal muscle and can mediate muscle wasting, an increased local production of NO in skeletal muscle by NOS II may have important implications for patients with severe heart failure.


Asunto(s)
Insuficiencia Cardíaca/enzimología , Músculo Esquelético/enzimología , Óxido Nítrico Sintasa/análisis , Adulto , Enfermedad Crónica , Expresión Génica , Humanos , Microscopía Inmunoelectrónica , Persona de Mediana Edad , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Reacción en Cadena de la Polimerasa , ARN Mensajero/análisis , ADN Polimerasa Dirigida por ARN
6.
J Am Coll Cardiol ; 32(5): 1179-86, 1998 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9809923

RESUMEN

OBJECTIVES: The purpose of the present study was to examine the expression of the endothelial-type nitric oxide synthase (NOS III) and the inducible-type NOS (NOS II) in human myocardium and their regulation in heart failure from patients with different etiologies. BACKGROUND: In heart failure, plasma levels of nitrates were found to be elevated. However, data on myocardial NOS expression in heart failure are conflicting. METHODS: Using RNase protection analysis and Western blotting, the expression of NOS III and NOS II was investigated in ventricular myocardium from nonfailing (NF) hearts (n=5) and from failing hearts of patients with idiopathic dilated cardiomyopathy (dCMP, n=14), ischemic cardiomyopathy (iCMP, n=9) or postmyocarditis cardiomyopathy (mCMP, n=7). Furthermore, immunohistochemical studies were performed to localize NOS III and NOS II within the ventricular myocardium. RESULTS: In failing human hearts, NOS III mRNA levels were increased to 180% in dCMP, 200% in iCMP and to 210% in mCMP as compared to NF hearts. Similarly, in Western blots (using constitutively expressed beta-tubulin as a reference) NOS III protein expression was increased about twofold in failing compared to NF hearts. Immunohistochemical studies with a selective antibody to NOS III showed no obvious differences in the staining of the endothelium of cardiac blood vessels from NF and failing human hearts. However, NOS III-immunoreactivity in cardiomyocytes was significantly more intense in failing compared to NF hearts. Low expression of NOS II mRNA was detected in only 2 of 30 failing human hearts and was not found in NF hearts. Inducible-type NOS protein was undetectable in either group. CONCLUSIONS: We conclude that the increased NOS III expression in the ventricular myocardium of failing human hearts may contribute to the contractile dysfunction observed in heart failure and/or may play a role in morphologic alterations such as hypertrophy and apoptosis of cardiomyocytes.


Asunto(s)
Insuficiencia Cardíaca/enzimología , Miocardio/enzimología , Óxido Nítrico Sintasa/metabolismo , Adulto , Western Blotting , Cardiomiopatía Dilatada/complicaciones , Cardiomiopatía Dilatada/enzimología , Expresión Génica , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/cirugía , Trasplante de Corazón , Ventrículos Cardíacos/enzimología , Humanos , Inmunohistoquímica , Técnicas In Vitro , Persona de Mediana Edad , Isquemia Miocárdica/complicaciones , Isquemia Miocárdica/enzimología , Miocarditis/complicaciones , Miocarditis/enzimología , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , ARN Mensajero/metabolismo , Donantes de Tejidos
7.
J Am Coll Cardiol ; 36(2): 628-34, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10933381

RESUMEN

OBJECTIVES: In the present study, we tested whether a nitrate-free interval is able to prevent increases in vascular superoxide (O2*-) and the development of hypersensitivity to vasoconstrictors and whether this may result in restoration of vascular nitroglycerin (NTG) sensitivity. BACKGROUND: Intermittent NTG-patch treatment (12 h patch on/patch-off) has been shown to increase ischemic periods in patients with stable coronary arteries, suggesting a rebound-like situation during the patch-off period. Recently, we demonstrated that long-term treatment with NTG induces tolerance, which was in part related to increases in vascular O2*- and increased vasoconstrictor sensitivity. METHODS: New Zealand white rabbits received a continuous application of NTG patches (0.4 mg/h) or an intermittent application of NTG patches (12 h patch on, 12 h patch off) for three days. Isometric tension studies were performed with aortic rings, and vascular O2*- was estimated using lucigenin-derived chemiluminescence (5 micromol/liter). Expression of the copper/zinc (Cu/Zn) superoxide dismutase (SOD) was assessed by Western blotting, and SOD activity was measured by autooxidation of 6-hydroxydopamine. RESULTS: Continuous treatment with NTG caused tolerance to NTG, cross-tolerance to the endothelium-dependent vasodilator acetylcholine, increased vascular O2*-, reduced Cu/Zn SOD expression and increased sensitivity to vasoconstrictors such as phenylephrine, serotonin and angiotensin II. On/off treatment with NTG improved tolerance, corrected endothelial dysfunction and decreased vascular O2*-. In addition the reduction in SOD expression was less pronounced, whereas increases in the sensitivity to vasoconstrictors such as phenylephrine and serotonin remained nearly unchanged. CONCLUSIONS: Enhanced vasoconstrictor sensitivity may explain, at least in part, the rebound phenomena observed in patients during a 12-h NTG patch-off period.


Asunto(s)
Endotelio Vascular/metabolismo , Nitroglicerina/farmacología , Superóxido Dismutasa/metabolismo , Vasoconstricción/fisiología , Acetilcolina/farmacología , Animales , Western Blotting , Tolerancia a Medicamentos , Endotelio Vascular/efectos de los fármacos , Femenino , Técnicas In Vitro , Masculino , Estrés Oxidativo/fisiología , Conejos , Factores de Tiempo , Vasoconstricción/efectos de los fármacos , Vasodilatadores/farmacología
8.
Trends Cardiovasc Med ; 3(3): 104-10, 1993.
Artículo en Inglés | MEDLINE | ID: mdl-21244948

RESUMEN

The endothelium-derived relaxing factor that mediates the endothelium-dependent vasodilatation first observed in 1980 has been identified as nitric oxide (NO). In addition to the endothelium, NO is formed in other cells such as neuronal cells of the brain (where it mediates synoptic plasticity), peripheral nonadrenergic noncholinergic (NANC) nerves (where it acts as an atypical neurotransmitter relaxing vascular and nonvascular smooth muscle), and various specialized epithelial cells. Other cell types such as macrophages and smooth muscle cells can be induced with bacterial endotoxin and/or cytokines to synthesize large amounts of the radical. At low concentrations, NO is an inter- and intracellular messenger molecule whose target enzyme is the soluble isoform of guanylyl cyclase. At high concentrations, the NO radical has cytostatic effects on parasitic microorganisms and tumor cells. In the vascular system, endothelium-derived NO is a physiologically significant vasodilator and inhibitor of platelet aggregation and adhesion. NANC nerve-derived NO may also contribute to vasodilatation. In addition, NO can prevent leukocyte adhesion to the endothelium by interfering with the adhesion molecule CD11/CD18, and NO has been shown to inhibit the proliferation of vascular smooth muscle cells. In sepsis and during cytokine therapy, a different NOS is induced in the vascular wall (presumably in smooth muscle cells) where it synthesizes large amounts of NO that contribute to the massive vasodilatation and shock.

9.
Cell Calcium ; 13(6-7): 427-34, 1992.
Artículo en Inglés | MEDLINE | ID: mdl-1380405

RESUMEN

NO synthase (NOS) catalyzes the oxidation of L-arginine to L-citrulline and nitric oxide (NO) or a NO-releasing compound. At least three isoforms of NOS exist (types I-III). The activities of the type I isoform purified from brain and the type III isoform purified from endothelial cells are regulated by the intracellular free calcium concentration ([Ca2+]i) and the Ca(2+)-binding protein calmodulin. At resting [Ca2+]i, both isozymes are inactive; they become fully active at [Ca2+]i greater than or equal to 500 nM Ca2+. Longer lasting increases in [Ca2+]i may downregulate NO formation, for in vitro phosphorylation by Ca2+/calmodulin protein kinase II decreases the Vmax of NOS. Besides the conversion of L-arginine, type I NOS, Ca2+/calmodulin dependently, generates H2O2 and reduces cytochrome c/P450. Other redox activities, i.e. the reduction of nitroblue tetrazolium to diformazan (NADPH-diaphorase) or of quinoid-dihydrobiopterin to tetrahydrobiopterin, by NOS appear to be Ca2+/calmodulin-independent.


Asunto(s)
Aminoácido Oxidorreductasas/metabolismo , Calcio/fisiología , Calmodulina/fisiología , Óxido Nítrico Sintasa
10.
Eur J Cell Biol ; 78(2): 134-42, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10099936

RESUMEN

We studied the expression of the NO synthase isoforms in an in vitro model of neural development using RT-PCR, Western blot and immunohistochemistry. Murine PCC7-Mz1 cells (Jostock et al., Eur. J. Cell Biol. 76, 63-76, 1998) differentiate in the presence of all-trans retinoic acid and dibutyryl cAMP along the neural pathway into neuron-like, fibroblast-like and astroglia-like cells. Undifferentiated cells showed immunofluorescent staining for neuronal-type NOS I and endothelial-type NOS III. This expression pattern was retained in those cells differentiating into neurofilament- and tau protein-positive neuronal cells. Thymocyte alloantigen (Thy1.2/CD 90.2)-positive fibroblasts, appearing around day 3, and glial fibrillary acidic protein (GFAP)-positive astroglial cells, appearing after day 6 of differentiation, stained negative for any NOS isoform. Starting at day 6 of differentiation, expression of inducible-type NOS II could be stimulated with cytokines in a subset of cells, which may represent activated astrocytes. NOS II was always undetectable in non-induced cultures. These data indicate that the ability of stem cells to express NOS I and NOS III is only retained when the cells differentiate along the neuronal lineage, while a small subpopulation of cells acquires the ability to express NOS II in response to cytokines.


Asunto(s)
Astrocitos/enzimología , Carcinoma Embrionario/enzimología , Fibroblastos/enzimología , Neuronas/enzimología , Óxido Nítrico Sintasa/biosíntesis , Animales , Western Blotting , Bucladesina/farmacología , Carcinoma Embrionario/patología , Diferenciación Celular/efectos de los fármacos , Citocinas/farmacología , Inducción Enzimática/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Ratones , Óxido Nítrico Sintasa de Tipo I , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tretinoina/farmacología , Células Tumorales Cultivadas
11.
J Invest Dermatol ; 117(5): 1200-5, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11710933

RESUMEN

Skin exposure to ultraviolet radiation from sunlight causes erythema and edema formation as well as inflammatory responses. As some of these ultraviolet-induced effects are potentially mediated by nitric oxide synthases, we examined the role of cytokines and ultraviolet A1 radiation (340-400 nm) on the expression of the nitric oxide synthase-2 in endothelia of normal human skin biopsies during short-term organ culture as well as expression and activity of the nitric oxide synthase-2 in in vitro cell cultures of human dermal endothelial cells. Both, cytokine challenge (interleukin-1beta + tumor necrosis factor-alpha + interferon-gamma) but also ultraviolet A1 exposure (50 J per cm2) in the absence of cytokines led to the expression of nitric oxide synthase-2 in human skin organ cultures as shown by immunohistochemistry. Moreover, exposing human dermal endothelial cell cultures to proinflammatory cytokines but also to ultraviolet A1 radiation (6-24 J per cm2) in the absence of cytokines resulted in significant nitric oxide synthase-2 mRNA and protein expression as well as enzyme activity. Ultraviolet A1 irradiation of cytokine activated cells led to further increases in nitric oxide synthase-2 mRNA, protein expression, and enzyme activity. Moreover, a reporter gene assay using a human nitric oxide synthase-2 promoter construct provide evidence that ultraviolet A1, in the absence of cytokines, induces nitric oxide synthase-2 expression and activity, as previously shown for cytokines. Thus, the results presented here demonstrate for the first time that in dermal endothelia of human skin ultraviolet A1 radiation alone represents a proinflammatory stimulus sufficient to initiate nitric oxide synthase-2 expression as well as activity comparable with the respective response seen in the presence of proinflammatory cytokines.


Asunto(s)
Óxido Nítrico Sintasa/metabolismo , Piel/enzimología , Piel/efectos de la radiación , Rayos Ultravioleta , Línea Celular , Citocinas/farmacología , Endotelio/citología , Endotelio/efectos de los fármacos , Endotelio/enzimología , Endotelio/efectos de la radiación , Inducción Enzimática , Humanos , Mediadores de Inflamación/farmacología , Interferón gamma/farmacología , Interleucina-1/farmacología , Óxido Nítrico Sintasa de Tipo II , Regiones Promotoras Genéticas/fisiología , Regiones Promotoras Genéticas/efectos de la radiación , Piel/citología , Piel/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
12.
Hypertension ; 23(6 Pt 2): 1121-31, 1994 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7515853

RESUMEN

Three isozymes of nitric oxide (NO) synthase (EC 1.14.13.39) have been identified and the cDNAs for these enzymes isolated. In humans, isozymes I (in neuronal and epithelial cells), II (in cytokine-induced cells), and III (in endothelial cells) are encoded for by three different genes located on chromosomes 12, 17, and 7, respectively. The deduced amino acid sequences of the human isozymes show less than 59% identity. Across species, amino acid sequences for each isoform are well conserved (> 90% for isoforms I and III, > 80% for isoform II). All isoforms use L-arginine and molecular oxygen as substrates and require the cofactors NADPH, 6(R)-5,6,7,8-tetrahydrobiopterin, flavin adenine dinucleotide, and flavin mononucleotide. They all bind calmodulin and contain heme. Isoform I is constitutively present in central and peripheral neuronal cells and certain epithelial cells. Its activity is regulated by Ca2+ and calmodulin. Its functions include long-term regulation of synaptic transmission in the central nervous system, central regulation of blood pressure, smooth muscle relaxation, and vasodilation via peripheral nitrergic nerves. It has also been implicated in neuronal death in cerebrovascular stroke. Expression of isoform II of NO synthase can be induced with lipopolysaccharide and cytokines in a multitude of different cells. Based on sequencing data there is no evidence for more than one inducible isozyme at this time. NO synthase II is not regulated by Ca2+; it produces large amounts of NO that has cytostatic effects on parasitic target cells by inhibiting iron-containing enzymes and causing DNA fragmentation. Induced NO synthase II is involved in the pathophysiology of autoimmune diseases and septic shock. Isoform III of NO synthase has been found mostly in endothelial cells. It is constitutively expressed, but expression can be enhanced, eg, by shear stress. Its activity is regulated by Ca2+ and calmodulin. NO from endothelial cells keeps blood vessels dilated, prevents the adhesion of platelets and white cells, and probably inhibits vascular smooth muscle proliferation.


Asunto(s)
Aminoácido Oxidorreductasas , Aminoácido Oxidorreductasas/metabolismo , Aminoácido Oxidorreductasas/fisiología , Clonación Molecular , Isoenzimas , Isoenzimas/metabolismo , Isoenzimas/fisiología , Aminoácido Oxidorreductasas/química , Animales , Arginina/fisiología , Humanos , Isoenzimas/química , Óxido Nítrico Sintasa , Distribución Tisular
13.
Hypertension ; 31(2): 582-8, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9461225

RESUMEN

Estrogens have been found to reduce the incidence of cardiovascular disease that has been ascribed in part to an increased expression and/or activity of the vasoprotective endothelial NO synthase (NOS III). Some reports have shown that the level of expression of this constitutive enzyme can be upregulated by estrogens. The current study investigates the molecular mechanism of the NOS III upregulation in human endothelial EA.hy 926 cells. Incubation of EA.hy 926 cells with 17beta-estradiol or the more stable 17alpha-ethinyl estradiol enhanced NOS III mRNA and protein expression up to 1.8-fold, without changing the stability of the NOS III mRNA. There was no enhancement of NOS III mRNA after incubation of EA.hy 926 cells with testosterone, progesterone, or dihydrocortisol or when 17alpha-ethinyl estradiol was added together with the estrogen antagonist RU58668, indicating a specific estrogenic response. Nuclear run-on assays indicated that the increase in NOS III mRNA is the result of an estrogen-induced enhancement of NOS III gene transcription. In transient transfection experiments using a 1.6 kb human NOS III promoter fragment (which contains no bona fide estrogen-responsive element, ERE), basal promoter activity was enhanced 1.7-fold by 17alpha-ethinyl estradiol. In electrophoretic mobility shift assays, nuclear extracts from estrogen-incubated EA.hy 926 cells showed no enhanced binding activity either for the ERE-like motif in the human NOS III promoter or for transcription factor GATA. However, binding of transcription factor Sp1 (which is essential for the activity of the human NOS III promoter) was significantly enhanced by estrogens. These data suggest that the estrogen stimulation of the NOS III promoter could be mediated in part by an increased activity of transcription factor Sp1.


Asunto(s)
Endotelio Vascular/enzimología , Congéneres del Estradiol/farmacología , Estradiol/farmacología , Etinilestradiol/farmacología , Óxido Nítrico Sintasa/genética , Factores de Transcripción/fisiología , Transcripción Genética/efectos de los fármacos , Secuencia de Bases , Línea Celular , Núcleo Celular/química , Secuencia de Consenso , ADN/genética , ADN/metabolismo , Estabilidad de Medicamentos , Endotelio Vascular/citología , Humanos , Isoenzimas/genética , Óxido Nítrico Sintasa/metabolismo , Regiones Promotoras Genéticas/genética , ARN Mensajero/química , ARN Mensajero/metabolismo , Extractos de Tejidos/metabolismo , Transfección
14.
FEBS Lett ; 410(2-3): 319-23, 1997 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-9237654

RESUMEN

We have investigated the expression of neuronal-type NO synthase I (NOS I) and inducible-type NOS II in guinea pig skeletal muscle (diaphragm). Expression of NOS I mRNA and protein was highest in muscle of specific pathogen-free animals, lower in normally bred animals, and lowest in lipopolysaccharide (LPS)-treated animals. NOS II mRNA and protein levels were highest in muscle of LPS-treated animals. Elevated NOS activity in muscle from LPS-treated animals was less susceptible to the NOS I-selective inhibitor N(G)-nitro-L-arginine. Expressional downregulation of NOS I in sepsis may have implications for contractile function of skeletal muscle.


Asunto(s)
Regulación hacia Abajo , Lipopolisacáridos/farmacología , Músculo Esquelético/enzimología , Neuronas/enzimología , Óxido Nítrico Sintasa/genética , Animales , Cobayas , Masculino , Óxido Nítrico Sintasa/metabolismo
15.
FEBS Lett ; 309(3): 402-4, 1992 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-1381323

RESUMEN

The enzyme responsible for the synthesis of endothelium-derived relaxing factor and/or nitric oxide in the endothelium has been described as a particulate enzyme, whereas other isoforms of nitric oxide synthase are soluble enzymes. Here we are reporting that endothelial cells metabolically incorporate myristate (C14), but not palmitate (C16), into nitric oxide synthase. We are postulating that the endothelial-derived nitric oxide synthase is a particulate enzyme because of the fatty acid acylation of the protein which 'anchors' the enzyme into the membrane either directly or via another membrane-bound protein.


Asunto(s)
Aminoácido Oxidorreductasas/metabolismo , Endotelio Vascular/enzimología , Animales , Bovinos , Electroforesis en Gel de Poliacrilamida , Óxido Nítrico Sintasa
16.
FEBS Lett ; 316(2): 175-80, 1993 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-7678401

RESUMEN

Complementary DNA clones corresponding to human brain nitric oxide (NO) synthase have been isolated. The deduced amino acid sequence revealed an overall identity with rat brain NO synthase of about 93% and contained all suggested consensus sites for binding of the co-factors. The cDNA transfected COS-1 cells showed significant NO synthase activity with the typical co-factor requirements. Unexpectedly, messenger RNA levels of this isoform of NO synthase was more abundant in human skeletal muscle than human brain. Moreover, we detected high NO synthase activity and the expressed protein in human skeletal muscle by Western blot analysis, indicating a possible novel function of NO in skeletal muscle.


Asunto(s)
Aminoácido Oxidorreductasas/genética , Cerebelo/enzimología , Músculos/enzimología , Adolescente , Adulto , Aminoácido Oxidorreductasas/biosíntesis , Secuencia de Aminoácidos , Animales , Sitios de Unión , Línea Celular , Clonación Molecular , ADN , Femenino , Humanos , Masculino , Datos de Secuencia Molecular , Óxido Nítrico Sintasa , ARN Mensajero/genética , Ratas , Homología de Secuencia de Aminoácido
17.
Neuroscience ; 59(2): 429-45, 1994 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-7516501

RESUMEN

Nitric oxide synthase immunoreactivity was detected in neurons and fibers of the rat pontine medulla. In the medulla, nitric oxide synthase-positive neurons and processes were observed in the gracile nucleus, spinal trigeminal nucleus, nucleus of the solitary tract, dorsal motor nucleus of the vagus, nucleus ambiguus, medial longitudinal fasciculus, reticular nuclei and lateral to the pyramidal tract. In the pons, intensely labeled neurons were observed in the pedunculopontine tegmental nucleus, paralemniscal nucleus, ventral tegmental nucleus, laterodorsal tegmental nucleus, and lateral and medial parabrachial nuclei. Labeled neurons and fibers were seen in the interpeduncular nuclei, dorsal and median raphe nuclei, central gray and dorsal central gray, and superior and inferior colliculi. Double-labeling techniques showed that a small population (< 5%) of nitric oxide synthase-positive neurons in the medulla also contained immunoreactivity to the aminergic neuron marker tyrosine hydroxylase. The majority of nitric oxide synthase-immunoreactive neurons in the dorsal and median raphe nuclei were 5-hydroxytryptamine-positive, whereas very few 5-hydroxytryptamine-positive cells in the caudal raphe nuclei were nitric oxide synthase-positive. Virtually all nitric oxide synthase-positive neurons in the pedunculopontine and laterodorsal tegmental nuclei were also choline acetyltransferase-positive, whereas nitric oxide synthase immunoreactivity was either low or not detected in choline acetyltransferase-positive neurons in the medulla. The results indicate a rostrocaudal gradient in the intensity of nitric oxide synthase immunoreactivity, i.e. it is highest in neurons of the tegmentum nuclei and neurons in the medulla are less intensely labeled. The majority of cholinergic and serotonergic neurons in the pons are nitric oxide synthase-positive, whereas the immunoreactivity was either too low to be detected or absent in the large majority of serotonergic, aminergic and cholinergic neurons in the medulla.


Asunto(s)
Aminoácido Oxidorreductasas/análisis , Bulbo Raquídeo/enzimología , Fibras Nerviosas/enzimología , Neuronas/enzimología , Puente/enzimología , Animales , Biomarcadores/análisis , Femenino , Fluoresceína-5-Isotiocianato , Inmunohistoquímica , Masculino , Bulbo Raquídeo/citología , Fibras Nerviosas/ultraestructura , Neuronas/citología , Óxido Nítrico Sintasa , Especificidad de Órganos , Puente/citología , Núcleos del Rafe/citología , Núcleos del Rafe/enzimología , Ratas , Ratas Sprague-Dawley , Serotonina/análisis , Tirosina 3-Monooxigenasa/análisis
18.
Neuroscience ; 79(1): 237-45, 1997 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9178879

RESUMEN

Neuronal nitric oxide synthase immunoreactivity was localized to sympathetic preganglionic neurons of the intermediolateral cell column and cyclic GMP immunoreactivity to nerve fibers projecting into the intermediolateral cell column of 20-25-day-old rats. Whole-cell patch-clamp recordings were made from sympathetic preganglionic neurons in spinal cord slices of immature rats and the role of nitric oxide and cyclic GMP on excitatory postsynaptic currents was studied. Superfusing the slices with the nitric oxide precursor L-arginine (300 microM) increased the amplitude of evoked excitatory postsynaptic currents as well as the frequency of spontaneous miniature excitatory postsynaptic currents in some neurons from minutes to over 1 h. The nitric oxide synthase inhibitor N(W)-nitro-L-arginine (100 microM) and the nitric oxide scavenger hemoglobin (100 microM) antagonized the potentiating effect of L-arginine. The nitric oxide donor sodium nitroprusside (100 microM) potentiated the synaptic currents in a manner similar to that of L-arginine and this effect was blocked by hemoglobin. The membrane-permeable cyclic GMP analogue dibutyryl guanosine 3',5'-cyclic monophosphate (350 microM), in the presence of the phosphodiesterase inhibitor 3-isobutyl-1-methylxanthine (750 microM), potentiated the evoked excitatory postsynaptic currents and increased the frequency of miniature excitatory postsynaptic currents; these effects were not prevented by hemoglobin. The results indicate that nitric oxide may facilitate the release of excitatory transmitters, possibly through a presynaptic cyclic GMP-dependent mechanism.


Asunto(s)
Potenciales Evocados/fisiología , Ganglios Simpáticos/fisiología , Neuronas/fisiología , Óxido Nítrico Sintasa/análisis , Nitroarginina/farmacología , Médula Espinal/citología , Transmisión Sináptica/fisiología , 1-Metil-3-Isobutilxantina/farmacología , Animales , Arginina/farmacología , GMP Cíclico/análisis , Estimulación Eléctrica , Potenciales Evocados/efectos de los fármacos , Hemoglobinas/farmacología , Inmunohistoquímica , Fibras Nerviosas/fisiología , Fibras Nerviosas/ultraestructura , Neuronas/citología , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Transmisión Sináptica/efectos de los fármacos
19.
Neuroscience ; 65(1): 231-9, 1995 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-7538645

RESUMEN

Chronic (5-14 days) preganglionic denervation of the rat superior cervical ganglia by sectioning the cervical sympathetic trunk resulted in a time-related partial or complete loss of nitric oxide synthase (isoform I)-immunoreactive fibers and terminals surrounding many sympathetic ganglionic neurons. Unexpectedly, denervation unmasked many varicose nitric oxide synthase-immunoreactive fibers, some of which could be traced the entire length of the superior cervical ganglia. Injection of the retrograde tracer Fluorogold into the superior cervical ganglia labeled a population of nodose ganglion cells and of dorsal root ganglion cells from C8 to T3 segments. When the same sections were processed for nitric oxide synthase-immunoreactivity, 40% of the Fluorogold-containing nodose ganglion cells also expressed nitric oxide synthase-immunoreactivity, whereas colocalization was observed in only a few dorsal root ganglion cells. Similarly, injection of Fluorogold into denervated superior cervical ganglia labeled a population of nodose ganglion cells. Sectioning of all nerve trunks associated with the superior cervical ganglion prior to injection of Fluorogold, except the cervical sympathetic trunk, resulted in no detectable labeling of Fluorogold in the ipsilateral nodose ganglion cells. These results indicate that a population of rat nodose ganglion cells contain nitric oxide synthase and that some of these neurons project their axons through the superior cervical ganglion and terminate in the peripheral target tissues. The possibility that nitric oxide synthase-immunoreactive vagal afferent fibers may participate in nociception is considered.


Asunto(s)
Vías Aferentes/fisiología , Aminoácido Oxidorreductasas/inmunología , Ganglio Cervical Superior/fisiología , Nervio Vago/fisiología , Aminoácido Oxidorreductasas/fisiología , Animales , Femenino , Inmunohistoquímica , Masculino , Óxido Nítrico Sintasa , Ganglio Nudoso/inmunología , Ratas , Ratas Sprague-Dawley
20.
Neuroscience ; 82(1): 255-65, 1998 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9483518

RESUMEN

This study was designed to evaluate whether the enzymes of the nitric oxide/cyclic-GMP pathway, nitric oxide synthase and soluble guanylyl cyclase, are functionally coupled in controlling catecholamine secretion in primary cultures of bovine chromaffin cells. In immunocytochemical studies, 80-85% of the tyrosine hydroxylase-positive chromaffin cells also possessed phenylethanolamine-N-methyltransferase, f1p4cating their capability to synthesize epinephrine. Immunoreactivity for neuronal-type nitric oxide synthase was found in over 90% of all chromaffin cells. Reverse transcription-polymerase chain reaction also demonstrated neuronal-type nitric oxide synthase messenger RNA. Immunoreactivity for soluble guanylyl cyclase was detectable in over 95% of chromaffin cells. Double-labeling immunofluorescence studies co-localized neuronal-type nitric oxide synthase and soluble guanylyl cyclase with tyrosine hydroxylase and phenylethanolamine-N-methyltransferase in the majority of chromaffin cells. Chromaffin cells possessed basal nitric oxide synthase activity which could be stimulated by acetylcholine and inhibited by NG-nitro-L-arginine methyl ester. Activation of soluble guanylyl cyclase by endogenously synthesized nitric oxide or the nitric oxide donor compound sodium nitroprusside was blocked by the inhibitor of soluble guanylyl cyclase 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one. Catecholamine release and the increase in cytosolic Ca2+ concentration evoked by acetylcholine were enhanced by inhibitors of the endogenous nitric oxide/cyclic-GMP pathway such as NG-nitro-L-arginine methyl ester, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one and the protein kinase G inhibitor Rp-8-pCPT-cGMPS. These data indicate that chromaffin cells possess an autocrine nitric oxide/cyclic-GMP pathway tonically controlling the inhibition of catecholamine release.


Asunto(s)
Catecolaminas/metabolismo , Células Cromafines/enzimología , Células Cromafines/metabolismo , Guanilato Ciclasa/metabolismo , Óxido Nítrico Sintasa/metabolismo , Glándulas Suprarrenales/enzimología , Glándulas Suprarrenales/metabolismo , Animales , Calcio/metabolismo , Bovinos , GMP Cíclico/metabolismo , Citosol/metabolismo , Feniletanolamina N-Metiltransferasa/metabolismo , Reacción en Cadena de la Polimerasa , ARN Mensajero/biosíntesis , ARN Mensajero/aislamiento & purificación , Tirosina 3-Monooxigenasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA