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












Base de datos
Intervalo de año de publicación
2.
Thromb Res ; 145: 84-92, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27513882

RESUMEN

INTRODUCTION: Coagulation and complement systems are simultaneously activated at sites of tissue injury, leading to thrombin generation and opsonisation with C3b. Thrombomodulin (TM) is a cell-bound regulator of thrombin activation, but can also enhance the regulatory activity of complement factor H (FH), thus accelerating the degradation of C3b into inactive iC3b. OBJECTIVES: This study sought to determine the biophysical interaction affinities of two recombinant TM analogs with thrombin, FH and C3b in order to analyze their ability to regulate serum complement activity. METHODS: Surface plasmon resonance (SPR) analysis was used to determine binding affinities of TM analogs with FH and C3b, and compared to thrombin as positive control. The capacity of the two recombinant TM analogs to regulate complement in serum was tested in standard complement hemolytic activity assays. RESULTS: SPR analysis showed that both TM analogs bind FH and C3b-Factor H with nanomolar and C3b with micromolar affinity; binding affinity for its natural ligand thrombin was several fold higher than for FH. At a physiological relevant concentration, TM inhibits complement hemolytic activity in serum via FH dependent and independent mechanisms. CONCLUSIONS: TM exhibits significant binding affinity for complement protein FH and C3b-FH complex and its soluble form is capable at physiologically relevant concentrations of inhibiting complement activation in serum.


Asunto(s)
Activación de Complemento/fisiología , Trombomodulina/metabolismo , Factor H de Complemento/metabolismo , Humanos , Unión Proteica , Resonancia por Plasmón de Superficie
3.
Mucosal Immunol ; 9(5): 1303-16, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26906404

RESUMEN

Human rhinovirus (HRV) infections are major contributors to the healthcare burden associated with acute exacerbations of chronic airway disease, such as chronic obstructive pulmonary disease and asthma. Cellular responses to HRV are mediated through pattern recognition receptors that may in part signal from membrane microdomains. We previously found Toll-like receptor signaling is reduced, by targeting membrane microdomains with a specific liposomal phosphatidylserine species, 1-stearoyl-2-arachidonoyl-sn-glycero-3-phospho-L-serine (SAPS). Here we explored the ability of this approach to target a clinically important pathogen. We determined the biochemical and biophysical properties and stability of SAPS liposomes and studied their ability to modulate rhinovirus-induced inflammation, measured by cytokine production, and rhinovirus replication in both immortalized and normal primary bronchial epithelial cells. SAPS liposomes rapidly partitioned throughout the plasma membrane and internal cellular membranes of epithelial cells. Uptake of liposomes did not cause cell death, but was associated with markedly reduced inflammatory responses to rhinovirus, at the expense of only modest non-significant increases in viral replication, and without impairment of interferon receptor signaling. Thus using liposomes of phosphatidylserine to target membrane microdomains is a feasible mechanism for modulating rhinovirus-induced signaling, and potentially a prototypic new therapy for viral-mediated inflammation.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Interacciones Huésped-Patógeno/efectos de los fármacos , Liposomas/farmacología , Fosfatidilserinas/farmacología , Mucosa Respiratoria/efectos de los fármacos , Rhinovirus/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras del Transporte Vesicular/deficiencia , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Línea Celular , Quimiocina CCL5/genética , Quimiocina CCL5/inmunología , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Células Epiteliales/inmunología , Células Epiteliales/virología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interferón beta/genética , Interferón beta/inmunología , Interleucina-8/genética , Interleucina-8/inmunología , Liposomas/síntesis química , Fosfatidilserinas/química , Éteres Fosfolípidos/química , Éteres Fosfolípidos/farmacología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/virología , Rhinovirus/crecimiento & desarrollo , Rhinovirus/inmunología , Transducción de Señal , Replicación Viral/efectos de los fármacos
4.
Allergy ; 70(1): 80-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25331546

RESUMEN

BACKGROUND: Alveolar macrophages are sentinels of the pulmonary mucosa and central to maintaining immunological homeostasis. However, their role in governing the response to allergen is not fully understood. Inappropriate responses to the inhaled environment manifest as asthma. METHODS: We utilized a mechanistic IL-13-driven model and a house dust mite allergen mucosal sensitization model of allergic airway disease to investigate the role of alveolar macrophages in regulating pulmonary inflammation. RESULTS: IL-13-dependent eosinophilic and Th2 inflammation was enhanced in mice depleted of alveolar macrophages using clodronate liposomes. Similarly, depletion of alveolar macrophages during house dust mite sensitization or established disease resulted in augmented Th2 immunity and increased allergen-specific IgG1 and IgE. Clodronate treatment also delayed the resolution of tissue inflammation following cessation of allergen challenge. Strikingly, tissue interstitial macrophages were elevated in alveolar macrophage-deficient mice identifying a new homeostatic relationship between different macrophage subtypes. A novel role for the macrophage-derived immunoregulatory cytokine IL-27 was identified in modulating Th2 inflammation following mucosal allergen exposure. CONCLUSIONS: In summary, alveolar macrophages are critical regulators of Th2 immunity and their dysregulation promotes an inflammatory environment with exacerbation of allergen-induced airway pathology. Manipulating IL-27 may provide a novel therapeutic strategy for the treatment of asthma.


Asunto(s)
Alérgenos/inmunología , Homeostasis , Pulmón/inmunología , Macrófagos Alveolares/inmunología , Animales , Antígenos Dermatofagoides/inmunología , Asma/inmunología , Asma/metabolismo , Asma/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Interleucina-13/metabolismo , Interleucina-13/farmacología , Interleucina-27/metabolismo , Leucocitos/inmunología , Leucocitos/metabolismo , Pulmón/metabolismo , Pulmón/patología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Ratones , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Células Th2/inmunología , Células Th2/metabolismo
5.
Arch Biochem Biophys ; 533(1-2): 55-61, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23500138

RESUMEN

Platelet activation represents a key event in normal hemostasis as well as during platelet plug formation related to thrombosis. Nitro-fatty acids are novel endogenously produced signaling mediators exerting pluripotent anti-inflammatory actions in cells and tissues. We have recently shown that nitroarachidonic acid inhibits thromboxane synthesis during platelet activation by affecting prostaglandin endoperoxide H synthase (PGHS). Herein, we investigated the regulation of human platelet activation by NO(2)AA and describe a novel mechanism involving protein kinase C (PKC) inhibition. NO(2)AA-mediated antiplatelet effects were characterized using mass spectrometry, confocal microscopy, flow cytometry, western blot and aggregometry. Incubation of NO(2)AA with human platelets caused a significant reduction in platelet sensitivity to thrombin, ADP, arachidonic acid (AA), and phorbol ester (PMA). These effects were cGMP-independent and did not involve Ca(2+) store-dependent mobilization. In contrast, signaling downstream of conventional PKC activation, such as α-granule secretion and extracellular signal regulated kinase 2 activation was strongly inhibited by NO(2)AA. Immunofluorescence confocal microscopy confirmed NO(2)AA-mediated inhibition of PKCα translocation to the membrane. In summary, we demonstrate that NO(2)AA inhibits platelet activation through modulation of PKCα activity as a potential novel mechanism for platelet regulation in vivo.


Asunto(s)
Ácido Araquidónico/farmacología , Activación Plaquetaria/efectos de los fármacos , Proteína Quinasa C/metabolismo , Animales , Ácido Araquidónico/metabolismo , Transporte Biológico , Bovinos , Humanos , Agregación Plaquetaria/efectos de los fármacos
6.
Platelets ; 24(7): 503-15, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22966961

RESUMEN

Raynaud's phenomenon (RP) describes the excessive vascular response of the digital vessels in response to cold exposure and emotional stress. It is typically the earliest manifestation of systemic sclerosis (SSc), a multisystem disease of unknown aetiology characterised by vasculopathy, inflammation and fibrosis. The biological actions of platelets are known to extend beyond primary haemostasis with a growing appreciation of their contribution to vascular function, inflammation and wound repair. This has led to a considerable body of work evaluating associations between platelet function analysis and RP/SSc. This review provides a conceptual framework upon which the potential contribution of platelets to vascular dysfunction, autoimmunity and tissue remodelling in RP and SSc is considered. We describe the existing evidence to support excessive platelet activation in RP and SSc, ranging from the early studies of platelet aggregability and circulating platelet-derived mediators, to the important findings of the recent work that has begun to explore the potential direct pathogenic role of platelets in established murine models of SSc. We shall describe and critically appraise the findings of previous therapeutic studies evaluating the use of anti-platelet agents in RP and SSc, along with their implications for future therapeutic intervention in these conditions.


Asunto(s)
Plaquetas/patología , Enfermedad de Raynaud/sangre , Esclerodermia Sistémica/sangre , Animales , Humanos
7.
Br J Pharmacol ; 167(3): 505-14, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22506619

RESUMEN

BACKGROUND AND PURPOSE: Excess morbidity/mortality in rheumatoid arthritis (RA) is associated with increased incidence of cardiovascular disease. In this 'proof-of-concept' study, vascular function was characterized in the murine collagen-induced arthritis (mCIA) model, the benchmark choice for evaluation of the pathological processes and assessment of new therapies. EXPERIMENTAL APPROACH: Mice in the very early stages of arthritis development [and appropriate naïve (non-immunized) age-matched controls] were used in the study. Blood pressure was measured using tail cuff plethysmography. Vascular function in rings of isolated aorta was studied with isometric tension myography. Levels of NO metabolites (NO(x)), MMP-9 protein and IL-1ß in plasma and MMP-9 protein in aortic homogenates were quantified. KEY RESULTS: Impaired vascular contractile responses in arthritis were unaffected by ex vivo inhibition of NOS (endothelial/neuronal and inducible) or COX activities. Endothelium-dependent and -independent relaxation, plasma NO(x) and blood pressure were unaffected by arthritis. Plasma and aortic homogenate MMP-9 protein levels were increased significantly in arthritis. Incubation of aortic tissues from naïve control animals with exogenous MMP-9 impaired subsequent contractile responses, mirroring that observed in arthritis. A role for IL-1ß in perpetuating contractile dysfunction and increasing aortic MMP-9 was excluded. CONCLUSIONS AND IMPLICATIONS: These data identify for the first time a relationship between early arthritis and contractile dysfunction and a possible role for MMP-9 therein, in the absence of overt endothelial dysfunction or increased NO production. As such, MMP-9 may constitute a significant target for early intervention in RA patients with a view to decreasing risk of cardiovascular disease.


Asunto(s)
Aorta Torácica/metabolismo , Artritis Experimental/fisiopatología , Metaloproteinasa 9 de la Matriz/metabolismo , Músculo Liso Vascular/metabolismo , Animales , Aorta Torácica/enzimología , Artritis Experimental/complicaciones , Presión Sanguínea , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/prevención & control , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Interleucina-1beta/sangre , Masculino , Ratones , Ratones Endogámicos DBA , Contracción Muscular , Miografía , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo
8.
Proc Natl Acad Sci U S A ; 98(14): 8006-11, 2001 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-11427723

RESUMEN

12/15-Lipoxygenase (LOX) activity is elevated in vascular diseases associated with impaired nitric oxide (( small middle dot)NO) bioactivity, such as hypertension and atherosclerosis. In this study, primary porcine monocytes expressing 12/15-LOX, rat A10 smooth muscle cells transfected with murine 12/15-LOX, and purified porcine 12/15-LOX all consumed *NO in the presence of lipid substrate. Suppression of LOX diene conjugation by *NO was also found, although the lipid product profile was unchanged. *NO consumption by porcine monocytes was inhibited by the LOX inhibitor, eicosatetraynoic acid. Rates of arachidonate (AA)- or linoleate (LA)-dependent *NO depletion by porcine monocytes (2.68 +/- 0.03 nmol x min(-1) x 10(6) cells(-1) and 1.5 +/- 0.25 nmol x min(-1) x 10(6) cells(-1), respectively) were several-fold greater than rates of *NO generation by cytokine-activated macrophages (0.1-0.2 nmol x min(-1) x 10(6) cells(-1)) and LA-dependent *NO consumption by primary porcine monocytes inhibited *NO activation of soluble guanylate cyclase. These data indicate that catalytic *NO consumption by 12/15-LOX modulates monocyte *NO signaling and suggest that LOXs may contribute to vascular dysfunction not only by the bioactivity of their lipid products, but also by serving as catalytic sinks for *NO in the vasculature.


Asunto(s)
Araquidonato 12-Lipooxigenasa/metabolismo , Guanilato Ciclasa/metabolismo , Monocitos/metabolismo , Óxido Nítrico/metabolismo , Animales , Catálisis , Técnicas de Cultivo de Célula , Activación Enzimática , Músculo Liso , Ratas , Especificidad por Sustrato , Porcinos , Transfección
9.
Circ Res ; 88(1): 12-21, 2001 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-11139468

RESUMEN

Nitric oxide ((.)NO) signaling pathways and lipid oxidation reactions are of central importance in both the maintenance of vascular homeostasis and the progression of vascular disease. Because both of these pathways involve free radical species that can also react together at extremely fast rates, convergent interactions between these pathways are expected. Biochemical and cell biology studies have defined multiple interactions of (.)NO with oxidizing lipids that could lead to either vascular protection or potentiation of inflammatory vascular injury. For example, low levels of (.)NO generated by endothelial nitric oxide synthase can terminate propagating lipid radicals and inhibit lipoxygenases, reactions that would be protective. Alternatively, if generated at elevated levels, for example, after inducible nitric oxide synthase expression in inflammation, (.)NO can be converted to prooxidant species, such as peroxynitrite (ONOO(-)) and nitrogen dioxide ((.)NO(2)), that can potentiate inflammatory injury to vascular cells. Finally, both enzymatic and nonenzymatic lipid oxidation reactions can influence (.)NO bioactivity by directly scavenging (.)NO or altering the induction and catalytic activity of nitric oxide synthase enzymes. In this review, we summarize the biochemical interactions between (.)NO and lipid oxidation reactions and discuss the recognized and potential roles of these reactions in the vasculature.


Asunto(s)
Peroxidación de Lípido , Óxido Nítrico/metabolismo , Animales , Humanos , Metabolismo de los Lípidos , Lípidos/química , Óxido Nítrico/química , Enfermedades Vasculares/metabolismo , Enfermedades Vasculares/patología
10.
Free Radic Res ; 35(5): 447-64, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11767404

RESUMEN

Nitric oxide (NO) is a major free radical modulator of smooth muscle tone, which under basal conditions acts to preserve vascular homeostasis through its anti-inflammatory properties. The biochemistry of NO, in particular, its rapid conversion in vivo into secondary reactive nitrogen species (RNS), its chemical nature as a free radical and its high diffusibility and hydrophobicity dictate that this species will interact with numerous biomolecules and enzymes. In this review, we consider the interactions of a number of enzymes found in the vasculature with NO and NO-derived RNS. All these enzymes are either homeostatic or promote the development of atherosclerosis and hypertension. Therefore their interactions with NO and NO-derived RNS will be of central importance in the initiation and progression of vascular disease. In some examples, (e.g. lipoxygenase, LOX), such interactions provide catalytic 'sinks' for NO, but for others, in particular peroxidases and prostaglandin H synthase (PGHS), reactions with NO may be detrimental. Nitric oxide and NO-derived RNS directly modulate the activity of vascular peroxidases and LOXs through a combination of effects, including transcriptional regulation, altering substrate availability, and direct reaction with enzyme turnover intermediates. Therefore, these interactions will have two major consequences: (i) depletion of NO levels available to cause vasorelaxation and prevent leukocyte/platelet adhesion and (ii) modulation of activity of the target enzymes, thereby altering the generation of bioactive signaling molecules involved in maintenance of vascular homeostasis, including prostaglandins and leukotrienes.


Asunto(s)
Lipooxigenasa/metabolismo , Óxido Nítrico/metabolismo , Peroxidasas/metabolismo , Especies de Nitrógeno Reactivo/metabolismo , Animales , Vasos Sanguíneos/metabolismo , Glutatión Peroxidasa/metabolismo , Homeostasis , Humanos , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo III , Prostaglandina-Endoperóxido Sintasas/metabolismo , Enfermedades Vasculares/etiología , Enfermedades Vasculares/metabolismo
11.
J Biol Chem ; 275(49): 38239-44, 2000 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-10993875

RESUMEN

Nitric oxide (( small middle dot)NO) plays a central role in vascular homeostasis via regulation of smooth muscle relaxation and platelet aggregation. Although mechanisms for ( small middle dot)NO formation are well known, removal pathways are less well characterized, particularly in cells that respond to ( small middle dot)NO through activation of soluble guanylate cyclase. Herein, we report that ( small middle dot)NO is catalytically consumed by prostaglandin H synthase-1 (PGHS-1) through acting as a reducing peroxidase substrate. With purified ovine PGHS-1, ( small middle dot)NO consumption requires peroxide (LOOH or H(2)O(2)), with a K(m)( (app)) for 15(S)hydroperoxyeicosatetraenoic acid (HPETE) of 3. 27 +/- 0.35 microm. During this, 2 mol ( small middle dot)NO are consumed per mol HPETE, and loss of HPETE hydroperoxy group occurs with retention of the conjugated diene spectrum. Hydroperoxide-stimulated ( small middle dot)NO consumption requires heme incorporation, is not inhibited by indomethacin, and is further stimulated by the reducing peroxidase substrate, phenol. PGHS-1-dependent ( small middle dot)NO consumption also occurs during arachidonate, thrombin, or activation of platelets (1-2 microm.min(-1) for typical plasma platelet concentrations) and prevents ( small middle dot)NO stimulation of platelet soluble guanylate cyclase. Platelet sensitivity to ( small middle dot)NO as an inhibitor of aggregation is greater using a platelet-activating stimulus () that does not cause ( small middle dot)NO consumption, indicating that this mechanism overcomes the anti-aggregatory effects of ( small middle dot)NO. Catalytic consumption of ( small middle dot)NO during eicosanoid synthesis thus represents both a novel proaggregatory function for PGHS-1 and a regulated mechanism for vascular ( small middle dot)NO removal.


Asunto(s)
Plaquetas/fisiología , Óxido Nítrico/metabolismo , Agregación Plaquetaria/fisiología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Animales , Ácido Araquidónico/farmacología , Catálisis , Bovinos , Ciclooxigenasa 1 , Glutatión/análogos & derivados , Glutatión/farmacología , Humanos , Peróxido de Hidrógeno/metabolismo , Isoenzimas/metabolismo , Cinética , Leucotrienos/metabolismo , Peróxidos Lipídicos/metabolismo , Proteínas de la Membrana , Compuestos Nitrosos/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , S-Nitrosoglutatión , Ovinos , Especificidad por Sustrato , Trombina/farmacología
12.
Arterioscler Thromb Vasc Biol ; 20(7): 1707-15, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10894807

RESUMEN

The regulation of nonenzymatic and enzymatic lipid oxidation reactions by nitric oxide (.NO) is potent and pervasive and reveals novel non-cGMP-dependent reactivities for this free radical inflammatory and signal transduction mediator.NO and its metabolites stimulate and inhibit lipid peroxidation reactions, modulate enzymatically catalyzed lipid oxidation, complex with lipid-reactive metals, and alter proinflammatory gene expression. Through these mechanisms,.NO can regulate nonenzymatic lipid oxidation and the production of inflammatory and vasoactive eicosanoids by prostaglandin endoperoxide synthase and lipoxygenase. The accumulation of macrophages and oxidized low density lipoprotein within the vascular wall can also be modulated by.NO. A key determinant of the pro-oxidant versus oxidant-protective influences of.NO is the underlying oxidative status of tissue. When.NO is in excess of surrounding oxidants, lipid oxidation and monocyte margination into the vascular wall are attenuated, producing antiatherogenic effects. However, when endogenous tissue rates of oxidant production are accelerated or when tissue oxidant defenses become depleted,.NO gives rise to secondary oxidizing species that can increase membrane and lipoprotein lipid oxidation as well as foam cell formation in the vasculature, thus promoting proatherogenic effects. In summary,.NO is a multifaceted molecule capable of reacting via multiple pathways to modulate lipid oxidation reactions, thereby impacting on tissue inflammatory reactions.


Asunto(s)
Arteriosclerosis/metabolismo , Endotelio Vascular/enzimología , Lipoproteínas/metabolismo , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico/metabolismo , Animales , Radicales Libres/metabolismo , Humanos , Metabolismo de los Lípidos , Nitratos/metabolismo , Oxidación-Reducción
13.
Free Radic Biol Med ; 28(7): 1017-29, 2000 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-10832063

RESUMEN

Manganese porphyrin complexes serve to catalytically scavenge superoxide, hydrogen peroxide, and peroxynitrite. Herein, reactions of manganese 5,10,15,20-tetrakis(N-ethylpyridinium-2-yl)porphyrin (MnTE-2-PyP(5+)) with lipids and lipid hydroperoxides (LOOH) are examined. In linoleic acid and human low-density lipoprotein (LDL), MnTE-2-PyP(5+) promotes oxidative reactions when biological reductants are not present. By redox cycling between Mn(+3) and Mn(+4) forms, MnTE-2-PyP(5+) initiates lipid peroxidation via decomposition of 13(S)hydroperoxyoctadecadienoic acid [13(S)HPODE], with a second-order rate constant of 8.9 x 10(3) M(-1)s(-1)and k(cat) = 0.32 s(-1). Studies of LDL oxidation demonstrate that: (i) MnTE-2-PyP(5+) can directly oxidize LDL, (ii) MnTE-2-PyP(5+) does not inhibit Cu-induced LDL oxidation, and (iii) MnTE-2-PyP(5+) plus a reductant partially inhibit lipid peroxidation. MnTE-2-PyP(5+) (1-5 microM) also significantly inhibits FeCl(3) plus ascorbate-induced lipid peroxidation of rat brain homogenate. In summary, MnTE-2-PyP(5+) initiates membrane lipid and lipoprotein oxidation in the absence of biological reductants, while MnTE-2-PyP(5+) inhibits lipid oxidation reactions initiated by other oxidants when reductants are present. It is proposed that, as the Mn(+3) resting redox state of MnTE-2-PyP(5+) becomes oxidized to the Mn(+4) redox state, LOOH is decomposed to byproducts that propagate lipid oxidation reactions. When the manganese of MnTE-2-PyP(5+) is reduced to the +2 state by biological reductants, antioxidant reactions of the metalloporphyrin are favored.


Asunto(s)
Lípidos/química , Lipoproteínas/química , Manganeso/química , Metaloporfirinas/química , Animales , Química Encefálica/efectos de los fármacos , Catálisis , Cromatografía en Capa Delgada , Depuradores de Radicales Libres/química , Depuradores de Radicales Libres/farmacología , Humanos , Ácido Linoleico/química , Ácidos Linoleicos/química , Peroxidación de Lípido , Peróxidos Lipídicos/química , Lipoproteínas LDL/sangre , Masculino , Espectrometría de Masas , Metaloporfirinas/farmacología , Oxidación-Reducción , Ratas , Ratas Sprague-Dawley
14.
J Biol Chem ; 274(29): 20083-91, 1999 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-10400618

RESUMEN

Analysis of purified soybean and rabbit reticulocyte 15-lipoxygenase (15-LOX) and PA317 cells transfected with human 15-LOX revealed a rapid rate of linoleate-dependent nitric oxide (.NO) uptake that coincided with reversible inhibition of product ((13S)-hydroperoxyoctadecadienoic acid, or (13S)-HPODE) formation. No reaction of .NO (up to 2 microM) with either native (Ered) or ferric LOXs (0.2 microM) metal centers to form nitrosyl complexes occurred at these .NO concentrations. During HPODE-dependent activation of 15-LOX, there was consumption of 2 mol of .NO/mol of 15-LOX. Stopped flow fluorescence spectroscopy showed that.NO (2.2 microM) did not alter the rate or extent of (13S)-HPODE-induced tryptophan fluorescence quenching associated with 15-LOX activation. Additionally, .NO does not inhibit the anaerobic peroxidase activity of 15-LOX, inferring that the inhibitory actions of .NO are due to reaction with the enzyme-bound lipid peroxyl radical, rather than impairment of (13S)-HPODE-dependent enzyme activation. From this, a mechanism of 15-LOX inhibition by .NO is proposed whereby reaction of .NO with EredLOO. generates Ered and LOONO, which hydrolyzes to (13S)-HPODE and nitrite (NO2-). Reactivation of Ered, considerably slower than dioxygenase activity, is then required to complete the catalytic cycle and leads to a net inhibition of rates of (13S)-HPODE formation. This reaction of .NO with 15-LOX inhibited. NO-dependent activation of soluble guanylate cyclase and consequent cGMP production. Since accelerated .NO production, enhanced 15-LOX gene expression, and 15-LOX product formation occurs in diverse inflammatory conditions, these observations indicate that reactions of .NO with lipoxygenase peroxyl radical intermediates will result in modulation of both .NO bioavailability and rates of production of lipid signaling mediators.


Asunto(s)
Araquidonato 15-Lipooxigenasa/metabolismo , Guanilato Ciclasa/metabolismo , Óxido Nítrico/metabolismo , Animales , Araquidonato 15-Lipooxigenasa/genética , Catálisis , Activación Enzimática , Humanos , Cinética , Ácido Linoleico/metabolismo , Oxidación-Reducción , Conejos , Glycine max/enzimología , Transfección
15.
Chem Res Toxicol ; 12(1): 83-92, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9894022

RESUMEN

Reactive nitrogen species derived from nitric oxide are potent oxidants formed during inflammation that can oxidize membrane and lipoprotein lipids in vivo. Herein, it is demonstrated that several of these species react with unsaturated fatty acid to yield nitrated oxidation products. Using HPLC coupled with both UV detection and electrospray ionization mass spectrometry, products of reaction of ONOO- with linoleic acid displayed mass/charge (m/z) characteristics of LNO2 (at least three products at m/z 324, negative ion mode). Further analysis by MS/MS gave a major fragment at m/z 46. Addition of a NO2 group was confirmed using [15N]ONOO- which gave a product at m/z 325, fragmenting to form a daughter ion at m/z 47. Formation of nitrated lipids was inhibited by bicarbonate, superoxide dismutase (SOD), and Fe3+-EDTA, while the yield of oxidation products was decreased by bicarbonate and SOD, but not by Fe3+-EDTA. Reaction of linoleic acid with both nitrogen dioxide (*NO2) or nitronium tetrafluoroborate (NO2BF4) also yielded nitrated lipid products (m/z 324), with HPLC retention times and MS/MS fragmentation patterns identical to the m/z 324 species formed by reaction of ONOO- with linoleic acid. Finally, reaction of HPODE, but not linoleate, with nitrous acid (HONO) or isobutyl nitrite (BuiONO) yielded a product at m/z 340, or 341 upon reacting with [15N]HONO. MS/MS analysis gave an NO2- fragment, and 15N NMR indicated that the product contained a nitro (RNO2) functional group, suggesting that the product was nitroepoxylinoleic acid [L(O)NO2]. This species could form via homolytic dissociation of LOONO to LO* and *NO2 and rearrangement of LO* to an epoxyallylic radical L(O)* followed by recombination of L(O)* with *NO2. Since unsaturated lipids of membranes and lipoproteins are critical targets of reactive oxygen and nitrogen species, these pathways lend insight into mechanisms for the formation of novel nitrogen-containing lipid products in vivo and provide synthetic strategies for further structural and functional studies.


Asunto(s)
Ácidos Grasos Insaturados/química , Nitratos/química , Óxido Nítrico/química , Dióxido de Nitrógeno/química , Ácido Nitroso/química , Oxidantes/química , Cromatografía Líquida de Alta Presión , Cromatografía Liquida , Concentración de Iones de Hidrógeno , Ácido Linoleico/química , Ácidos Linoleicos/química , Peróxidos Lipídicos/química , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Oxidación-Reducción
16.
Methods Enzymol ; 301: 454-70, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-9919594

RESUMEN

Reactions of linoleate (and presumably other unsaturated fatty acids) with reactive nitrogen species that form in biological systems from secondary reactions of .NO yield two main nitration product groups, LNO2 (formed by ONOO-, .NO2, or NO2+ reaction with linoleate), and LONO2 (formed by HONO reaction with 13(S)-HPODE, or .NO termination with LOO.). Comparison of HPLC retention times and m/z for lipid nitration products indicate that the mechanisms of nitrated product formation converge at several points: (i) The initial product of HONO attack on LOOH will be LOONO, which is identical to the initial termination product of LOO. reaction with .NO. (ii) Dissociation of LOONO to give LO. and .NO2 via caged radicals, which recombine to give LONO2 (m/z 340) will occur, regardless of how LOONO is formed (Fig. 7). (iii) In some experiments, the reaction of O2- (where oxidation is initiated by xanthine oxidase-derived O2- production and metal-dependent decomposition of H2O2) with .NO will result in generation of ONOO-. Nitration of unsaturated lipid by this species will yield a species demonstrated herein to be LNO2. Lipid oxidation leads to formation of bioactive products, including hydroxides, hydroperoxides, and isoprostanes. In vivo, nitrated lipids (LNO2, LONO2) may also possess bioactivity, for example through eicosanoid receptor binding activity, or by acting as antagonists/competitive inhibitors of eicosanoid receptor-ligand interactions. In addition, nitrated lipids could mediate signal transduction via direct .NO donation, transnitrosation, or following reductive metabolism. Similar bioactive products are formed following ONOO- reaction with glucose, glycerol, and other biomolecules.


Asunto(s)
Ácidos Grasos Insaturados/metabolismo , Óxido Nítrico/metabolismo , Animales , Cromatografía Líquida de Alta Presión/métodos , Radicales Libres/metabolismo , Humanos , Peroxidación de Lípido , Nitratos/metabolismo , Dióxido de Nitrógeno/metabolismo , Ácido Nitroso/metabolismo
17.
Mol Aspects Med ; 19(4-5): 221-357, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-10231805

RESUMEN

Since its initial discovery as an endogenously produced bioactive mediator, nitric oxide (.NO) has been found to play a critical role in the cellular function of nearly all organ systems. Furthermore, aberrant production of .NO or reactive nitrogen species (RNS) derived from .NO, has been implicated in a number of pathological conditions, such as acute lung disease, atherosclerosis and septic shock. While .NO itself is fairly non-toxic, secondary RNS are oxidants and nitrating agents that can modify both the structure and function of numerous biomolecules both in vitro, and in vivo. The mechanisms by which RNS mediate toxicity are largely dictated by its unique reactivity. The study of how reactive nitrogen species (RNS) derived from .NO interact with biomolecules such as proteins, carbohydrates and lipids, to modify both their structure and function is an area of active research, which is lending major new insights into the mechanisms underlying their pathophysiological role in human disease. In the context of .NO-dependent pathophysiology, these biochemical reactions will play a major role since they: (i) lead to removal of .NO and decreased efficiency of .NO as an endothelial-derived relaxation factor (e.g. in hypertension, atherosclerosis) and (ii) lead to production of other intermediate species and covalently modified biomolecules that cause injury and cellular dysfunction during inflammation. Although the physical and chemical properties of .NO and .NO-derived RNS are well characterised, extrapolating this fundamental knowledge to a complicated biological environment is a current challenge for researchers in the field of .NO and free radical research. In this review, we describe the impact of .NO and .NO-derived RNS on biological processes primarily from a biochemical standpoint. In this way, it is our intention to outline the most pertinent and relevant reactions of RNS, as they apply to a diverse array of pathophysiological states. Since reactions of RNS in vivo are likely to be vast and complex, our aim in this review is threefold: (i) address the major sources and reactions of .NO-derived RNS in biological systems, (ii) describe current knowledge regarding the functional consequences underlying .NO-dependent covalent modification of specific biomolecules, and (iii) to summarise and critically evaluate the available evidence implicating these reactions in human pathology. To this end, three areas of special interest have been chosen for detailed description, namely, formation and role of S-nitrosothiols, modulation of lipid oxidation/nitration by RNS, and tyrosine nitration mechanisms and consequences.


Asunto(s)
Óxido Nítrico/fisiología , Animales , Radicales Libres/metabolismo , Humanos , Metabolismo de los Lípidos , Óxido Nítrico/biosíntesis , Nitrógeno/metabolismo , Oxidación-Reducción , Compuestos de Sulfhidrilo/metabolismo , Tirosina/metabolismo
18.
Biochem J ; 327 ( Pt 1): 203-8, 1997 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-9355754

RESUMEN

The purified 15-lipoxygenase from rabbit reticulocytes is capable of oxidizing NADH in the presence of linoleic acid and oxygen. This co-oxidation proceeds at a rate that amounts to approx. 7% of linoleic acid oxygenation rates. Although NADH inhibits the lipoxygenase reaction with linoleic acid as substrate (46% inhibition at 0.2 mM NADH), the reaction specificity of the enzyme was not altered since (13S)-hydroperoxy-(9Z,11E)-octadecadienoic acid was identified as the major reaction product. NADH oxidation was inhibited by NAD+ (uncompetitive with respect to linoleate and mixed/competitive with respect to NADH), and NADPH or NMNH could substitute for NADH with slightly different apparent Km values. NADH oxidation was enhanced at lower oxygen tension, but was completely prevented under anaerobic conditions. Computer-assisted modelling of 15-lipoxygenase/NADH interaction and sequence alignments of mammalian lipoxygenases with NADH-dependent enzymes suggested that there is no specific binding of the coenzyme at the putative fatty acid-binding site of lipoxygenases. These results suggest that NAD(P)H might be oxidized by a radical intermediate formed during the dioxygenase cycle of the lipoxygenase reaction but that NADH oxidation might not proceed at the active site of the enzyme. The mechanism and possible biological consequences of 15-lipoxygenase-catalysed NAD(P)H oxidation are discussed.


Asunto(s)
Araquidonato 15-Lipooxigenasa/metabolismo , NADP/metabolismo , NAD/metabolismo , Animales , Sitios de Unión , Simulación por Computador , Ácidos Grasos Insaturados/metabolismo , Radicales Libres/metabolismo , Cinética , Ácido Linoleico/metabolismo , Peróxidos Lipídicos/química , Peróxidos Lipídicos/metabolismo , Inhibidores de la Lipooxigenasa/farmacología , NAD/farmacología , Oxidación-Reducción , Oxígeno/metabolismo , Conejos , Reticulocitos/enzimología , Superóxidos/metabolismo
19.
FASEB J ; 11(2): 181-8, 1997 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9039961

RESUMEN

Hydrogen peroxide (H2O2) or 4-hydroxy-2,3-trans-nonenal (HNE) treatment of rat vascular smooth muscle cells (A7r5) caused induction of aldose reductase mRNA. Induction was dose (10-100 microM H2O2, 1-10 microM HNE) and time dependent, reaching a maximum (three- to fourfold) after 7-12 h. Treatment of cells with actinomycin D confirmed de novo synthesis of aldose reductase mRNA. H2O2-induced expression was prevented by catalase but unaffected by Desferal, indicating that metal catalyzed degradation of peroxide was not involved. Induction of enzymatically active aldose reductase by H2O2 and HNE was confirmed using Western blotting and enzyme assays. Aldose reductase can metabolize several aldehyde compounds including HNE, a major toxic product of lipid peroxidation. Inclusion of Sorbinil, an aldose reductase inhibitor, in toxicity assays resulted in a significant (twofold) enhancement of HNE-mediated killing of A7r5 cells, suggesting a protective role of aldose reductase against HNE-induced cell death. These data indicate that the induction of aldose reductase during oxidative stress might represent an important cellular antioxidant defense mechanism.


Asunto(s)
Aldehído Reductasa/biosíntesis , Imidazolidinas , Músculo Liso/metabolismo , Estrés Oxidativo , Aldehído Reductasa/antagonistas & inhibidores , Aldehído Reductasa/genética , Aldehídos/farmacología , Aldehídos/toxicidad , Animales , Arsenitos/farmacología , Catalasa/farmacología , Células Cultivadas , Deferoxamina/farmacología , Inducción Enzimática/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Peróxido de Hidrógeno/toxicidad , Imidazoles/farmacología , Músculo Liso/citología , Músculo Liso/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , ARN/análisis , ARN Mensajero/biosíntesis , Ratas , Compuestos de Sodio/farmacología
20.
Biochemistry ; 36(49): 15216-23, 1997 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-9398249

RESUMEN

The reaction between nitric oxide (*NO) and lipid peroxyl radicals (LOO*) has been proposed to account for the potent inhibitory properties of *NO toward lipid peroxidation processes; however, the mechanisms of this reaction, including kinetic parameters and nature of termination products, have not been defined. Here, the reaction between linoleate peroxyl radicals and *NO was examined using 2, 2'-azobis(2-amidinopropane) hydrochloride-dependent oxidation of linoleate. Addition of *NO (0.5-20 microM) to peroxidizing lipid led to cessation of oxygen uptake, which resumed at original rates when all *NO had been consumed. At high *NO concentrations (>3 microM), the time of inhibition (Tinh) of chain propagation became increasingly dependent on oxygen concentration, due to the competing reaction of oxygen with *NO. Kinetic analysis revealed that a simple radical-radical termination reaction (*NO:ROO* = 1:1) does not account for the inhibition of lipid oxidation by *NO, and at least two molecules of *NO are consumed per termination reaction. A mechanism is proposed whereby *NO first reacts with LOO* (k = 2 x 10(9) M-1 s-1) to form LOONO. Following decomposition of LOONO to LO* and *NO2, a second *NO is consumed via reaction with LO*, with the composite rate constant for this reaction being k = 7 x 10(4) M-1 s-1. At equal concentrations, greater inhibition of oxidation was observed with *NO than with alpha-tocopherol. Since *NO reacts with LOO* at an almost diffusion-limited rate, steady state concentrations of 30 nM *NO would effectively compete with endogenous alpha-tocopherol concentrations (about 20 microM) as a scavenger of LOO* in the lipid phase. This indicates that biological *NO concentrations (up to 2 microM) will significantly influence peroxidation reactions in vivo.


Asunto(s)
Peroxidación de Lípido , Peróxidos Lipídicos/química , Óxido Nítrico/química , Vitamina E/química , Simulación por Computador , Radicales Libres , Cinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...