Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Antioxidants (Basel) ; 11(12)2022 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-36552550

RESUMEN

Parkinson's disease (PD) is due to the oxidation of alpha synuclein (αSyn) contributing to motor impairment. We developed a transgenic mouse model of PD that overexpresses the mutated human αSyn gene (A53T) crossed to a mouse expressing the human MPO gene. This model exhibits increased oxidation and chlorination of αSyn leading to greater motor impairment. In the current study, the hMPO-A53T mice were treated with thiocyanate (SCN-) which is a favored substrate of MPO as compared to chlorine. We show that hMPO-A53T mice treated with SCN- have less chlorination in the brain and show an improvement in motor skills compared to the nontreated hMPO-A53T mice. Interestingly, in the hMPO-A53T mice we found a possible link between MPO-related disease and the glymphatic system which clears waste including αSyn from the brain. The untreated hMPO-A53T mice exhibited an increase in the size of periventricular glymphatic vessels expressing the glymphatic marker LYVE1 and aquaporin 4 (AQP4). These vessels also exhibited an increase in MPO and HOCl-modified epitopes in the glymphatic vessels correlating with loss of ependymal cells lining the ventricles. These findings suggest that MPO may significantly promote the impairment of the glymphatic waste removal system thus contributing to neurodegeneration in PD. Moreover, the inhibition of MPO chlorination/oxidation by SCN- may provide a potential therapeutic approach to this disease.

2.
Free Radic Biol Med ; 141: 115-140, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31175983

RESUMEN

α-Synuclein (αSyn) is central to the neuropathology of Parkinson's disease (PD) due to its propensity for misfolding and aggregation into neurotoxic oligomers. Nitration/oxidation of αSyn leads to dityrosine crosslinking and aggregation. Myeloperoxidase (MPO) is an oxidant-generating enzyme implicated in neurodegenerative diseases. In the present work we have examined the impact of MPO in PD through analysis of postmortem PD brain and in a novel animal model in which we crossed a transgenic mouse expressing the human MPO (hMPO) gene to a mouse expressing human αSyn-A53T mutant (A53T) (hMPO-A53T). Surprisingly, our results show that in PD substantia nigra, the hMPO gene is expressed in neurons containing aggregates of nitrated αSyn as well as MPO-generated HOCl-modified epitopes. In our hMPO-A53T mouse model, we also saw hMPO expression in neurons but not mouse MPO. In the mouse model, hMPO was expressed in neurons colocalizing with nitrated αSyn, carbamylated lysine, nitrotyrosine, as well as HOCl-modified epitopes/proteins. RNAscope in situ hybridization confirmed hMPO mRNA expression in neurons. Interestingly, the hMPO protein expressed in hMPO-A53T brain is primarily the precursor proMPO, which enters the secretory pathway potentially resulting in interneuronal transmission of MPO and oxidative species. Importantly, the hMPO-A53T mouse model, when compared to the A53T model, exhibited significant exacerbation of motor impairment on rotating rods, balance beams, and wire hang tests. Further, hMPO expression in the A53T model resulted in earlier onset of end stage paralysis. Interestingly, there was a high concentration of αSyn aggregates in the stratum lacunosum moleculare of hippocampal CA2 region, which has been associated in humans with accumulation of αSyn pathology and neural atrophy in dementia with Lewy bodies. This accumulation of αSyn aggregates in CA2 was associated with markers of endoplasmic reticulum (ER) stress and the unfolded protein response with expression of activating transcription factor 4 (ATF4), C/EBP homologous protein (CHOP), MPO, and cleaved caspase-3. Together these findings suggest that MPO plays an important role in nitrative and oxidative damage that contributes to αSyn pathology in synucleinopathies.


Asunto(s)
Modelos Animales de Enfermedad , Trastornos Motores/enzimología , Neuronas/enzimología , Enfermedad de Parkinson/enzimología , Peroxidasa/metabolismo , Sustancia Negra/enzimología , Animales , Carbono/química , Epítopos/química , Femenino , Humanos , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Transgénicos , Trastornos Motores/fisiopatología , Destreza Motora , Nitrógeno/química , Estrés Oxidativo , Enfermedad de Parkinson/fisiopatología , Peroxidasa/genética , Sustancia Negra/citología
3.
J Biol Chem ; 293(17): 6374-6386, 2018 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-29496995

RESUMEN

Protein carbamylation by cyanate is a post-translational modification associated with several (patho)physiological conditions, including cardiovascular disorders. However, the biochemical pathways leading to protein carbamylation are incompletely characterized. This work demonstrates that the heme protein myeloperoxidase (MPO), which is secreted at high concentrations at inflammatory sites from stimulated neutrophils and monocytes, is able to catalyze the two-electron oxidation of cyanide to cyanate and promote the carbamylation of taurine, lysine, and low-density lipoproteins. We probed the role of cyanide as both electron donor and low-spin ligand by pre-steady-state and steady-state kinetic analyses and analyzed reaction products by MS. Moreover, we present two further pathways of carbamylation that involve reaction products of MPO, namely oxidation of cyanide by hypochlorous acid and reaction of thiocyanate with chloramines. Finally, using an in vivo approach with mice on a high-fat diet and carrying the human MPO gene, we found that during chronic exposure to cyanide, mimicking exposure to pollution and smoking, MPO promotes protein-bound accumulation of carbamyllysine (homocitrulline) in atheroma plaque, demonstrating a link between cyanide exposure and atheroma. In summary, our findings indicate that cyanide is a substrate for MPO and suggest an additional pathway for in vivo cyanate formation and protein carbamylation that involves MPO either directly or via its reaction products hypochlorous acid or chloramines. They also suggest that chronic cyanide exposure could promote the accumulation of carbamylated proteins in atherosclerotic plaques.


Asunto(s)
Cianatos , Cianuros , Peroxidasa , Placa Aterosclerótica/enzimología , Carbamilación de Proteína , Animales , Citrulina/análogos & derivados , Citrulina/química , Citrulina/genética , Citrulina/metabolismo , Cianatos/química , Cianatos/metabolismo , Cianuros/química , Cianuros/metabolismo , Humanos , Ratones , Ratones Noqueados , Oxidación-Reducción , Peroxidasa/química , Peroxidasa/genética , Peroxidasa/metabolismo , Placa Aterosclerótica/genética , Placa Aterosclerótica/patología
4.
PLoS One ; 11(2): e0149391, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26890638

RESUMEN

Among the human heme-peroxidase family, myeloperoxidase (MPO) has a unique disulfide-linked oligomeric structure resulting from multi-step processing of the pro-protein monomer (proMPO) after it exits the endoplasmic reticulum (ER). Related family members undergo some, but not all, of the processing steps involved with formation of mature MPO. Lactoperoxidase has its pro-domain proteolytically removed and is a monomer in its mature form. Eosinophil peroxidase undergoes proteolytic removal of its pro-domain followed by proteolytic separation into heavy and light chains and is a heterodimer. However, only MPO undergoes both these proteolytic modifications and then is further oligomerized into a heterotetramer by a single inter-molecular disulfide bond. The details of how and where the post-ER processing steps of MPO occur are incompletely understood. We report here that T47D breast cancer cells stably transfected with an MPO expression plasmid are able to efficiently replicate all of the processing steps that lead to formation of the mature MPO heterotetramer. MPO also traffics to the lysosome granules of T47D cells where it accumulates, allowing in-depth immunofluorescent microscopy studies of MPO trafficking and storage for the first time. Using this novel cell model we show that formation of MPO's single inter-molecular disulfide bond can occur normally in the absence of the proteolytic events that lead to separation of the MPO heavy and light chains. We further demonstrate that Cys319, which forms MPO's unique inter-molecular disulfide bond, is important for events that precede this step. Mutation of this residue alters the glycosylation and catalytic activity of MPO and blocks its entry into the endocytic pathway where proteolytic processing and disulfide bonding occur. Finally, using the endocytic trafficking of lysosomal hydrolases as a guide, we investigate the role of candidate receptors in the endocytic trafficking of MPO.


Asunto(s)
Expresión Génica , Lisosomas/metabolismo , Peroxidasa/genética , Peroxidasa/metabolismo , Procesamiento Proteico-Postraduccional , Catepsinas/antagonistas & inhibidores , Línea Celular , Línea Celular Tumoral , Cistina/metabolismo , Disulfuros , Retículo Endoplásmico , Activación Enzimática , Técnicas de Silenciamiento del Gen , Glicosilación , Humanos , Espacio Intracelular , Manosafosfatos/metabolismo , Mutación , Peroxidasa/química , Multimerización de Proteína , Transporte de Proteínas , Proteolisis , ARN Interferente Pequeño/genética
5.
Tumour Biol ; 35(1): 141-8, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23893381

RESUMEN

Myeloperoxidase (MPO) is an oxidant generating enzyme normally restricted to myeloid cells, however aberrant MPO expression has been found to occur in non-myeloid cells in some disease states. The functional -463GA promoter polymorphism alters MPO expression levels. The -463G is within an SP1 binding site and is associated with higher gene expression. The G allele is most frequent with ~62% of European populations being GG homozygotes. The GA polymorphism has been associated with risk or survival in a variety of cancers including lung and breast cancer. In this study we determined the frequency of the -463G/A polymorphism in 230 ovarian cancer patients, 75 patients with borderline ovarian tumors, and 299 healthy controls. The GG genotype was found to be overrepresented in patients with early stage ovarian cancer (83.3% GG, p = 0.008) as compared to healthy controls (62% GG), suggesting that MPO oxidants may increase risk. Immunohistochemical analysis revealed MPO expression in a subset of columnar ovarian epithelial carcinoma cells in early stage carcinomas.


Asunto(s)
Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Peroxidasa/genética , Alelos , Estudios de Casos y Controles , Femenino , Expresión Génica , Frecuencia de los Genes , Genotipo , Humanos , Ganglios Linfáticos/patología , Metástasis de la Neoplasia , Estadificación de Neoplasias , Neoplasias Ováricas/metabolismo , Peroxidasa/metabolismo , Polimorfismo Genético , Regiones Promotoras Genéticas
6.
NMR Biomed ; 24(8): 1023-8, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21538638

RESUMEN

MR techniques using hyperpolarized (13)C have successfully produced examples of angiography and intermediary metabolic imaging, but, to date, no receptor imaging has been attempted. The goal of this study was to synthesize and evaluate a novel hyperpolarizable molecule, 2,2,3,3-tetrafluoropropyl 1-(13)C-propionate-d(2,3,3) (TFPP), for the detection of atheromatous plaques in vivo. TFPP binds to lipid bilayers and its use in hyperpolarized MR could prove to be a major step towards receptor imaging. The precursor, 2,2,3,3-tetrafluoropropyl 1-(13)C-acrylate-d(2,3,3) (TFPA), binds to 1,2-dimyristoylphosphatidylcholine lipid bilayers with a 1.6-ppm chemical shift in the (19)F MR spectrum. This molecule was designed to be hyperpolarized through the addition of parahydrogen to the (13)C-acrylate moiety by parahydrogen-induced polarization. TFPA was hyperpolarized to TFPP to an extent similar to that of the hydroxyethylacrylate to hydroxyethylpropionate transition: 17 ± 4% for TFPP versus 20% for hydroxyethylpropionate; T(1) relaxation times (45 ± 2 s versus 55 ± 2 s) were comparable and the hyperpolarized properties of TFPP were characterized. Hydroxyethylacrylate, like TFPA, has a chemical structure with an acrylate moiety, but does not contain the lipid-binding tetrafluoropropyl functional group. Hyperpolarized TFPP binds to the lipid bilayer, appearing as a second, chemically shifted (13)C hyperpolarized MR signal with a further reduction in the longitudinal relaxation time (T(1) = 21 ± 1 s). In aortas harvested from low-density lipoprotein receptor knock-out mice fed with a high-fat diet for 9 months, and in which atheroma is deposited in the aorta and heart, TFPP showed greater binding to lipid on the intimal surface than in control mice fed a normal diet. When TFPP was hyperpolarized and administered in vivo to atheromatous mice in a pilot study, increased binding was observed on the endocardial surface of the intact heart compared with normally fed controls. Hyperpolarized TFPP has bio-sensing specificity for lipid, coupled with a 42,000-fold sensitivity gain in the MR signal at 4.7 T. Binding of TFPP with lipids results in the formation of a characteristic second peak in MRS. TFPP therefore has the potential to act as an in vivo molecular probe for atheromatous plaque imaging and may serve as a model of receptor-targeted bio-imaging with enhanced MR sensitivity.


Asunto(s)
Hidrógeno/química , Imagen por Resonancia Magnética/métodos , Espectroscopía de Resonancia Magnética/métodos , Placa Aterosclerótica/metabolismo , Receptores de LDL/análisis , Animales , Ratones , Ratones Endogámicos C57BL , Putrescina/análogos & derivados , Putrescina/metabolismo , Receptores de LDL/deficiencia
7.
J Biol Chem ; 284(5): 3158-3169, 2009 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-19059911

RESUMEN

Myeloperoxidase (MPO) is expressed in Alzheimer disease (AD) but not normal aged brain. A functional -463G/A MPO promoter polymorphism has been associated with AD risk through as yet unidentified mechanisms. Here we report that human MPO-463G allele, but not MPO-463A or mouse MPO, is strongly expressed in astrocytes and deposited in plaques in huMPO transgenic mice crossed to the APP23 model. MPO is similarly expressed in astrocytes in human AD tissue. In cortical homogenates of the MPOG-APP23 model, MPO expression correlated with increased levels of a lipid peroxidation product, 4-hydroxynonenal. Fluorescence high-performance liquid chromatography and electrospray ionization mass spectroscopy identified selective accumulation of phospholipid hydroperoxides in two classes of anionic phospholipids, phosphatidylserine (PS-OOH) and phosphatidylinositol (PI-OOH). The same molecular species of PS-OOH and PI-OOH were elevated in human AD brains as compared with non-demented controls. Augmented lipid peroxidation in MPOG-APP23 mice correlated with greater memory deficits. We suggest that aberrant huMPO expression in astrocytes leads to a specific pattern of phospholipid peroxidation and neuronal dysfunction contributing to AD.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Astrocitos/enzimología , Modelos Animales de Enfermedad , Trastornos de la Memoria/enzimología , Peroxidasa/metabolismo , Fosfolípidos/metabolismo , Alelos , Animales , Encéfalo/enzimología , Cromatografía Líquida de Alta Presión , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunohistoquímica , Hibridación in Situ , Peroxidación de Lípido , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Peroxidasa/genética , Espectrometría de Fluorescencia , Espectrometría de Masa por Ionización de Electrospray
8.
Nat Med ; 13(10): 1176-84, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17828273

RESUMEN

Post-translational modification and functional impairment of proteins through carbamylation is thought to promote vascular dysfunction during end-stage renal disease. Cyanate, a reactive species in equilibrium with urea, carbamylates protein lysine residues to form epsilon-carbamyllysine (homocitrulline), altering protein structure and function. We now report the discovery of an alternative and quantitatively dominant mechanism for cyanate formation and protein carbamylation at sites of inflammation and atherosclerotic plaque: myeloperoxidase-catalyzed oxidation of thiocyanate, an anion abundant in blood whose levels are elevated in smokers. We also show that myeloperoxidase-catalyzed lipoprotein carbamylation facilitates multiple pro-atherosclerotic activities, including conversion of low-density lipoprotein into a ligand for macrophage scavenger receptor A1 recognition, cholesterol accumulation and foam-cell formation. In two separate clinical studies (combined n = 1,000 subjects), plasma levels of protein-bound homocitrulline independently predicted increased risk of coronary artery disease, future myocardial infarction, stroke and death. We propose that protein carbamylation is a mechanism linking inflammation, smoking, uremia and coronary artery disease pathogenesis.


Asunto(s)
Aterosclerosis/metabolismo , Proteínas Sanguíneas/metabolismo , Carbamatos/metabolismo , Inflamación/patología , Fumar/metabolismo , Uremia/sangre , Animales , Aterosclerosis/patología , Colesterol/biosíntesis , Citrulina/análogos & derivados , Citrulina/biosíntesis , Citrulina/sangre , Ensayos Clínicos como Asunto , Enfermedad de la Arteria Coronaria/etiología , Enfermedad de la Arteria Coronaria/patología , Cianatos/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Células Espumosas/citología , Humanos , Células Jurkat , Lipoproteínas LDL/metabolismo , Lipoproteínas LDL/farmacología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Oxidación-Reducción , Peroxidasa/metabolismo , Valor Predictivo de las Pruebas , Procesamiento Proteico-Postraduccional , Receptores de LDL/metabolismo , Receptores Depuradores de Clase A/metabolismo
9.
Biochem Biophys Res Commun ; 349(2): 846-54, 2006 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-16956579

RESUMEN

Myeloperoxidase (MPO) is an oxidant-generating enzyme expressed in macrophages and implicated in atherosclerosis and cholesterol homeostasis. LXRalpha and PPARalpha regulate genes involved in cholesterol metabolism and the inflammatory response in macrophages. Here, we examine the effect of LXR and PPARalpha ligands on MPO expression. LXR and PPARalpha, as heterodimers with RXR, are shown to bind overlapping sites in an Alu receptor response element (AluRRE) in the MPO promoter. The LXR ligand T0901317 suppresses MPO mRNA expression in primary human macrophages, and in bone marrow cells and macrophages from huMPO transgenic mice. The PPARalpha ligand GW9578 downregulates MPO expression in GMCSF-macrophages, while upregulating in MCSF-macrophages. In contrast, the mouse MPO gene, which lacks the primate-specific AluRRE, is not regulated by LXR or PPARalpha ligands. These findings identify human MPO as a novel LXR and PPARalpha target gene, consistent with the role of these receptors in regulation of proinflammatory genes in macrophages.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , PPAR alfa/metabolismo , Peroxidasa/biosíntesis , Peroxidasa/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Aterosclerosis/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Núcleo Celular/metabolismo , Humanos , Hidrocarburos Fluorados , Leucocitos Mononucleares/metabolismo , Receptores X del Hígado , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores Nucleares Huérfanos , Sulfonamidas/farmacología
10.
Mediators Inflamm ; 2006(2): 36735, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16883063

RESUMEN

The severity of cystic fibrosis (CF) pulmonary disease is not directly related to CFTR genotype but depends upon several parameters, including neutrophil-dominated inflammation. Identification of agents modulating inflammation constitutes a relevant goal. Myeloperoxidase (MPO) is involved in both microbicidal and proinflammatory neutrophil activities. The aim of this study was to evaluate whether the -463GA MPO promoter polymorphism is linked to clinical severity of CF-associated pulmonary inflammation. This polymorphism significantly affects the level of MPO gene expression in leukocytes and the G allele is more expressing than the A allele. We show that MPO genotype significantly influences the severity of pulmonary disease in early stages, prior to the development of chronic lung infections, with GG genotype being associated with more severe CF disease. Our findings indicate that the level of MPO gene expression influences the CF pathogenesis, presumably reflecting cellular damage by MPO-generated oxidants or other activity of MPO in airway inflammation.


Asunto(s)
Fibrosis Quística/sangre , Fibrosis Quística/enzimología , Regulación Enzimológica de la Expresión Génica , Peroxidasa/genética , Polimorfismo Genético , Regiones Promotoras Genéticas , Adolescente , Adulto , Alelos , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Femenino , Genotipo , Humanos , Pulmón/metabolismo , Masculino , Factores Sexuales
11.
J Lipid Res ; 47(7): 1366-77, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16639078

RESUMEN

Myeloperoxidase (MPO) is an oxidant-generating enzyme present in macrophages at atherosclerotic lesions and implicated in coronary artery disease (CAD). Although mouse models are important for investigating the role of MPO in atherosclerosis, neither mouse MPO nor its oxidation products are detected in lesions in murine models. To circumvent this problem, we generated transgenic mice expressing two functionally different human MPO alleles, with either G or A at position -463, and crossed these to the LDL receptor-deficient (LDLR(-/-)) mouse. The -463G allele is linked to higher MPO expression and increased CAD incidence in humans. Both MPO alleles were expressed in a subset of lesions in high-fat-fed LDLR(-/-) mice, notably at necrotic lesions with cholesterol clefts. MPOG-expressing LDLR(-/-) males (but not females) developed significantly higher serum cholesterol, triglycerides, and glucose, all correlating with increased weight gain/obesity, implicating MPO in lipid homeostasis. The MPOG- and MPOA-expressing LDLR(-/-) males also exhibited significantly larger aortic lesions than control LDLR(-/-) males. The human MPO transgenic model will facilitate studies of MPO involvement in atherosclerosis and lipid homeostasis.


Asunto(s)
Aterosclerosis/enzimología , Aterosclerosis/genética , Hiperlipidemias/enzimología , Hiperlipidemias/genética , Obesidad/enzimología , Obesidad/genética , Peroxidasa/genética , Peroxidasa/metabolismo , Alelos , Animales , Aterosclerosis/patología , Grasas de la Dieta/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Humanos , Hiperlipidemias/patología , Hígado/enzimología , Hígado/patología , Macrófagos/enzimología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Obesidad/patología , Regiones Promotoras Genéticas , Receptores de LDL/deficiencia , Receptores de LDL/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Caracteres Sexuales
12.
Nitric Oxide ; 13(1): 42-53, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15893945

RESUMEN

Nitric oxide (NO) plays key roles in vasodilation and host defense, yet the overproduction of NO by inducible nitric oxide synthase (iNOS) at inflammatory sites can also be pathogenic. Here, we investigate the role of MPO in modulating the induction of iNOS by IFNgamma/LPS (IL). In monocyte-macrophages (Mvarphi) treated with IL, MPO gene expression was found to be downregulated as iNOS was upregulated. In Mvarphi from MPO-knockout (KO) mice, the induction of iNOS by IL was earlier and higher than in MPO-positive cells, suggesting MPO is inhibitory. Consistent with that interpretation, the addition of purified MPO enzyme to cultured macrophages inhibited iNOS induction by IL. In addition, an inhibitor of MPO enzyme, 4-aminobenzohydrazide, enhanced iNOS induction in MPO-positive cells, but not in MPO-KO cells. Similarly, taurine, a scavenger of MPO-generated HOCl, enhanced iNOS induction in MPO-positive cells, but not in MPO-KO cells. MPO affects an early event, suppressing iNOS induction when added within 2h of IL, but not when added several hours after IL. The suppression by MPO was alleviated by NO donor, sodium nitroprusside, suggesting the suppression results from scavenging of NO by MPO. This interpretation is consistent with earlier reports that MPO consumes NO, and that low levels of NO donor augment induction of iNOS by IFNgamma/LPS. The implication of these findings is that MPO acts as gatekeeper, suppressing the deleterious induction of iNOS at inflammatory sites by illegitimate signals. The combined signaling of IFNgamma/LPS overrides the gatekeeper function by suppressing MPO gene expression.


Asunto(s)
Macrófagos/enzimología , Óxido Nítrico Sintasa/metabolismo , Peroxidasa/farmacología , Animales , Regulación hacia Abajo , Inducción Enzimática/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica , Humanos , Interferón gamma/farmacología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Ratones , Ratones Noqueados , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo II , Nitritos/metabolismo , Peroxidasa/metabolismo , Regulación hacia Arriba
13.
Biochem Biophys Res Commun ; 331(2): 442-51, 2005 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-15850779

RESUMEN

Statins, inhibitors of HMG-CoA reductase, have pleiotropic benefits independent of cholesterol levels, including anti-oxidant and anti-inflammatory effects. Here, we investigate the effect of statins on myeloperoxidase (MPO) expression. MPO, expressed in foam cell macrophages, was recently shown to oxidize the ApoA-1 component of HDL, impairing ABCA-1 mediated cholesterol efflux. High levels of serum MPO correlate with increased risk of CAD events. Findings here show that statins strongly inhibit MPO mRNA expression in human and murine monocyte-macrophages. Suppression was reversed by downstream intermediates of HMG-CoA reductase, mevalonate, and geranylgeranylpyrophosphate, but not farnesylpyrophosphate. An inhibitor of geranylgeranyltransferase, GGTI-286, mimics the effects of statins, indicating geranylgeranylation is key to MPO expression. Reduction of MPO mRNA levels was observed in vivo in leukocytes from statin-fed mice, correlating with reductions in MPO protein and enzyme activity. These findings suggest that the pleiotropic protections afforded by statins may be due in part to suppression of MPO expression.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Peroxidasa/genética , Alelos , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Células Cultivadas , Humanos , Macrófagos/metabolismo , Ácido Mevalónico/farmacología , Ratones , Ratones Transgénicos , ARN Mensajero/genética , ARN Mensajero/metabolismo
15.
J Biol Chem ; 279(9): 8300-15, 2004 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-14668325

RESUMEN

A functional myeloperoxidase (MPO) promoter polymorphism, -463GA, has been associated with incidence or severity of inflammatory diseases, including atherosclerosis and Alzheimer's disease, and some cancers. The polymorphism is within an Alu element encoding four hexamer repeats recognized by nuclear receptors (AluRRE). Here we show that peroxisome proliferator-activated receptor gamma (PPARgamma) agonists strongly regulate MPO gene expression through the AluRRE. Opposite effects were observed in granulocyte/macrophage colony-stimulating factor (GMCSF)- versus macrophage colony-stimulating factor (MCSF)-derived macrophages (Mphi): Expression was markedly up-regulated (mean 26-fold) in MCSF-Mphi and down-regulated (34-fold) in GMCSF-Mphi. This was observed with rosiglitazone and three other PPARgamma ligands of the thiazolidinedione class, as well as the natural prostaglandin metabolite 15-deoxy-Delta(12,14) prostaglandin J(2). The selective PPARgamma antagonist, GW9662, blocked both the positive and negative effects on MPO expression. Gel retardation assays showed PPARgamma bound hexamers 3/4, and estrogen receptor-alpha bound hexamers 1/2, with -463A in hexamer 1 enhancing binding. Estrogen blocked PPARgamma effects on MPO expression, especially for the A allele. Charcoal filtration of fetal calf serum eliminated the block of PPARgamma, whereas replenishing the medium with 17beta-estradiol reinstated the block. These findings suggest a model in which estrogen receptor binds the AluRRE, preventing PPARgamma binding to the adjacent site. The positive and negative regulation by PPARgamma ligands, and the block by estrogen, was also observed in transgenic mice expressing the G and A alleles. The mouse MPO gene, which lacks the primate-specific AluRRE, was unresponsive to PPARgamma ligands, suggesting the human MPO transgenes will enhance the utility of mouse models for diseases involving MPO, such as atherosclerosis and Alzheimer's.


Asunto(s)
Estrógenos/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Macrófagos/enzimología , Peroxidasa/genética , Polimorfismo Genético , Prostaglandina D2/análogos & derivados , Receptores Citoplasmáticos y Nucleares/agonistas , Factores de Transcripción/agonistas , Elementos Alu/genética , Animales , Arteriosclerosis/enzimología , Sitios de Unión , Células de la Médula Ósea/enzimología , Células Cultivadas , Estradiol/farmacología , Receptor alfa de Estrógeno , Genotipo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Factor Estimulante de Colonias de Macrófagos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Peroxidasa/análisis , Regiones Promotoras Genéticas/genética , Prostaglandina D2/farmacología , ARN Mensajero/análisis , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de LDL/deficiencia , Receptores de Ácido Retinoico/metabolismo , Receptores X Retinoide , Rosiglitazona , Tiazolidinedionas/farmacología , Factores de Transcripción/metabolismo
16.
Lung Cancer ; 37(1): 35-40, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12057865

RESUMEN

Myeloperoxidase (MPO) is a metabolic/oxidative enzyme found in neutrophils and monocytes that contributes to pulmonary carcinogenesis through activation of specific procarcinogens including benzo[a]pyrene intermediates, 4-aminobiphenyl and the arylamines. There is a G-->A polymorphism located in the 5' untranslated region of the MPO gene that may be responsible for reduced transcriptional activity due to the decreased binding affinity for the SP1 transcription factor. Individuals with one or two copies of the A-allele may be afforded protection due to decreased transcriptional activity of MPO and subsequent decreased metabolic activation of procarcinogens. Previous studies have reported a range of protective effects in different ethnic populations. We employed a restriction fragment length polymorphism-polymerase chain reaction (RFLP-PCR) assay to identify the MPO genotypes in 375 lung cancer cases and 378 healthy controls, all of whom were Caucasian. Our results demonstrate a reduced risk of lung cancer when the A-allele genotypes (G/A+A/A) were combined (odds ratio (OR)=0.66; 95% confidence interval (CI) 0.49-0.90). We also noted a protective effect (OR=0.63; 95% CI 0.45-0.87) in ever smokers with the A-allele genotypes which was not evident in never smokers (OR=1.14; 95% CI 0.42-3.11). We observed an incremental decrease in the protective effects as cigarette pack-years increased. Thus, lightest smokers were provided the greatest protection. When the data were stratified by gender, there was a statistically significant reduced risk of lung cancer among men (OR=0.55; 95% CI 0.36-0.84), but not among women (OR=0.81; 95% CI 0.55-1.26) for the A-allele genotypes. Lastly, an age effect was evident only in men but not women. The protective effects of the A-allele genotypes decreased with increasing age. This report provides further support for the hypothesis that a single nucleotide polymorphism in the MPO gene is a protective factor in lung cancer carcinogenesis.


Asunto(s)
Predisposición Genética a la Enfermedad , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/prevención & control , Peroxidasa/genética , Polimorfismo Genético , Factores de Edad , Anciano , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Mutación Puntual , Reacción en Cadena de la Polimerasa , Polimorfismo de Longitud del Fragmento de Restricción , Factores Sexuales , Fumar/efectos adversos
17.
Clin Immunol ; 103(2): 154-60, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12027420

RESUMEN

Wegener's granulomatosis, microscopic polyangiitis, and Churg Strauss syndrome are forms of systemic vasculitides in which neutrophils and monocyte macrophages infiltrate the walls of small blood vessels, leading to destruction and occlusion. These diseases are associated with autoantibodies directed against granular components of neutrophils and monocytes, i.e., antineutrophil cytoplasmic antibodies (ANCA). The most common target antigens of ANCA in these vasculitides are myeloperoxidase (MPO) and proteinase 3 (PR3). ANCA-stimulated neutrophils injure endothelial cells, a process that is dependent upon the production of reactive oxygen radicals and the release of granular components such as MPO and PR3. Here we investigate whether a common functional MPO promoter polymorphism (-463 G/A) is associated with increased incidence and clinical aspects of ANCA-associated small vessel vasculitis. Genotyping was carried out for 142 patients with ANCA-associated small vessel vasculitis and 129 ethnically matched controls. The GG genotype was found to be associated with an increased risk for MPO-ANCA-associated vasculitis in females (86% GG, P = 0.045), but not males (64% GG, P = 1.0). Interestingly, the MPO A allele is associated with an increased incidence of relapses (P = 0.012) and an earlier age at diagnosis (P = 0.03) of MPO-ANCA-associated vasculitis. Both these associations are specific for MPO-ANCA and are not observed in patients with PR3-ANCA-associated vasculitis. These findings suggest that MPO expression levels influence the disease course of MPO-ANCA-associated vasculitis and further support the view that genetic factors are involved in the pathophysiology of this autoimmune disease.


Asunto(s)
Peroxidasa/genética , Polimorfismo Genético , Vasculitis/enzimología , Vasculitis/genética , Anticuerpos Anticitoplasma de Neutrófilos/metabolismo , Síndrome de Churg-Strauss/enzimología , Síndrome de Churg-Strauss/genética , Síndrome de Churg-Strauss/inmunología , Genotipo , Glomerulonefritis/enzimología , Glomerulonefritis/genética , Glomerulonefritis/inmunología , Granulomatosis con Poliangitis/enzimología , Granulomatosis con Poliangitis/genética , Granulomatosis con Poliangitis/inmunología , Humanos , Monocitos/inmunología , Neutrófilos/inmunología , Fenotipo , Vasculitis/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA