Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 44(2): e20-e38, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38095105

RESUMEN

BACKGROUND: High cholesterol levels in pancreatic ß-cells cause oxidative stress and decrease insulin secretion. ß-cells can internalize apo (apolipoprotein) A-I, which increases insulin secretion. This study asks whether internalization of apoA-I improves ß-cell insulin secretion by reducing oxidative stress. METHODS: Ins-1E cells were cholesterol-loaded by incubation with cholesterol-methyl-ß-cyclodextrin. Insulin secretion in the presence of 2.8 or 25 mmol/L glucose was quantified by radioimmunoassay. Internalization of fluorescently labeled apoA-I by ß-cells was monitored by flow cytometry. The effects of apoA-I internalization on ß-cell gene expression were evaluated by RNA sequencing. ApoA-I-binding partners on the ß-cell surface were identified by mass spectrometry. Mitochondrial oxidative stress was quantified in ß-cells and isolated islets with MitoSOX and confocal microscopy. RESULTS: An F1-ATPase ß-subunit on the ß-cell surface was identified as the main apoA-I-binding partner. ß-cell internalization of apoA-I was time-, concentration-, temperature-, cholesterol-, and F1-ATPase ß-subunit-dependent. ß-cells with internalized apoA-I (apoA-I+ cells) had higher cholesterol and cell surface F1-ATPase ß-subunit levels than ß-cells without internalized apoA-I (apoA-I- cells). The internalized apoA-I colocalized with mitochondria and was associated with reduced oxidative stress and increased insulin secretion. The IF1 (ATPase inhibitory factor 1) attenuated apoA-I internalization and increased oxidative stress in Ins-1E ß-cells and isolated mouse islets. Differentially expressed genes in apoA-I+ and apoA-I- Ins-1E cells were related to protein synthesis, the unfolded protein response, insulin secretion, and mitochondrial function. CONCLUSIONS: These results establish that ß-cells are functionally heterogeneous, and apoA-I restores insulin secretion in ß-cells with elevated cholesterol levels by improving mitochondrial redox balance.


Asunto(s)
Células Secretoras de Insulina , Insulina , Ratones , Animales , Insulina/farmacología , Apolipoproteína A-I/metabolismo , Células Secretoras de Insulina/metabolismo , Colesterol/metabolismo , Glucosa/metabolismo , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/farmacología
2.
Int J Mol Sci ; 25(8)2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38673843

RESUMEN

Neutrophil-myeloperoxidase (MPO) is a heme-containing peroxidase which produces excess amounts of hypochlorous acid during inflammation. While pharmacological MPO inhibition mitigates all indices of experimental colitis, no studies have corroborated the role of MPO using knockout (KO) models. Therefore, we investigated MPO deficient mice in a murine model of colitis. Wild type (Wt) and MPO-deficient mice were treated with dextran sodium sulphate (DSS) in a chronic model of experimental colitis with three acute cycles of DSS-induced colitis over 63 days, emulating IBD relapse and remission cycles. Mice were immunologically profiled at the gut muscoa and the faecal microbiome was assessed via 16S rRNA amplicon sequencing. Contrary to previous pharmacological antagonist studies targeting MPO, MPO-deficient mice showed no protection from experimental colitis during cyclical DSS-challenge. We are the first to report drastic faecal microbiota shifts in MPO-deficient mice, showing a significantly different microbiome profile on Day 1 of treatment, with a similar shift and distinction on Day 29 (half-way point), via qualitative and quantitative descriptions of phylogenetic distances. Herein, we provide the first evidence of substantial microbiome shifts in MPO-deficiency, which may influence disease progression. Our findings have significant implications for the utility of MPO-KO mice in investigating disease models.


Asunto(s)
Colitis , Sulfato de Dextran , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Ratones Noqueados , Peroxidasa , Animales , Peroxidasa/metabolismo , Peroxidasa/genética , Ratones , Colitis/microbiología , Colitis/inducido químicamente , Colitis/genética , Heces/microbiología , Eliminación de Gen , ARN Ribosómico 16S/genética , Ratones Endogámicos C57BL
3.
Chem Res Toxicol ; 34(6): 1681-1692, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-34085520

RESUMEN

The heme enzyme myeloperoxidase (MPO) is a key mediator of endothelial dysfunction and a therapeutic target in cardiovascular disease. During inflammation, MPO released by circulating leukocytes is internalized by endothelial cells and transcytosed into the subendothelial extracellular matrix of diseased vessels. At this site, MPO mediates endothelial dysfunction by catalytically consuming nitric oxide (NO) and producing reactive oxidants, hypochlorous acid (HOCl) and the nitrogen dioxide radical (•NO2). Accordingly, there is interest in developing MPO inhibitors that effectively target endothelial-localized MPO. Here we studied a series of piperidine nitroxides conjugated to polyamine moieties as novel endothelial-targeted MPO inhibitors. Electron paramagnetic resonance analysis of cell lysates showed that polyamine conjugated nitroxides were efficiently internalized into endothelial cells in a heparan sulfate dependent manner. Nitroxides effectively inhibited the consumption of MPO's substrate hydrogen peroxide (H2O2) and formation of HOCl catalyzed by endothelial-localized MPO, with their efficacy dependent on both nitroxide and conjugated-polyamine structure. Nitroxides also differentially inhibited protein nitration catalyzed by both purified and endothelial-localized MPO, which was dependent on •NO2 scavenging rather than MPO inhibition. Finally, nitroxides uniformly inhibited the catalytic consumption of NO by MPO in human plasma. These studies show for the first time that nitroxides effectively inhibit local oxidative reactions catalyzed by endothelial-localized MPO. Novel polyamine-conjugated nitroxides, ethylenediamine-TEMPO and putrescine-TEMPO, emerged as efficacious nitroxides uniquely exhibiting high endothelial cell uptake and efficient inhibition of MPO-catalyzed HOCl production, protein nitration, and NO oxidation. Polyamine-conjugated nitroxides represent a versatile class of antioxidant drugs capable of targeting endothelial-localized MPO during vascular inflammation.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Óxido Nítrico/farmacología , Peroxidasa/antagonistas & inhibidores , Poliaminas/farmacología , Biocatálisis , Células Endoteliales/enzimología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Humanos , Óxido Nítrico/química , Óxido Nítrico/metabolismo , Oxidación-Reducción , Peroxidasa/metabolismo , Poliaminas/química , Poliaminas/metabolismo
4.
Biochemistry ; 58(7): 974-986, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30585477

RESUMEN

The heme enzyme indoleamine 2,3-dioxygenase-1 (IDO1) catalyzes the first reaction of l-tryptophan oxidation along the kynurenine pathway. IDO1 is a central immunoregulatory enzyme with important implications for inflammation, infectious disease, autoimmune disorders, and cancer. Here we demonstrate that IDO1 is a mammalian nitrite reductase capable of chemically reducing nitrite to nitric oxide (NO) under hypoxia. Ultraviolet-visible absorption and resonance Raman spectroscopy showed that incubation of dithionite-reduced, ferrous-IDO1 protein (FeII-IDO1) with nitrite under anaerobic conditions resulted in the time-dependent formation of an FeII-nitrosyl IDO1 species, which was inhibited by substrate l-tryptophan, dependent on the concentration of nitrite or IDO1, and independent of the concentration of the reductant, dithionite. The bimolecular rate constant for IDO1 nitrite reductase activity was determined as 5.4 M-1 s-1 (pH 7.4, 23 °C), which was comparable to that measured for myoglobin (3.6 M-1 s-1; pH 7.4, 23 °C), an efficient and biologically important mammalian heme-based nitrite reductase. IDO1 nitrite reductase activity was pH-dependent but differed with myoglobin in that it showed a reduced proton dependency at pH >7. Electron paramagnetic resonance studies measuring NO production showed that the conventional IDO1 dioxygenase reducing cofactors, ascorbate and methylene blue, enhanced IDO1's nitrite reductase activity and the time- and IDO1 concentration-dependent release of NO in a manner inhibited by l-tryptophan or the IDO inhibitor 1-methyl-l-tryptophan. These data identify IDO1 as an efficient mammalian nitrite reductase that is capable of generating NO under anaerobic conditions. IDO1's nitrite reductase activity may have important implications for the enzyme's biological actions when expressed within hypoxic tissues.


Asunto(s)
Indolamina-Pirrol 2,3,-Dioxigenasa/química , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Nitrito Reductasas/metabolismo , Anaerobiosis , Espectroscopía de Resonancia por Spin del Electrón , Hemo/química , Hemo/metabolismo , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Óxido Nítrico/química , Óxido Nítrico/metabolismo , Nitrito Reductasas/química , Nitritos/química , Nitritos/metabolismo , Protones , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espectrofotometría Ultravioleta , Espectrometría Raman
5.
Proteomics ; 18(12): e1700253, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29437277

RESUMEN

The recognition of pathogen-derived peptides by T lymphocytes is the cornerstone of adaptive immunity, whereby intracellular antigens are degraded in the cytosol and short peptides assemble with class I human leukocyte antigen (HLA) molecules in the ER. These peptide-HLA complexes egress to the cell surface and are scrutinized by cytotoxic CD8+ T-cells leading to the eradication of the infected cell. Here, naturally presented HLA-B*57:01 bound peptides derived from the envelope protein of the human immunodeficiency virus (HIVenv) are identified. HIVenv peptides are present at a very small percentage of the overall HLA-B*57:01 peptidome (<0.1%) and both native and posttranslationally modified forms of two distinct HIV peptides are identified. Notably, a peptide bearing a natively encoded C-terminal tryptophan residue is also present in a modified form containing a kynurenine residue. Kynurenine is a major product of tryptophan catabolism and is abundant during inflammation and infection. Binding of these peptides at a molecular level and their immunogenicity in preliminary functional studies are examined. Modest immune responses are observed to the modified HIVenv peptide, highlighting a potential role for kynurenine-modified peptides in the immune response to HIV and other viral infections.


Asunto(s)
Linfocitos B/inmunología , Epítopos/inmunología , Productos del Gen env/inmunología , Antígenos VIH/inmunología , VIH-1/inmunología , Antígenos HLA-B/inmunología , Procesamiento Proteico-Postraduccional , Linfocitos B/virología , Células Cultivadas , Epítopos/metabolismo , Productos del Gen env/metabolismo , Antígenos VIH/metabolismo , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Antígenos HLA-B/química , Antígenos HLA-B/metabolismo , Humanos
6.
Clin Sci (Lond) ; 129(7): 601-72, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26186743

RESUMEN

IDO1 (indoleamine 2,3-dioxygenase 1) is a member of a unique class of mammalian haem dioxygenases that catalyse the oxidative catabolism of the least-abundant essential amino acid, L-Trp (L-tryptophan), along the kynurenine pathway. Significant increases in knowledge have been recently gained with respect to understanding the fundamental biochemistry of IDO1 including its catalytic reaction mechanism, the scope of enzyme reactions it catalyses, the biochemical mechanisms controlling IDO1 expression and enzyme activity, and the discovery of enzyme inhibitors. Major advances in understanding the roles of IDO1 in physiology and disease have also been realised. IDO1 is recognised as a prominent immune regulatory enzyme capable of modulating immune cell activation status and phenotype via several molecular mechanisms including enzyme-dependent deprivation of L-Trp and its conversion into the aryl hydrocarbon receptor ligand kynurenine and other bioactive kynurenine pathway metabolites, or non-enzymatic cell signalling actions involving tyrosine phosphorylation of IDO1. Through these different modes of biochemical signalling, IDO1 regulates certain physiological functions (e.g. pregnancy) and modulates the pathogenesis and severity of diverse conditions including chronic inflammation, infectious disease, allergic and autoimmune disorders, transplantation, neuropathology and cancer. In the present review, we detail the current understanding of IDO1's catalytic actions and the biochemical mechanisms regulating IDO1 expression and activity. We also discuss the biological functions of IDO1 with a focus on the enzyme's immune-modulatory function, its medical implications in diverse pathological settings and its utility as a therapeutic target.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Indolamina-Pirrol 2,3,-Dioxigenasa/fisiología , Animales , Infecciones Bacterianas/metabolismo , Catálisis , Humanos , Quinurenina/química , Ratones , Modelos Biológicos , Neoplasias/embriología , Enfermedades del Sistema Nervioso/metabolismo , Oxidación-Reducción , Conformación Proteica , Procesamiento Proteico-Postraduccional , Transducción de Señal , Especificidad por Sustrato , Triptófano/química
7.
Arterioscler Thromb Vasc Biol ; 34(10): 2261-7, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25147338

RESUMEN

OBJECTIVE: Therapeutic interventions that increase plasma levels of high-density lipoproteins and apolipoprotein A-I (apoA-I) A-I, the major high-density lipoprotein apolipoprotein, improve glycemic control in people with type 2 diabetes mellitus. High-density lipoproteins and apoA-I also enhance insulin synthesis and secretion in isolated pancreatic islets and clonal ß-cell lines. This study identifies the signaling pathways that mediate these effects. APPROACH AND RESULTS: Incubation with apoA-I increased cAMP accumulation in Ins-1E cells in a concentration-dependent manner. The increase in cAMP levels was inhibited by preincubating the cells with the cell-permeable, transmembrane adenylate cyclase inhibitor, 2'5' dideoxyadenosine, but not with KH7, which inhibits soluble adenylyl cyclases. Incubation of Ins-1E cells with apoA-I resulted in colocalization of ATP-binding cassette transporter A1 with the Gαs subunit of a heterotrimeric G-protein and a Gαs subunit-dependent increase in insulin secretion. Incubation of Ins-1E cells with apoA-I also increased protein kinase A phosphorylation and reduced the nuclear localization of forkhead box protein O1 (FoxO1). Preincubation of Ins-1E cells with the protein kinase A-specific inhibitors, H89 and PKI amide, prevented apoA-I from increasing insulin secretion and mediating the nuclear exclusion of FoxO1. Transfection of Ins-1E cells with a mutated FoxO1 that is restricted to the nucleus confirmed the requirement for FoxO1 nuclear exclusion by blocking insulin secretion in apoA-I-treated Ins-1E cells. ApoA-I also increased Irs1, Irs2, Ins1, Ins2, and Pdx1 mRNA levels. CONCLUSIONS: ApoA-I increases insulin synthesis and secretion via a heterotrimeric G-protein-cAMP-protein kinase A-FoxO1-dependent mechanism that involves transmembrane adenylyl cyclases and increased transcription of key insulin response and ß-cell survival genes.


Asunto(s)
Apolipoproteína A-I/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Factores de Transcripción Forkhead/metabolismo , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Células Secretoras de Insulina/enzimología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Inhibidores de Adenilato Ciclasa , Adenilil Ciclasas/metabolismo , Animales , Línea Celular Tumoral , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Insulina/biosíntesis , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Ratas , Receptores Depuradores de Clase B/genética , Receptores Depuradores de Clase B/metabolismo , Transducción de Señal , Factores de Tiempo , Transfección , Regulación hacia Arriba
8.
J Biol Chem ; 288(3): 1548-67, 2013 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-23209301

RESUMEN

The heme enzyme indoleamine 2,3-dioxygenase (IDO) is a key regulator of immune responses through catalyzing l-tryptophan (l-Trp) oxidation. Here, we show that hydrogen peroxide (H(2)O(2)) activates the peroxidase function of IDO to induce protein oxidation and inhibit dioxygenase activity. Exposure of IDO-expressing cells or recombinant human IDO (rIDO) to H(2)O(2) inhibited dioxygenase activity in a manner abrogated by l-Trp. Dioxygenase inhibition correlated with IDO-catalyzed H(2)O(2) consumption, compound I-mediated formation of protein-centered radicals, altered protein secondary structure, and opening of the distal heme pocket to promote nonproductive substrate binding; these changes were inhibited by l-Trp, the heme ligand cyanide, or free radical scavengers. Protection by l-Trp coincided with its oxidation into oxindolylalanine and kynurenine and the formation of a compound II-type ferryl-oxo heme. Physiological peroxidase substrates, ascorbate or tyrosine, enhanced rIDO-mediated H(2)O(2) consumption and attenuated H(2)O(2)-induced protein oxidation and dioxygenase inhibition. In the presence of H(2)O(2), rIDO catalytically consumed nitric oxide (NO) and utilized nitrite to promote 3-nitrotyrosine formation on IDO. The promotion of H(2)O(2) consumption by peroxidase substrates, NO consumption, and IDO nitration was inhibited by l-Trp. This study identifies IDO as a heme peroxidase that, in the absence of substrates, self-inactivates dioxygenase activity via compound I-initiated protein oxidation. l-Trp protects against dioxygenase inactivation by reacting with compound I and retarding compound II reduction to suppress peroxidase turnover. Peroxidase-mediated dioxygenase inactivation, NO consumption, or protein nitration may modulate the biological actions of IDO expressed in inflammatory tissues where the levels of H(2)O(2) and NO are elevated and l-Trp is low.


Asunto(s)
Hemo/química , Peróxido de Hidrógeno/química , Indolamina-Pirrol 2,3,-Dioxigenasa/química , Peroxidasas/química , Biocatálisis , Dicroismo Circular , Escherichia coli/genética , Hemo/metabolismo , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Cinética , Óxido Nítrico/química , Oxidación-Reducción , Peroxidasas/metabolismo , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Soluciones , Espectrometría Raman
9.
Heart Lung Circ ; 23(12): 1110-7, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25047283

RESUMEN

BACKGROUND: The Medtronic Freestyle bioprosthesis (FSB) provides an alternative to other prostheses for both aortic valve and aortic root surgery. This paper is a systematic review of the post-operative outcomes in patients with aortic valve and/or aortic root disease following FSB implantation. METHODS: Electronic databases were searched for primary analysis, prospective randomised studies comparing the FSB with an alternative aortic prosthesis were included. Additionally, case series that included data for at least 100 individual operated patients were used for secondary analysis. RESULTS: Among three identified randomised studies, 199 FSB cases were compared with homografts, and stented and an alternative stentless bioprosthesis. The FSB showed comparable hospital mortality (4.5% vs. 5.3%) and eight-year actuarial survival (80±5.0% versus 77±6.0%) with the homograft (respectively) and comparable reduction in left ventricular mass index relative to other prosthesis types. Over 6000 individual patients were included in the selected 15 case series. Weighted mean operative mortality, neurological event rate and five-year actuarial survival was 5.2%, 5.5% and 77.8%, respectively. CONCLUSION: The FSB performed comparably against alternative prostheses regarding in-hospital mortality, long-term survival and reduction in left ventricular mass index. Included case series demonstrated robust post-operative outcomes in both the short and long term.


Asunto(s)
Aorta , Bioprótesis , Prótesis Vascular , Cardiopatías Congénitas , Enfermedades de las Válvulas Cardíacas , Prótesis Valvulares Cardíacas , Animales , Válvula Aórtica/fisiopatología , Válvula Aórtica/cirugía , Enfermedad de la Válvula Aórtica Bicúspide , Cardiopatías Congénitas/mortalidad , Cardiopatías Congénitas/fisiopatología , Cardiopatías Congénitas/cirugía , Enfermedades de las Válvulas Cardíacas/mortalidad , Enfermedades de las Válvulas Cardíacas/fisiopatología , Enfermedades de las Válvulas Cardíacas/cirugía , Humanos
10.
Atherosclerosis ; 392: 117519, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38581737

RESUMEN

BACKGROUND AND AIMS: Atherosclerosis is the primary underlying cause of myocardial infarction and stroke, which are the major causes of death globally. Heparanase (Hpse) is a pro-inflammatory extracellular matrix degrading enzyme that has been implicated in atherogenesis. However, to date the precise roles of Hpse in atherosclerosis and its mechanisms of action are not well defined. This study aims to provide new insights into the contribution of Hpse in different stages of atherosclerosis in vivo. METHODS: We generated Hpse gene-deficient mice on the atherosclerosis-prone apolipoprotein E gene knockout (ApoE-/-) background to investigate the impact of Hpse gene deficiency on the initiation and progression of atherosclerosis after 6 and 14 weeks high-fat diet feeding, respectively. Atherosclerotic lesion development, blood serum profiles, lesion composition and aortic immune cell populations were evaluated. RESULTS: Hpse-deficient mice exhibited significantly reduced atherosclerotic lesion burden in the aortic sinus and aorta at both time-points, independent of changes in plasma cholesterol levels. A significant reduction in the necrotic core size and an increase in smooth muscle cell content were also observed in advanced atherosclerotic plaques of Hpse-deficient mice. Additionally, Hpse deficiency reduced circulating and aortic levels of VCAM-1 at the initiation and progression stages of disease and circulating MCP-1 levels in the initiation but not progression stage. Moreover, the aortic levels of total leukocytes and dendritic cells in Hpse-deficient ApoE-/- mice were significantly decreased compared to control ApoE-/-mice at both disease stages. CONCLUSIONS: This study identifies Hpse as a key pro-inflammatory enzyme driving the initiation and progression of atherosclerosis and highlighting the potential of Hpse inhibitors as novel anti-inflammatory treatments for cardiovascular disease.


Asunto(s)
Aorta , Aterosclerosis , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Glucuronidasa , Ratones Noqueados para ApoE , Placa Aterosclerótica , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/enzimología , Aterosclerosis/metabolismo , Glucuronidasa/deficiencia , Glucuronidasa/genética , Glucuronidasa/metabolismo , Aorta/patología , Aorta/metabolismo , Aorta/enzimología , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/enzimología , Enfermedades de la Aorta/metabolismo , Dieta Alta en Grasa , Apolipoproteínas E/genética , Apolipoproteínas E/deficiencia , Ratones Endogámicos C57BL , Masculino , Molécula 1 de Adhesión Celular Vascular/metabolismo , Ratones , Ratones Noqueados , Seno Aórtico/patología , Necrosis
11.
Best Pract Res Clin Endocrinol Metab ; 37(3): 101689, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36008277

RESUMEN

The high density lipoprotein (HDL) fraction of human plasma consists of multiple subpopulations of spherical particles that are structurally uniform, but heterogeneous in terms of size, composition and function. Numerous epidemiological studies have established that an elevated high density lipoprotein cholesterol (HDL-C) level is associated with decreased cardiovascular risk. However, with several recent randomised clinical trials of HDL-C raising agents failing to reduce cardiovascular events, contemporary research is transitioning towards clinical development of the cardioprotective functions of HDLs and the identification of functions that can be exploited for treatment of other diseases. This review describes the origins of HDLs and the causes of their compositional and functional heterogeneity. It then summarises current knowledge of how cardioprotective and other functions of HDLs are regulated. The final section of the review summarises recent advances in the clinical development of HDL-targeted therapies.


Asunto(s)
Apolipoproteína A-I , Lipoproteínas HDL , Humanos
12.
J Am Heart Assoc ; 12(4): e027600, 2023 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-36789837

RESUMEN

Excessive inflammation and impaired healing of cardiac tissue following a myocardial infarction (MI) can drive the development of heart failure. Cardiac repair begins immediately after the onset of MI and continues for months. The repair process can be divided into the following 3 overlapping phases, each having distinct functions and sequelae: the inflammatory phase, the proliferative phase, and the maturation phase. Macrophages, neutrophils, and lymphocytes are present in the myocardium throughout the repair process and govern the duration and function of each of these phases. However, changes in the functions of these cell types across each phase are poorly characterized. Numerous immunomodulatory therapies that specifically target inflammation have been developed for promoting cardiac repair and preventing heart failure after MI. However, these treatments have been largely unsuccessful in large-scale clinical randomized controlled trials. A potential explanation for this failure is the lack of a thorough understanding of the time-dependent evolution of the functions of immune cells after a major cardiovascular event. Failure to account for this temporal plasticity in cell function may reduce the efficacy of immunomodulatory approaches that target cardiac repair. This review is concerned with how the functions of different immune cells change with time following an MI. Improved understanding of the temporal changes in immune cell function is important for the future development of effective and targeted treatments for preventing heart failure after MI.


Asunto(s)
Insuficiencia Cardíaca , Infarto del Miocardio , Humanos , Miocardio/metabolismo , Insuficiencia Cardíaca/terapia , Insuficiencia Cardíaca/metabolismo , Reperfusión , Inflamación/metabolismo , Remodelación Ventricular/fisiología
13.
J Biol Chem ; 286(15): 12901-11, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21266577

RESUMEN

Urate and myeloperoxidase (MPO) are associated with adverse outcomes in cardiovascular disease. In this study, we assessed whether urate is a likely physiological substrate for MPO and if the products of their interaction have the potential to exacerbate inflammation. Urate was readily oxidized by MPO and hydrogen peroxide to 5-hydroxyisourate, which decayed to predominantly allantoin. The redox intermediates of MPO were reduced by urate with rate constants of 4.6 × 10(5) M(-1) s(-1) for compound I and 1.7 × 10(4) M(-1) s(-1) for compound II. Urate competed with chloride for oxidation by MPO and at hyperuricemic levels is expected to be a substantive substrate for the enzyme. Oxidation of urate promoted super-stoichiometric consumption of glutathione, which indicates that it is converted to a free radical intermediate. In combination with superoxide and hydrogen peroxide, MPO oxidized urate to a reactive hydroperoxide. This would form by addition of superoxide to the urate radical. Urate also enhanced MPO-dependent consumption of nitric oxide. In human plasma, stimulated neutrophils produced allantoin in a reaction dependent on the NADPH oxidase, MPO and superoxide. We propose that urate is a physiological substrate for MPO that is oxidized to the urate radical. The reactions of this radical with superoxide and nitric oxide provide a plausible link between urate and MPO in cardiovascular disease.


Asunto(s)
Peróxido de Hidrógeno/metabolismo , Hiperuricemia/enzimología , Neutrófilos/enzimología , Peroxidasa/metabolismo , Superóxidos/metabolismo , Alantoína/biosíntesis , Alantoína/química , Enfermedades Cardiovasculares/enzimología , Humanos , Peróxido de Hidrógeno/química , Inflamación , NADPH Oxidasas/química , NADPH Oxidasas/metabolismo , Oxidación-Reducción , Peroxidasa/química , Especificidad por Sustrato , Superóxidos/química , Ácido Úrico
14.
Cells ; 11(20)2022 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-36291066

RESUMEN

Cardiovascular disease (CVD) is the leading cause of death and disability worldwide, and its management places a huge burden on healthcare systems through hospitalisation and treatment. Atherosclerosis is a chronic inflammatory disease of the arterial wall resulting in the formation of lipid-rich, fibrotic plaques under the subendothelium and is a key contributor to the development of CVD. As such, a detailed understanding of the mechanisms involved in the development of atherosclerosis is urgently required for more effective disease treatment and prevention strategies. Heparanase is the only mammalian enzyme known to cleave heparan sulfate of heparan sulfate proteoglycans, which is a key component of the extracellular matrix and basement membrane. By cleaving heparan sulfate, heparanase contributes to the regulation of numerous physiological and pathological processes such as wound healing, inflammation, tumour angiogenesis, and cell migration. Recent evidence suggests a multifactorial role for heparanase in atherosclerosis by promoting underlying inflammatory processes giving rise to plaque formation, as well as regulating lesion stability. This review provides an up-to-date overview of the role of heparanase in physiological and pathological processes with a focus on the emerging role of the enzyme in atherosclerosis.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Animales , Humanos , Proteoglicanos de Heparán Sulfato , Glucuronidasa , Heparitina Sulfato , Aterosclerosis/terapia , Placa Aterosclerótica/terapia , Lípidos , Mamíferos
15.
Pharmacol Ther ; 221: 107711, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33137376

RESUMEN

Myeloperoxidase (MPO) is a prominent mammalian heme peroxidase and a fundamental component of the innate immune response against microbial pathogens. In recent times, MPO has received considerable attention as a key oxidative enzyme capable of impairing the bioactivity of nitric oxide (NO) and promoting endothelial dysfunction; a clinically relevant event that manifests throughout the development of inflammatory cardiovascular disease. Increasing evidence indicates that during cardiovascular disease, MPO is released intravascularly by activated leukocytes resulting in its transport and sequestration within the vascular endothelium. At this site, MPO catalyzes various oxidative reactions that are capable of promoting vascular inflammation and impairing NO bioactivity and endothelial function. In particular, MPO catalyzes the production of the potent oxidant hypochlorous acid (HOCl) and the catalytic consumption of NO via the enzyme's NO oxidase activity. An emerging paradigm is the ability of MPO to also influence endothelial function via non-catalytic, cytokine-like activities. In this review article we discuss the implications of our increasing knowledge of the versatility of MPO's actions as a mediator of cardiovascular disease and endothelial dysfunction for the development of new pharmacological agents capable of effectively combating MPO's pathogenic activities. More specifically, we will (i) discuss the various transport mechanisms by which MPO accumulates into the endothelium of inflamed or diseased arteries, (ii) detail the clinical and basic scientific evidence identifying MPO as a significant cause of endothelial dysfunction and cardiovascular disease, (iii) provide an up-to-date coverage on the different oxidative mechanisms by which MPO can impair endothelial function during cardiovascular disease including an evaluation of the contributions of MPO-catalyzed HOCl production and NO oxidation, and (iv) outline the novel non-enzymatic mechanisms of MPO and their potential contribution to endothelial dysfunction. Finally, we deliver a detailed appraisal of the different pharmacological strategies available for targeting the catalytic and non-catalytic modes-of-action of MPO in order to protect against endothelial dysfunction in cardiovascular disease.


Asunto(s)
Enfermedades Cardiovasculares , Peroxidasa , Enfermedades Vasculares , Animales , Enfermedades Cardiovasculares/tratamiento farmacológico , Endotelio Vascular/metabolismo , Humanos , Ácido Hipocloroso/metabolismo , Óxido Nítrico/metabolismo , Oxidación-Reducción , Peroxidasa/metabolismo , Peroxidasa/farmacología , Enfermedades Vasculares/metabolismo
16.
Free Radic Biol Med ; 166: 255-264, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33539947

RESUMEN

During vascular inflammation, the leukocyte-derived enzyme myeloperoxidase (MPO) is transcytosed across the endothelium and into the sub-endothelial extracellular matrix, where it promotes endothelial dysfunction by catalytically consuming nitric oxide (NO) produced by endothelial NO synthase (eNOS). In the presence of chloride ions and hydrogen peroxide (H2O2), MPO forms the oxidant hypochlorous acid (HOCl). Here we examined the short-term implications of HOCl produced by endothelial-transcytosed MPO for eNOS activity. Incubation of MPO with cultured aortic endothelial cells (ECs) resulted in its transport into the sub-endothelium. Exposure of MPO-containing ECs to low micromolar concentrations of H2O2 yielded enhanced rates of H2O2 consumption that correlated with HOCl formation and increased eNOS enzyme activity. The MPO-dependent activation of eNOS occurred despite reduced cellular uptake of the eNOS substrate l-arginine, which involved a decrease in the maximal activity (Vmax), but not substrate affinity (Km), of the major endothelial l-arginine transporter, cationic amino acid transporter-1. Activation of eNOS in MPO-containing ECs exposed to H2O2 involved a rapid elevation in cytosolic calcium and increased eNOS phosphorylation at Ser-1179 and de-phosphorylation at Thr-497. These signaling events were attenuated by intracellular calcium chelation, removal of extracellular calcium and inhibition of phospholipase C. This study shows that stimulation of endothelial-transcytosed MPO activates eNOS by promoting phospholipase C-dependent calcium signaling and altered eNOS phosphorylation at Ser-1179 and Thr-497. This may constitute a compensatory signaling response of ECs aimed at maintaining eNOS activity and NO production in the face of MPO-catalyzed oxidative stress.


Asunto(s)
Óxido Nítrico Sintasa de Tipo III , Peroxidasa , Calcio/metabolismo , Señalización del Calcio , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Peróxido de Hidrógeno/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Peroxidasa/metabolismo , Fosfolipasas de Tipo C/metabolismo
17.
Sci Rep ; 11(1): 14993, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34294791

RESUMEN

The treatment of periodontitis has numerous positive effects on established chronic health conditions, including cardiovascular disease and diabetes. However, ethical considerations do limit the establishment of human trials to investigate whether periodontitis promotes the early stages of chronic conditions. Therefore, the aim of this study was to investigate whether periodontitis induces endothelial dysfunction in hyperlipidemic apolipoprotein E gene-deficient (ApoE-/-) mice. Forty-five 8-week-old ApoE-/- mice were challenged by oral lavage with Porphyromonas gingivalis and Streptococcus gordonii for 4 weeks. A subgroup of animals (n = 15-17/group) was placed in a metabolic chamber immediately before euthanasia at 4 weeks to measure VO2/CO2 concentrations and voluntary locomotion. In infected and control animals alveolar bone levels were measured by x-ray imaging and endothelial function was determined by measuring endothelial-dependent vasorelaxation of aortic rings. The mRNA expression levels of serum amyloid A and tumor necrosis factor were determined in liver tissues by qRT PCR and protein concentrations in serum by ELISA. Caecal contents were analysed by sequencing to determine changes to the gut microbiota to investigate linkages between microbiome and systemic changes. The results showed that oral lavage of P. gingivalis and S. gordonii for 4 weeks, initiated periodontitis in ApoE-/- mice, similar to the human situation. The oral inflammation was accompanied by a significant increase in mRNA expression of pro-inflammatory mediators serum amyloid A1 and tumor necrosis factor in the liver. Mice with periodontitis also exhibited impaired endothelial-dependent vasorelaxation responses to acetylcholine. This systemic response was connected to increased energy expenditure, locomotion and respiratory quotient. No differences were detected in caecal microbiota between the infected and control animals. Overall, this is the first report that provide evidence that periodontitis induces endothelial dysfunction in mice. Other systemic responses observed in response to the local reaction need further investigation. The study suggests that early prevention of periodontitis may help limit the early stages of endothelial dysfunction that is linked to atherogenesis in humans.


Asunto(s)
Apolipoproteínas E/genética , Infecciones por Bacteroidaceae/diagnóstico por imagen , Hiperlipidemias/genética , Periodontitis/microbiología , Placa Aterosclerótica/diagnóstico por imagen , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Modelos Animales de Enfermedad , Metabolismo Energético , Microbioma Gastrointestinal , Técnicas de Inactivación de Genes , Hiperlipidemias/microbiología , Masculino , Ratones , Periodontitis/diagnóstico por imagen , Periodontitis/genética , Filogenia , Placa Aterosclerótica/microbiología , Porphyromonas gingivalis/patogenicidad , Análisis de Secuencia de ARN , Proteína Amiloide A Sérica/genética , Proteína Amiloide A Sérica/metabolismo , Streptococcus gordonii/patogenicidad , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Rayos X
18.
Biochemistry ; 49(3): 591-600, 2010 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-20000778

RESUMEN

The heme enzyme indoleamine 2,3-dioxygenase (IDO) plays an important immune regulatory role by catalyzing the oxidative degradation of l-tryptophan. Here we show that the selenezal drug ebselen is a potent IDO inhibitor. Exposure of human macrophages to ebselen inhibited IDO activity in a manner independent of changes in protein expression. Ebselen inhibited the activity of recombinant human IDO (rIDO) with an apparent inhibition constant of 94 +/- 17 nM. Optical and resonance Raman spectroscopy showed that ebselen altered the active site heme of rIDO by inducing a transition of the ferric heme iron from the predominantly high- to low-spin form and by lowering the vibrational frequency of the Fe-CO stretch of the CO complex, indicating an opening of the distal heme pocket. Substrate binding studies showed that ebselen enhanced nonproductive l-tryptophan binding, while circular dichroism indicated that the drug reduced the helical content and protein stability of rIDO. Thiol labeling and mass spectrometry revealed that ebselen reacted with multiple cysteine residues of IDO. Removal of cysteine-bound ebselen with dithiothreitol reversed the effects of the drug on the heme environment and significantly restored enzyme activity. These findings indicate that ebselen inhibits IDO activity by reacting with the enzyme's cysteine residues that result in changes to protein conformation and active site heme, leading to an increase in the level of nonproductive substrate binding. This study highlights that modification of cysteine residues is a novel and effective means of inhibiting IDO activity. It also suggests that IDO is under redox control and that the enzyme represents a previously unrecognized in vivo target of ebselen.


Asunto(s)
Azoles/química , Azoles/farmacología , Cisteína/química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Indolamina-Pirrol 2,3,-Dioxigenasa/química , Compuestos de Organoselenio/química , Compuestos de Organoselenio/farmacología , Sitios de Unión , Catálisis , Dicroismo Circular , Cisteína/genética , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Isoindoles , Cinética , Conformación Proteica , Espectrometría Raman
19.
Antioxidants (Basel) ; 9(12)2020 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-33266084

RESUMEN

Systemic glutathione deficiency, inflammation, and oxidative stress are hallmarks of cystic fibrosis (CF), an inherited disease that causes persistent lung infections and severe damage to the respiratory system and many of the body organs. Improvements to current antioxidant therapeutic strategies are needed. The dietary supplement, γ-glutamylcysteine (GGC), which is the immediate precursor to glutathione, rapidly boosts cellular glutathione levels following a single dose in healthy individuals. Efficacy of GGC against oxidative stress induced by Pseudomonas aeruginosa, which is a common and chronic pathogen infecting lungs of CF patients, remains unassessed. Primary mucocilliary differentiated airway (bronchial and/or nasal) epithelial cells were created from four individuals with CF. Airway oxidative stress and inflammation was induced by P. aeruginosa lipopolysaccharide (LPS). Parameters including global proteomics alterations, cell redox state (glutathione, oxidative stress), pro-inflammatory mediators (IL-8, IDO-1), and cellular health (membrane integrity, stress granule formation, cell metabolic viability) were assayed under six experimental conditions: (1) Mock, (2) LPS-challenged (3) therapeutic, (4) prophylactic (5) therapeutic and prophylactic and (6) GGC alone. Proteomic analysis identified perturbation of several pathways related to cellular respiration and stress responses upon LPS challenge. Most of these were resolved when cells were treated with GGC. While GGC did not resolve LPS-induced IL-8 and IDO-1 activity, it effectively attenuated LPS-induced oxidative stress and stress granule formation, while significantly increasing total intracellular glutathione levels, metabolic viability and improving epithelial cell barrier integrity. Both therapeutic and prophylactic treatments were successful. Together, these findings indicate that GGC has therapeutic potential for treatment and prevention of oxidative stress-related damage to airways in cystic fibrosis.

20.
Circulation ; 118(13): 1347-57, 2008 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-18809807

RESUMEN

BACKGROUND: Oxidative injury and dysfunction of the vascular endothelium are early and causal features of many vascular diseases. Single antioxidant strategies to prevent vascular injury have met with mixed results. METHODS AND RESULTS: Here, we report that induction of a metabolic stress response with adenosine monophosphate kinase (AMPK) prevents oxidative endothelial cell injury. This response is characterized by stabilization of the mitochondrion and increased mitochondrial biogenesis, resulting in attenuation of oxidative c-Jun N-terminal kinase (JNK) activation. We report that peroxisome proliferator coactivator 1alpha is a key downstream target of AMPK that is both necessary and sufficient for the metabolic stress response and JNK attenuation. Moreover, induction of the metabolic stress response in vivo attenuates reactive oxygen species-mediated JNK activation and endothelial dysfunction in response to angiotensin II in wild-type mice but not in animals lacking either the endothelial isoform of AMPK or peroxisome proliferator coactivator 1alpha. CONCLUSIONS: These data highlight AMPK and peroxisome proliferator coactivator 1alpha as potential therapeutic targets for the amelioration of endothelial dysfunction and, as a consequence, vascular disease.


Asunto(s)
Células Endoteliales/enzimología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Estrés Oxidativo/fisiología , Enfermedades Vasculares/metabolismo , Adaptación Fisiológica/fisiología , Adenilato Quinasa/genética , Adenilato Quinasa/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Animales , Células COS , Muerte Celular/fisiología , Chlorocebus aethiops , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Proteínas de Choque Térmico/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Ratones Endogámicos C57BL , Mutagénesis , Oxidantes/farmacología , Estrés Oxidativo/efectos de los fármacos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , ARN Interferente Pequeño , Ribonucleótidos , Factores de Transcripción/metabolismo , Venas Umbilicales/citología , Enfermedades Vasculares/patología , Enfermedades Vasculares/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA