Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
Am J Physiol Regul Integr Comp Physiol ; 326(6): R552-R566, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38586887

RESUMEN

Nitric oxide is produced at low micromolar levels following the induction of inducible nitric oxide synthase (iNOS) and is responsible for mediating the inhibitory actions of cytokines on glucose-stimulated insulin secretion by islets of Langerhans. It is through the inhibition of mitochondrial oxidative metabolism, specifically aconitase and complex 4 of the electron transport chain, that nitric oxide inhibits insulin secretion. Nitric oxide also attenuates protein synthesis, induces DNA damage, activates DNA repair pathways, and stimulates stress responses (unfolded protein and heat shock) in ß-cells. In this report, the time- and concentration-dependent effects of nitric oxide on the expression of six genes known to participate in the response of ß-cells to this free radical were examined. The genes included Gadd45α (DNA repair), Puma (apoptosis), Hmox1 (antioxidant defense), Hsp70 (heat shock), Chop (UPR), and Ppargc1α (mitochondrial biogenesis). We show that nitric oxide stimulates ß-cell gene expression in a narrow concentration range of ∼0.5-1 µM or levels corresponding to iNOS-derived nitric oxide. At concentrations greater than 1 µM, nitric oxide fails to stimulate gene expression in ß-cells, and this is associated with the inhibition of mitochondrial oxidative metabolism. This narrow concentration range of responses is ß-cell selective, as the actions of nitric oxide in non-ß-cells (α-cells, mouse embryonic fibroblasts, and macrophages) are concentration dependent. Our findings suggest that ß-cells respond to a narrow concentration range of nitric oxide that is consistent with the levels produced following iNOS induction, and that these concentration-dependent actions are selective for insulin-containing cells.


Asunto(s)
Proteínas Reguladoras de la Apoptosis , Regulación de la Expresión Génica , Células Secretoras de Insulina , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico , Animales , Óxido Nítrico/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Factor de Transcripción CHOP/metabolismo , Factor de Transcripción CHOP/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo Oxigenasa (Desciclizante)/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Insulina/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/genética , Ratas , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Proteínas de la Membrana , Hemo-Oxigenasa 1
2.
Int J Mol Sci ; 22(3)2021 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-33513958

RESUMEN

Microparticles or microvesicles (MPs/MVs) are sub-cellular vesicles with a growing number of known biological functions. Microvesicles from a variety of parent cells within the vascular system increase in numerous pathological states. Red blood cell-derived MVs (RMVs) are relatively less studied than other types of circulating MVs despite red blood cells (RBCs) being the most abundant intravascular cell. This may be in part due the echoes of past misconceptions that RBCs were merely floating anucleate bags of hemoglobin rather than dynamic and responsive cells. The initial aim of this study was to maximize the concentration of RMVs derived from various blood or blood products by focusing on the optimal isolation conditions without creating more MVs from artificial manipulation. We found that allowing RBCs to sediment overnight resulted in a continuum in size of RBC membrane-containing fragments or vesicles extending beyond the 1 µm size limit suggested by many as the maximal size of an MV. Additionally, dilution and centrifugation factors were studied that altered the resultant MV population concentration. The heterogeneous size of RMVs was confirmed in mice models of hemolytic anemia. This methodological finding establishes a new paradigm in that it blurs the line between RBC, fragment, and RMV as well as suggests that the concentration of circulating RMVs may be widely underestimated given that centrifugation removes the majority of such RBC-derived membrane-containing particles.


Asunto(s)
Anemia Hemolítica/sangre , Micropartículas Derivadas de Células/genética , Centrifugación , Eritrocitos/citología , Anemia Hemolítica/genética , Anemia Hemolítica/patología , Animales , Linaje de la Célula/genética , Recuento de Eritrocitos , Hemoglobinas/genética , Humanos , Ratones
3.
Am J Physiol Regul Integr Comp Physiol ; 318(5): R1004-R1013, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32292063

RESUMEN

Both reactive nitrogen and oxygen species (RNS and ROS), such as nitric oxide, peroxynitrite, and hydrogen peroxide, have been implicated as mediators of pancreatic ß-cell damage during the pathogenesis of autoimmune diabetes. While ß-cells are thought to be vulnerable to oxidative damage due to reportedly low levels of antioxidant enzymes, such as catalase and glutathione peroxidase, we have shown that they use thioredoxin reductase to detoxify hydrogen peroxide. Thioredoxin reductase is an enzyme that participates in the peroxiredoxin antioxidant cycle. Peroxiredoxins are expressed in ß-cells and, when overexpressed, protect against oxidative stress, but the endogenous roles of peroxiredoxins in the protection of ß-cells from oxidative damage are unclear. Here, using either glucose oxidase or menadione to continuously deliver hydrogen peroxide, or the combination of dipropylenetriamine NONOate and menadione to continuously deliver peroxynitrite, we tested the hypothesis that ß-cells use peroxiredoxins to detoxify both of these reactive species. Either pharmacological peroxiredoxin inhibition with conoidin A or specific depletion of cytoplasmic peroxiredoxin 1 (Prdx1) using siRNAs sensitizes INS 832/13 cells and rat islets to DNA damage and death induced by hydrogen peroxide or peroxynitrite. Interestingly, depletion of peroxiredoxin 2 (Prdx2) had no effect. Together, these results suggest that ß-cells use cytoplasmic Prdx1 as a primary defense mechanism against both ROS and RNS.


Asunto(s)
Daño del ADN , Peróxido de Hidrógeno/toxicidad , Células Secretoras de Insulina/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Peroxirredoxinas/metabolismo , Ácido Peroxinitroso/toxicidad , Animales , Muerte Celular , Línea Celular Tumoral , Citoplasma/enzimología , Citoprotección , Inhibidores Enzimáticos/farmacología , Células Secretoras de Insulina/enzimología , Células Secretoras de Insulina/patología , Masculino , Peroxirredoxinas/antagonistas & inhibidores , Peroxirredoxinas/genética , Quinoxalinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas Sprague-Dawley , Transducción de Señal , Tiorredoxina Reductasa 1/metabolismo
4.
Nitric Oxide ; 104-105: 36-43, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32891753

RESUMEN

It is well established that myoglobin supports mitochondrial respiration through the storage and transport of oxygen as well as through the scavenging of nitric oxide. However, during ischemia/reperfusion (I/R), myoglobin and mitochondria both propagate myocardial injury through the production of oxidants. Nitrite, an endogenous signaling molecule and dietary constituent, mediates potent cardioprotection after I/R and this effect relies on its interaction with both myoglobin and mitochondria. While independent mechanistic studies have demonstrated that nitrite-mediated cardioprotection requires the presence of myoglobin and the post-translational S-nitrosation of critical cysteine residues on mitochondrial complex I, it is unclear whether myoglobin directly catalyzes the S-nitrosation of complex I or whether mitochondrial-dependent nitrite reductase activity contributes to S-nitrosation. Herein, using purified myoglobin and isolated mitochondria, we characterize and directly compare the nitrite reductase activities of mitochondria and myoglobin and assess their contribution to mitochondrial S-nitrosation. We demonstrate that myoglobin is a significantly more efficient nitrite reductase than isolated mitochondria. Further, deoxygenated myoglobin catalyzes the nitrite-dependent S-nitrosation of mitochondrial proteins. This reaction is enhanced in the presence of oxidized (Fe3+) myoglobin and not significantly affected by inhibitors of mitochondrial respiration. Using a Chinese Hamster Ovary cell model stably transfected with human myoglobin, we show that both myoglobin and mitochondrial complex I expression are required for nitrite-dependent attenuation of cell death after anoxia/reoxygenation. These data expand the understanding of myoglobin's role both as a nitrite reductase to a mediator of S-nitrosation and as a regulator of mitochondrial function, and have implications for nitrite-mediated cardioprotection after I/R.


Asunto(s)
Citoprotección/fisiología , Mitocondrias/metabolismo , Mioglobina/metabolismo , Nitrito Reductasas/metabolismo , Nitritos/metabolismo , Animales , Células CHO , Hipoxia de la Célula/fisiología , Cricetulus , Cisteína/química , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Humanos , Proteínas Mitocondriales/química , Proteínas Mitocondriales/metabolismo , Nitrosación
5.
Arch Biochem Biophys ; 649: 47-52, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29752896

RESUMEN

Far red/near infrared (R/NIR) energy is a novel therapy, but its mechanism of action is poorly characterized. Cytochrome c oxidase (Cco) of the mitochondrial electron transport chain is considered the primary photoacceptor for R/NIR to photolyze a putative heme nitrosyl in Cco to liberate free nitric oxide (NO). We previously observed R/NIR light directly liberates NO from nitrosylated hemoglobin and myoglobin, and recently suggested S-nitrosothiols (RSNO) and dinitrosyl iron complexes (DNIC) may be primary sources of R/NIR-mediated NO. Here we indicate R/NIR light exposure induces wavelength dependent dilation of murine facial artery, with longer wavelengths (740, and 830 nm) exhibiting reduced potency when compared to 670 nm. R/NIR also stimulated NO release from pure solutions of low molecular weight RSNO (GSNO and SNAP) and glutathione dinitrosyl iron complex (GSH-DNIC) in a power- and wavelength-dependent manner, with the greatest effect at 670 nm. NO release from SNAP using 670 was nearly ten-fold more than GSNO or GSH-DNIC, with no substantial difference in NO production at 740 nm and 830 nm. Thermal effects of irradiation on vasodilation or NO release from S-nitrosothiols and DNIC was minimal. Our results suggest 670 nm is the optimal wavelength for R/NIR treatment of certain vascular-related diseases.


Asunto(s)
Arterias/efectos de los fármacos , Hierro/farmacología , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico/metabolismo , Óxidos de Nitrógeno/farmacología , S-Nitrosotioles/farmacología , Vasodilatación/efectos de los fármacos , Animales , Arterias/efectos de la radiación , Rayos Infrarrojos , Luz , Ratones Endogámicos C57BL , Vasodilatación/efectos de la radiación
6.
Nitric Oxide ; 65: 1-9, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28111306

RESUMEN

Dinitrosyl iron complexes (DNIC) spontaneously form in aqueous solutions of Fe(II), nitric oxide (NO), and various anions. They exist as an equilibrium between diamagnetic, dimeric (bi-DNIC) and paramagnetic, monomeric (mono-DNIC) forms. Thiolate groups (e.g., on glutathione or protein cysteine residues) are the most biologically relevant anions to coordinate to Fe(II). Low molecular weight DNIC have previously been suggested to be important mediators of NO biology in cells, and emerging literature supports their role in the control of iron-dependent cellular processes. Recently, it was shown that DNIC may be one of the most abundant NO-derived products in cells and may serve as intermediates in the cellular formation of S-nitrosothiols. In this work, we examined the stability of low molecular weight DNIC and investigated issues with their detection in the presence of other NO-dependent metabolites such as S-nitrosothiols. By using spectrophotometric, Electron Paramagnetic Resonance, ozone-based chemiluminesence, and HPLC techniques we established that at neutral pH, bi-DNIC remain stable for hours, whereas excess thiol results in decomposition to form nitrite. NO was also detected during the decomposition, but no S-nitrosothiol formation was observed. Importantly, mercury chloride accelerated the degradation of DNIC; thus, the implications of this finding for the diagnostic use of mercury chloride in the detection of S-nitrosothiols were determined in simple and complex biological systems. We conclude S-nitrosothiol levels may have been substantially overestimated in all methods where mercury chloride has been used.


Asunto(s)
Compuestos Ferrosos/análisis , S-Nitrosotioles/análisis , Animales , Cisteína/análogos & derivados , Cisteína/química , Cisteína/farmacología , Compuestos Ferrosos/química , Compuestos Ferrosos/metabolismo , Glutatión/análisis , Glutatión/química , Humanos , Concentración de Iones de Hidrógeno , Lipopolisacáridos/farmacología , Luminiscencia , Células MCF-7 , Ratones , Óxido Nítrico/análisis , Óxido Nítrico/metabolismo , Nitritos/análisis , Nitritos/síntesis química , Células RAW 264.7 , S-Nitrosotioles/química , S-Nitrosotioles/metabolismo , S-Nitrosotioles/farmacología , Espermina/análogos & derivados , Espermina/farmacología
7.
Blood ; 124(26): 3978-81, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25339362

RESUMEN

High mobility group box 1 (HMGB1) is a chromatin-binding protein that maintains DNA structure. On cellular activation or injury, HMGB1 is released from activated immune cells or necrotic tissues and acts as a damage-associated molecular pattern to activate Toll-like receptor 4 (TLR4). Little is known concerning HMGB1 release and TLR4 activity and their role in the pathology of inflammation of sickle cell disease (SCD). Circulating HMGB1 levels were increased in both humans and mice with SCD compared with controls. Furthermore, sickle plasma increased HMGB1-dependent TLR4 activity compared with control plasma. HMGB1 levels were further increased during acute sickling events (vasoocclusive crises in humans or hypoxia/reoxygenation injury in mice). Anti-HMGB1 neutralizing antibodies reduced the majority of sickle plasma-induced TLR4 activity both in vitro and in vivo. These findings show that HMGB1 is the major TLR4 ligand in SCD and likely plays a critical role in SCD-mediated inflammation.


Asunto(s)
Anemia de Células Falciformes/metabolismo , Proteína HMGB1/metabolismo , Inflamación/metabolismo , Receptor Toll-Like 4/metabolismo , Anemia de Células Falciformes/inmunología , Animales , Regulación de la Expresión Génica , Humanos , Hipoxia/patología , Ligandos , Ratones , Ratones Endogámicos C57BL , Oxígeno/metabolismo , Transducción de Señal
8.
Biochim Biophys Acta ; 1840(2): 892-900, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23988402

RESUMEN

BACKGROUND: S-nitrosothiols have been recognized as biologically-relevant products of nitric oxide that are involved in many of the diverse activities of this free radical. SCOPE OF REVIEW: This review serves to discuss current methods for the detection and analysis of protein S-nitrosothiols. The major methods of S-nitrosothiol detection include chemiluminescence-based methods and switch-based methods, each of which comes in various flavors with advantages and caveats. MAJOR CONCLUSIONS: The detection of S-nitrosothiols is challenging and prone to many artifacts. Accurate measurements require an understanding of the underlying chemistry of the methods involved and the use of appropriate controls. GENERAL SIGNIFICANCE: Nothing is more important to a field of research than robust methodology that is generally trusted. The field of S-nitrosation has developed such methods but, as S-nitrosothiols are easy to introduce as artifacts, it is vital that current users learn from the lessons of the past. This article is part of a Special Issue entitled Current methods to study reactive oxygen species - pros and cons and biophysics of membrane proteins. Guest Editor: Christine Winterbourn.


Asunto(s)
Mediciones Luminiscentes/métodos , S-Nitrosotioles/análisis , Animales , Humanos
9.
Blood Cells Mol Dis ; 54(2): 183-8, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25488613

RESUMEN

Humans and mice with sickle cell disease (SCD) have rigid red blood cells (RBCs). Omega-3 fatty acids, such as docosahexanoic acid (DHA), may influence RBC deformability via incorporation into the RBC membrane. In this study, sickle cell (SS) mice were fed natural ingredient rodent diets supplemented with 3% DHA (DHA diet) or a control diet matched in total fat (CTRL diet). After 8weeks of feeding, we examined the RBCs for: 1) stiffness, as measured by atomic force microscopy; 2) deformability, as measured by ektacytometry; and 3) percent irreversibly sickled RBCs on peripheral blood smears. Using atomic force microscopy, it is found that stiffness is increased and deformability decreased in RBCs from SS mice fed CTRL diet compared to wild-type mice. In contrast, RBCs from SS mice fed DHA diet had markedly decreased stiffness and increased deformability compared to RBCs from SS mice fed CTRL diet. Furthermore, examination of peripheral blood smears revealed less irreversibly sickled RBCs in SS mice fed DHA diet as compared to CTRL diet. In summary, our findings indicate that DHA supplementation improves RBC flexibility and reduces irreversibly sickled cells by 40% in SS mice. These results point to potential therapeutic benefits of dietary omega-3 fatty acids in SCD.


Asunto(s)
Anemia de Células Falciformes/dietoterapia , Suplementos Dietéticos , Ácidos Docosahexaenoicos/administración & dosificación , Membrana Eritrocítica/efectos de los fármacos , Anemia de Células Falciformes/sangre , Anemia de Células Falciformes/patología , Animales , Modelos Animales de Enfermedad , Recuento de Eritrocitos , Deformación Eritrocítica/efectos de los fármacos , Membrana Eritrocítica/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Microscopía de Fuerza Atómica
10.
Biochim Biophys Acta ; 1830(5): 3173-81, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23416062

RESUMEN

BACKGROUND: S-Nitrosoglutathione (GSNO) is the S-nitrosated derivative of glutathione and is thought to be a critical mediator of the down stream signaling effects of nitric oxide (NO). GSNO has also been implicated as a contributor to various disease states. SCOPE OF REVIEW: This review focuses on the chemical nature of GSNO, its biological activities, the evidence that it is an endogenous mediator of NO action, and implications for therapeutic use. MAJOR CONCLUSIONS: GSNO clearly exerts its cellular actions through both NO- and S-nitrosation-dependent mechanisms; however, the chemical and biological aspects of this compound should be placed in the context of S-nitrosation as a whole. GENERAL SIGNIFICANCE: GSNO is a central intermediate in formation and degradation of cellular S-nitrosothiols with potential therapeutic applications; thus, it remains an important molecule of study. This article is part of a Special Issue entitled Cellular functions of glutathione.


Asunto(s)
S-Nitrosoglutatión/metabolismo , Animales , Glutatión/química , Glutatión/metabolismo , Humanos , Óxido Nítrico/química , Óxido Nítrico/metabolismo , Nitrosación , S-Nitrosoglutatión/química , S-Nitrosotioles/química , S-Nitrosotioles/metabolismo
11.
Blood ; 120(20): 4229-37, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23007404

RESUMEN

A nitric oxide synthase (NOS)-like activity has been demonstrated in human red blood cells (RBCs), but doubts about its functional significance, isoform identity and disease relevance remain. Using flow cytometry in combination with the nitric oxide (NO)-imaging probe DAF-FM we find that all blood cells form NO intracellularly, with a rank order of monocytes > neutrophils > lymphocytes > RBCs > platelets. The observation of a NO-related fluorescence within RBCs was unexpected given the abundance of the NO-scavenger oxyhemoglobin. Constitutive normoxic NO formation was abolished by NOS inhibition and intracellular NO scavenging, confirmed by laser-scanning microscopy and unequivocally validated by detection of the DAF-FM reaction product with NO using HPLC and LC-MS/MS. Using immunoprecipitation, ESI-MS/MS-based peptide sequencing and enzymatic assay we further demonstrate that human RBCs contain an endothelial NOS (eNOS) that converts L-(3)H-arginine to L-(3)H-citrulline in a Ca(2+)/calmodulin-dependent fashion. Moreover, in patients with coronary artery disease, red cell eNOS expression and activity are both lower than in age-matched healthy individuals and correlate with the degree of endothelial dysfunction. Thus, human RBCs constitutively produce NO under normoxic conditions via an active eNOS isoform, the activity of which is compromised in patients with coronary artery disease.


Asunto(s)
Enfermedad de la Arteria Coronaria/enzimología , Eritrocitos/enzimología , Óxido Nítrico Sintasa de Tipo III/sangre , Adulto , Secuencia de Aminoácidos , Arginina/sangre , Cromatografía Líquida de Alta Presión , Citrulina/sangre , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/patología , Endotelio Vascular/patología , Citometría de Flujo , Fluoresceínas/análisis , Colorantes Fluorescentes/análisis , Humanos , Inmunoprecipitación , Espectrometría de Masas , Microscopía Confocal , Datos de Secuencia Molecular , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/sangre , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/fisiología , Oxihemoglobinas/metabolismo , Alineación de Secuencia , Análisis de Secuencia de Proteína , Homología de Secuencia de Aminoácido
12.
Free Radic Biol Med ; 215: 112-126, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38336101

RESUMEN

Murine sickle cell disease (SCD) results in damage to multiple organs, likely mediated first by vasculopathy. While the mechanisms inducing vascular damage remain to be determined, nitric oxide bioavailability and sterile inflammation are both considered to play major roles in vasculopathy. Here, we investigate the effects of high mobility group box-1 (HMGB1), a pro-inflammatory damage-associated molecular pattern (DAMP) molecule on endothelial-dependent vasodilation and lung morphometrics, a structural index of damage in sickle (SS) mice. SS mice were treated with either phosphate-buffered saline (PBS), hE-HMGB1-BP, an hE dual-domain peptide that binds and removes HMGB1 from the circulation via the liver, 1-[4-(aminocarbonyl)-2-methylphenyl]-5-[4-(1H-imidazol-1-yl)phenyl]-1H-pyrrole-2-propanoic acid (N6022) or N-acetyl-lysyltyrosylcysteine amide (KYC) for three weeks. Human umbilical vein endothelial cells (HUVEC) were treated with recombinant HMGB1 (r-HMGB1), which increases S-nitrosoglutathione reductase (GSNOR) expression by ∼80%, demonstrating a direct effect of HMGB1 to increase GSNOR. Treatment of SS mice with hE-HMGB1-BP reduced plasma HMGB1 in SS mice to control levels and reduced GSNOR expression in facialis arteries isolated from SS mice by ∼20%. These changes were associated with improved endothelial-dependent vasodilation. Treatment of SS mice with N6022 also improved vasodilation in SS mice suggesting that targeting GSNOR also improves vasodilation. SCD decreased protein nitrosothiols (SNOs) and radial alveolar counts (RAC) and increased GSNOR expression and mean linear intercepts (MLI) in lungs from SS mice. The marked changes in pulmonary morphometrics and GSNOR expression throughout the lung parenchyma in SS mice were improved by treating with either hE-HMGB1-BP or KYC. These data demonstrate that murine SCD induces vasculopathy and chronic lung disease by an HMGB1- and GSNOR-dependent mechanism and suggest that HMGB1 and GSNOR might be effective therapeutic targets for reducing vasculopathy and chronic lung disease in humans with SCD.


Asunto(s)
Anemia de Células Falciformes , Benzamidas , Proteína HMGB1 , Enfermedades Pulmonares , Lesión Pulmonar , Pirroles , Enfermedades Vasculares , Humanos , Animales , Ratones , Lesión Pulmonar/etiología , Proteína HMGB1/genética , Células Endoteliales/metabolismo , Anemia de Células Falciformes/tratamiento farmacológico , Anemia de Células Falciformes/genética , Inflamación , Enfermedades Vasculares/etiología
13.
Biochemistry ; 52(25): 4364-72, 2013 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-23718265

RESUMEN

When produced at physiological levels, reactive oxygen species (ROS) can act as signaling molecules to regulate normal vascular function. Produced under pathological conditions, ROS can contribute to the oxidative damage of cellular components (e.g., DNA and proteins) and trigger cell death. Moreover, the reaction of superoxide with nitric oxide (NO) produces the strong oxidant peroxynitrite and decreases NO bioavailability, both of which may contribute to activation of cell death pathways. The effects of ROS generated from the 1,4-naphthoquinones alone and in combination with NO on the activation status of poly(ADP-ribose) polymerase (PARP) and cell viability were examined. Treatment with redox cycling quinones activates PARP, and this stimulatory effect is attenuated in the presence of NO. Mitochondria play a central role in cell death signaling pathways and are a target of oxidants. We show that simultaneous exposure of endothelial cells to NO and ROS results in mitochondrial dysfunction, ATP and NAD(+) depletion, and cell death. Alone, NO and ROS have only minor effects on cellular bioenergetics. Further, PARP inhibition does not attenuate reduced cell viability or mitochondrial dysfunction. These results show that concomitant exposure to NO and ROS impairs energy metabolism and triggers PARP-independent cell death. While superoxide-mediated PARP activation is attenuated in the presence of NO, PARP inhibition does not modify the loss of mitochondrial function or adenine and pyridine nucleotide pools and subsequent bioenergetic dysfunction. These findings suggest that the mechanisms by which ROS and NO induce endothelial cell death are closely linked to the maintenance of mitochondrial function and not overactivation of PARP.


Asunto(s)
Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Naftoquinonas/toxicidad , Óxido Nítrico/química , Poli(ADP-Ribosa) Polimerasas/metabolismo , Animales , Aorta/enzimología , Aorta/metabolismo , Aorta/patología , Bovinos , Muerte Celular/efectos de los fármacos , Células Cultivadas , Endotelio Vascular/enzimología , Metabolismo Energético/efectos de los fármacos , Mitocondrias/enzimología , Mitocondrias/metabolismo , Mitocondrias/patología , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Especies Reactivas de Oxígeno/farmacología
14.
J Biol Chem ; 287(22): 18262-74, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22493289

RESUMEN

The reductive nitrosylation of ferric (met)hemoglobin is of considerable interest and remains incompletely explained. We have previously observed that at low NO concentrations the reaction with tetrameric hemoglobin occurs with an observed rate constant that is at least 5 times faster than that observed at higher concentrations. This was ascribed to a faster reaction of NO with a methemoglobin-nitrite complex. We now report detailed studies of this reaction of low NO with methemoglobin. Nitric oxide paradoxically reacts with ferric hemoglobin with faster observed rate constants at the lower NO concentration in a manner that is not affected by changes in nitrite concentration, suggesting that it is not a competition between NO and nitrite, as we previously hypothesized. By evaluation of the fast reaction in the presence of allosteric effectors and isolated ß- and α-chains of hemoglobin, it appears that NO reacts with a subpopulation of ß-subunit ferric hemes whose population is influenced by quaternary state, redox potential, and hemoglobin dimerization. To further characterize the role of nitrite, we developed a system that oxidizes nitrite to nitrate to eliminate nitrite contamination. Removal of nitrite does not alter reaction kinetics, but modulates reaction products, with a decrease in the formation of S-nitrosothiols. These results are consistent with the formation of NO(2)/N(2)O(3) in the presence of nitrite. The observed fast reductive nitrosylation observed at low NO concentrations may function to preserve NO bioactivity via primary oxidation of NO to form nitrite or in the presence of nitrite to form N(2)O(3) and S-nitrosothiols.


Asunto(s)
Hemoglobinas/metabolismo , Óxido Nítrico/metabolismo , Compuestos Nitrosos/metabolismo , Regulación Alostérica , Humanos , Concentración de Iones de Hidrógeno , Oxidación-Reducción , Unión Proteica
15.
Am J Physiol Lung Cell Mol Physiol ; 304(6): L445-55, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23333803

RESUMEN

Lipopolysaccharide (LPS)-mediated endothelial activation contributes to lung inflammation and alveolar remodeling seen in premature infants with bronchopulmonary dysplasia (BPD). The mechanisms underlying LPS-mediated oxidative stress and proinflammatory signaling in human pulmonary microvascular endothelial cells (HPMEC) remain unclear. We hypothesized that NADPH oxidase (Nox) mediates LPS-induced endothelial activation in HPMEC by regulating phosphorylation of Toll-like receptor (TLR) pathway proteins. LPS-induced expression of intercellular adhesion molecule 1 (ICAM-1) was associated with increased 2-OH-E(+) (marker for superoxide formation) levels and was attenuated by apocynin and the Nox inhibitor, VAS2870. LPS triggered membrane translocation of p67phox, suggesting activation of Nox2. Silencing Nox2, but not Nox4, suppressed LPS-induced ICAM-1 expression in HPMEC. Immunoprecipitation studies showed that inhibitor of κ-B kinase-ß (IKK-ß) serine phosphorylation induced by LPS was inhibited by Nox2 silencing. We examined whether Nox2-dependent, LPS-mediated IKK-ß phosphorylation was regulated by protein phosphatase 2A (PP2A) or TGF-ß associated kinase-1 (TAK1) in HPMEC. LPS increased PP2A activity in HPMEC, and inhibition of PP2A did not alter LPS-mediated ICAM-1 expression but attenuated IKK-ß phosphorylation. TAK1 inhibition decreased LPS-induced ICAM-1 expression in HPMEC, and Nox2 silencing attenuated LPS-mediated TAK1 phosphorylation (Thr184/187). We demonstrate that Nox2 regulates LPS-mediated endothelial activation in pulmonary endothelial cells by modulating phosphorylation of key kinases in the TLR signaling cascade. Our data support a novel mechanism by which Nox-dependent signaling regulates proinflammatory signaling in pulmonary endothelial cells. Inhibition of vascular Nox may potentially limit lung injury and alveolar remodeling caused by infections in BPD.


Asunto(s)
Células Endoteliales/enzimología , Quinasa I-kappa B/metabolismo , Lipopolisacáridos/farmacología , Glicoproteínas de Membrana/fisiología , Microvasos/citología , NADPH Oxidasas/fisiología , Procesamiento Proteico-Postraduccional , Células Cultivadas , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Isoenzimas/fisiología , Pulmón/irrigación sanguínea , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , NADPH Oxidasa 2 , NADPH Oxidasa 4 , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Proteína Fosfatasa 2/metabolismo , Proteína Fosfatasa 2/fisiología , Transporte de Proteínas , ARN Interferente Pequeño/genética , Superóxidos/metabolismo
16.
Am J Physiol Heart Circ Physiol ; 304(2): H328-36, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23125208

RESUMEN

Hemolysis can saturate the hemoglobin (Hb)/heme scavenging system, resulting in increased circulating cell-free Hb (CF-Hb) in hereditary and acquired hemolytic disease. While recent studies have suggested a central role for intravascular hemolysis and CF-Hb in the development of vascular dysfunction, this concept has stimulated considerable debate. This highlights the importance of determining the contribution of CF-Hb to vascular complications associated with hemolysis. Therefore, a novel Hb-binding peptide was synthesized and linked to a small fragment of apolipoprotein E (amino acids 141-150) to facilitate endocytic clearance. Plasma clearance of hE-Hb-b10 displayed a rapid phase t(1/2) of 16 min and slow phase t(1/2) of 10 h, trafficking primarily through the liver. Peptide hE-Hb-B10 decreased CF-Hb in mice treated with phenylhydrazine, a model of acute hemolysis. Administration of hE-Hb-B10 also attenuated CF-Hb in two models of chronic hemolysis: Berkeley sickle cell disease (SS) mice and mice with severe hereditary spherocytosis (HS). The hemolytic rate was unaltered in either chronic hemolysis model, supporting the conclusion that hE-Hb-B10 promotes CF-Hb clearance without affecting erythrocyte lysis. Interestingly, hE-Hb-B10 also decreased plasma ALT activity in SS and HS mice. Although acetylcholine-mediated facialis artery vasodilation was not improved by hE-Hb-B10 treatment, the peptide shifted vascular response in favor of NO-dependent vasodilation in SS mice. Taken together, these data demonstrate that hE-Hb-B10 decreases CF-Hb with a concomitant reduction in liver injury and changes in vascular response. Therefore, hE-Hb-B10 can be used to investigate the different roles of CF-Hb in hemolytic pathology and may have therapeutic benefit in the treatment of CF-Hb-mediated tissue damage.


Asunto(s)
Anemia Hemolítica/tratamiento farmacológico , Apolipoproteínas E/farmacología , Endocitosis/efectos de los fármacos , Hemoglobinas/metabolismo , Hemólisis , Hígado/efectos de los fármacos , Enfermedad Aguda , Anemia Hemolítica/sangre , Anemia Hemolítica/etiología , Anemia Hemolítica/fisiopatología , Anemia de Células Falciformes/sangre , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/tratamiento farmacológico , Animales , Apolipoproteínas E/sangre , Apolipoproteínas E/farmacocinética , Enfermedad Crónica , Modelos Animales de Enfermedad , Semivida , Humanos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Óxido Nítrico/metabolismo , Fragmentos de Péptidos/sangre , Fragmentos de Péptidos/farmacología , Péptidos/sangre , Péptidos/farmacología , Fenilhidrazinas , Unión Proteica , Transporte de Proteínas , Esferocitosis Hereditaria/sangre , Esferocitosis Hereditaria/complicaciones , Esferocitosis Hereditaria/tratamiento farmacológico , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
17.
Cytometry A ; 83(2): 242-50, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23125136

RESUMEN

There are increased levels of circulating microparticles (MPs) in several disease states. Flow cytometry is a common method to examine MPs, but their small size necessitates the use of markers to distinguish specifically MPs from artifact. Annexin V, which binds phosphatidylserine, is a commonly used marker for MP detection. Annexin V requires millimolar calcium ion for optimum binding. Ca(++) can precipitate with phosphate in phosphate-buffered saline (PBS). Calcium-phosphate microprecipitates were formed by titrating Ca(++) into PBS and examined using flow cytometry. Calcium-phosphate microprecipitates were compared with MPs derived from aged donor blood units. Microprecipitates were ∼0.7-0.9 µm in diameter compared with standard beads of known size. The microprecipitates disappeared with the addition of Ca(++) chelator. When we added fluorescently labeled antibodies to microprecipitates, the median fluorescent signal increased with increasing Ca(++) concentration regardless of specificity of the antibody. When repeated with a biological sample, there was an apparent increase in the fluorescent signal that returned to baseline after Ca(++) chelation. The flow cytometry signal of calcium-phosphate microprecipitates overlaps with the MP signal. Since Ca(++) is essential for annexin V binding, it is essential to avoid artifacts from calcium-phosphate microprecipitates when using any buffer or biological fluid containing phosphate. This also highlights the potential utility of flow cytometry for the analysis of crystals in biological fluids.


Asunto(s)
Artefactos , Fosfatos de Calcio/química , Citometría de Flujo/normas , Anexina A5/química , Micropartículas Derivadas de Células , Precipitación Química , Técnica del Anticuerpo Fluorescente , Humanos , Luz , Tamaño de la Partícula , Fosfatidilserinas/química , Unión Proteica , Estándares de Referencia , Dispersión de Radiación
18.
Biochem J ; 444(3): 561-71, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22458763

RESUMEN

Recent studies have highlighted the fact that cancer cells have an altered metabolic phenotype, and this metabolic reprogramming is required to drive the biosynthesis pathways necessary for rapid replication and proliferation. Specifically, the importance of citric acid cycle-generated intermediates in the regulation of cancer cell proliferation has been recently appreciated. One function of MCTs (monocarboxylate transporters) is to transport the citric acid cycle substrate pyruvate across the plasma membrane and into mitochondria, and inhibition of MCTs has been proposed as a therapeutic strategy to target metabolic pathways in cancer. In the present paper, we examined the effect of different metabolic substrates (glucose and pyruvate) on mitochondrial function and proliferation in breast cancer cells. We demonstrated that cancer cells proliferate more rapidly in the presence of exogenous pyruvate when compared with lactate. Pyruvate supplementation fuelled mitochondrial oxygen consumption and the reserve respiratory capacity, and this increase in mitochondrial function correlated with proliferative potential. In addition, inhibition of cellular pyruvate uptake using the MCT inhibitor α-cyano-4-hydroxycinnamic acid impaired mitochondrial respiration and decreased cell growth. These data demonstrate the importance of mitochondrial metabolism in proliferative responses and highlight a novel mechanism of action for MCT inhibitors through suppression of pyruvate-fuelled mitochondrial respiration.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Mitocondrias/metabolismo , Transportadores de Ácidos Monocarboxílicos/antagonistas & inhibidores , Transportadores de Ácidos Monocarboxílicos/metabolismo , Ácido Pirúvico/antagonistas & inhibidores , Ácido Pirúvico/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Respiración de la Célula/efectos de los fármacos , Respiración de la Célula/fisiología , Ácidos Cumáricos/farmacología , Femenino , Humanos , Mitocondrias/efectos de los fármacos , Ácido Pirúvico/farmacología
19.
Biochem J ; 442(1): 191-7, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22070099

RESUMEN

S-nitrosothiols are products of nitric oxide (NO) metabolism that have been implicated in a plethora of signalling processes. However, mechanisms of S-nitrosothiol formation in biological systems are uncertain, and no efficient protein-mediated process has been identified. Recently, we observed that ferric cytochrome c can promote S-nitrosoglutathione formation from NO and glutathione by acting as an electron acceptor under anaerobic conditions. In the present study, we show that this mechanism is also robust under oxygenated conditions, that cytochrome c can promote protein S-nitrosation via a transnitrosation reaction and that cell lysate depleted of cytochrome c exhibits a lower capacity to synthesize S-nitrosothiols. Importantly, we also demonstrate that this mechanism is functional in living cells. Lower S-nitrosothiol synthesis activity, from donor and nitric oxide synthase-generated NO, was found in cytochrome c-deficient mouse embryonic cells as compared with wild-type controls. Taken together, these data point to cytochrome c as a biological mediator of protein S-nitrosation in cells. This is the most efficient and concerted mechanism of S-nitrosothiol formation reported so far.


Asunto(s)
Citocromos c/metabolismo , S-Nitrosotioles/metabolismo , Aerobiosis , Anaerobiosis , Animales , Antimicina A/farmacología , Células Cultivadas , Embrión de Mamíferos/metabolismo , Glutatión/metabolismo , Ratones , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo
20.
Br J Haematol ; 154(4): 502-11, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21595649

RESUMEN

Normally, cell free haemoglobin is bound by haptoglobin and efficiently cleared. However, the chronic haemolysis in sickle cell disease (SCD) overwhelms haptoglobin binding capacity and protein turnover, resulting in elevated cell free haemoglobin. Cell free haemoglobin acts as both a scavenger of vasoactive nitric oxide and a pro-oxidant. In addition, methaemoglobin (metHb) releases the haem moiety, which can bind to albumin to form methaemalbumin (metHSA). This study used electron paramagnetic resonance to detect metHSA in SCD plasma and demonstrated that haptoglobin prevents haem transfer from metHb to HSA. MetHSA may either provide a second line of defence against haemoglobin/haem-mediated oxidation or contribute to the pro-oxidant environment of SCD plasma. We demonstrated that HSA inhibited oxidative protein modification induced by metHb. Additionally, we showed that while metHb induced haem oxygenase 1 (HO-1), an indicator of oxidative stress, HSA attenuated metHb induction of this enzyme, thereby limiting the potential benefits of HO-1. Furthermore, HO-1 induction by metHSA was less than HO-1 induction by equimolar metHb not bound to albumin. Our findings confirm the presence of metHSA in SCD and suggest that haem transfer from metHb to HSA reduces the oxidative effects of free haemoglobin/haem on endothelium with both beneficial (reduced protein oxidation) and potentially harmful (reduced HO-1 induction) outcomes.


Asunto(s)
Anemia de Células Falciformes/sangre , Hemo-Oxigenasa 1/metabolismo , Metemalbúmina/biosíntesis , Animales , Bovinos , Células Cultivadas , Endotelio Vascular/metabolismo , Haptoglobinas/fisiología , Hemo/metabolismo , Humanos , Peroxidación de Lípido/fisiología , Metemalbúmina/metabolismo , Oxidación-Reducción , Unión Proteica/fisiología , Albúmina Sérica/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA