Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Pharm ; 19(1): 18-25, 2022 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-34846906

RESUMEN

Oxidative stress underlies the pathology of many human diseases, including the doxorubicin-induced off-target cardiotoxicity in cancer chemotherapies. Since current diagnostic procedures are only capable of monitoring cardiac function, a noninvasive means of detecting biochemical changes in redox status prior to irreversible functional changes is highly desirable for both early diagnosis and prognosis. We designed a novel 18F-labeled molecular probe, 18F-FPBT, for the direct detection of superoxide in vivo using positron emission tomography (PET). 18F-FPBT was radiosynthesized in one step by nucleophilic radiofluorination. In vitro, 18F-FPBT showed rapid and selective oxidation by superoxide (around 60% in 5 min) compared to other physiological ROS. In healthy mice and rats, 18F-FBPT is distributed to all major organs in the first few minutes post injection and is rapidly cleared via both renal and hepatobiliary routes with minimal background retention in the heart. In a rat model of doxorubicin-induced cardiotoxicity, 18F-FBPT showed significantly higher (P < 0.05) uptake in the hearts of treated animals compared to healthy controls. These results warrant further optimization of 18F-FBPT for clinical translation.


Asunto(s)
Cardiotoxicidad/diagnóstico por imagen , Tomografía de Emisión de Positrones/métodos , Animales , Cardiotoxicidad/etiología , Modelos Animales de Enfermedad , Doxorrubicina/toxicidad , Ecocardiografía , Radioisótopos de Flúor , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Ratas , Ratas Wistar
2.
J Cell Mol Med ; 24(6): 3534-3548, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32040259

RESUMEN

Cardiac ischaemia-reperfusion (I/R) injury has been attributed to stress signals arising from an impaired mitochondrial electron transport chain (ETC), which include redox imbalance, metabolic stalling and excessive production of reactive oxygen species (ROS). The alternative oxidase (AOX) is a respiratory enzyme, absent in mammals, that accepts electrons from a reduced quinone pool to reduce oxygen to water, thereby restoring electron flux when impaired and, in the process, blunting ROS production. Hence, AOX represents a natural rescue mechanism from respiratory stress. This study aimed to determine how respiratory restoration through xenotopically expressed AOX affects the re-perfused post-ischaemic mouse heart. As expected, AOX supports ETC function and attenuates the ROS load in post-anoxic heart mitochondria. However, post-ischaemic cardiac remodelling over 3 and 9 weeks was not improved. AOX blunted transcript levels of factors known to be up-regulated upon I/R such as the atrial natriuretic peptide (Anp) whilst expression of pro-fibrotic and pro-apoptotic transcripts were increased. Ex vivo analysis revealed contractile failure at nine but not 3 weeks after ischaemia whilst label-free quantitative proteomics identified an increase in proteins promoting adverse extracellular matrix remodelling. Together, this indicates an essential role for ETC-derived signals during cardiac adaptive remodelling and identified ROS as a possible effector.


Asunto(s)
Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/fisiopatología , Transducción de Señal , Remodelación Ventricular , Animales , Biocatálisis , Transporte de Electrón , Matriz Extracelular/metabolismo , Masculino , Ratones , Mitocondrias Cardíacas/metabolismo , Proteínas Mitocondriales/metabolismo , Contracción Miocárdica , Isquemia Miocárdica/complicaciones , Isquemia Miocárdica/genética , Daño por Reperfusión Miocárdica/complicaciones , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/patología , Miocardio/ultraestructura , Oxidorreductasas/metabolismo , Proteínas de Plantas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
3.
Sci Rep ; 9(1): 499, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30679497

RESUMEN

Copper-64-Diacetyl-bis(N4-methylthiosemicarbazone) [64Cu][Cu(ATSM)] is a hypoxia-targeting PET tracer with applications in oncology and cardiology. Upon entering a hypoxic cell, [64Cu][Cu(II)(ATSM)] is reduced to a putative [64Cu][Cu(I)(ATSM)]- species which dissociates to deposit radiocopper, thereby providing hypoxic contrast. This process may be dependent upon protonation arising from intracellular acidosis. Since acidosis is a hallmark of ischemic tissue and tumors, the hypoxia specificity of [64Cu][Cu(ATSM)] may be confounded by changes in intracellular pH. We have therefore determined the influence of intracellular pH on [64Cu][Cu(ATSM)] pharmacokinetics. Using isolated perfused rat hearts, acidosis was induced using an ammonium pre-pulse method, with and without hypoxic buffer perfusion. Cardiac [64Cu][Cu(ATSM)] pharmacokinetics were determined using NaI detectors, with intracellular pH and cardiac energetics monitored in parallel by 31P NMR. To distinguish direct acidotic effects on tracer pharmacokinetics from acidosis-induced hypocontractility, parallel studies used lidocaine perfusion to abolish cardiac contraction. Hypoxic myocardium trapped [64Cu][Cu(ATSM)] despite no evidence of it being acidotic when characterised by 31P NMR. Independent induction of tissue acidosis had no direct effect on [64Cu][Cu(ATSM)] pharmacokinetics in either normoxic or hypoxic hearts, beyond decreasing cardiac oxygen consumption to alleviate hypoxia and decrease tracer retention, leading us to conclude that tissue acidosis does not mediate the hypoxia selectivity of [64Cu][Cu(ATSM)].


Asunto(s)
Radioisótopos de Cobre , Espectroscopía de Resonancia Magnética , Isquemia Miocárdica , Miocardio , Acidosis , Animales , Radioisótopos de Cobre/farmacocinética , Radioisótopos de Cobre/farmacología , Masculino , Isquemia Miocárdica/diagnóstico por imagen , Isquemia Miocárdica/metabolismo , Miocardio/metabolismo , Miocardio/patología , Perfusión , Radiofármacos/farmacocinética , Radiofármacos/farmacología , Ratas , Ratas Wistar
4.
Sci Rep ; 9(1): 216, 2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30659226

RESUMEN

By the time cardiotoxicity-associated cardiac dysfunction is detectable by echocardiography it is often beyond meaningful intervention. 99mTc-sestamibi is used clinically to image cardiac perfusion by single photon emission computed tomography (SPECT) imaging, but as a lipophilic cation its distribution is also governed by mitochondrial membrane potential (ΔΨm). Correcting scans for variations in perfusion (using a ΔΨm-independent perfusion tracer such as (bis(N-ethoxy-N-ethyldithiocarbamato)nitrido 99mTc(V)) (99mTc-NOET) could allow 99mTc-sestamibi to be repurposed to specifically report on ΔΨm as a readout of evolving cardiotoxicity. Isolated rat hearts were perfused within a γ-detection apparatus to characterize the pharmacokinetics of 99mTc-sestamibi and 99mTc-NOET in response to mitochondrial perturbation by hypoxia, ionophore (CCCP) or doxorubicin. All interventions induced 99mTc-sestamibi washout; hypoxia from 24.9 ± 2.6% ID to 0.4 ± 6.2%, CCCP from 22.8 ± 2.5% ID to -3.5 ± 3.1%, and doxorubicin from 23.0 ± 2.2% ID to 17.8 ± 0.7, p < 0.05. Cardiac 99mTc-NOET retention (34.0 ± 8.0% ID) was unaffected in all cases. Translating to an in vivo rat model, 2 weeks after bolus doxorubicin injection, there was a dose-dependent loss of cardiac 99mTc-sestamibi retention (from 2.3 ± 0.3 to 0.9 ± 0.2 ID/g with 10 mg/kg (p < 0.05)), while 99mTc-NOET retention (0.93 ± 0.16 ID/g) was unaffected. 99mTc-NOET therefore traps in myocardium independently of the mitochondrial perturbations that induce 99mTc-sestamibi washout, demonstrating proof-of-concept for an imaging approach to detect evolving cardiotoxicity.


Asunto(s)
Cardiotoxicidad/diagnóstico por imagen , Tomografía Computarizada de Emisión de Fotón Único/métodos , Animales , Antraciclinas/toxicidad , Circulación Coronaria/fisiología , Corazón/diagnóstico por imagen , Masculino , Miocardio/metabolismo , Compuestos de Organotecnecio/farmacocinética , Perfusión/métodos , Cintigrafía , Radiofármacos/farmacocinética , Ratas , Ratas Wistar , Tecnecio Tc 99m Sestamibi/metabolismo
5.
J Mol Cell Cardiol ; 123: 88-91, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30118790

RESUMEN

Ischemia-reperfusion (IR) injury occurs when blood supply to an organ is disrupted and then restored, and underlies many disorders, notably myocardial infarction and stroke. While reperfusion of ischemic tissue is essential for survival, it also initiates cell death through generation of mitochondrial reactive oxygen species (ROS). Recent work has revealed a novel pathway underlying ROS production at reperfusion in vivo in which the accumulation of succinate during ischemia and its subsequent rapid oxidation at reperfusion drives ROS production at complex I by reverse electron transport (RET). Pharmacologically inhibiting ischemic succinate accumulation, or slowing succinate metabolism at reperfusion, have been shown to be cardioprotective against IR injury. Here, we determined whether ischemic preconditioning (IPC) contributes to cardioprotection by altering kinetics of succinate accumulation and oxidation during IR. Mice were subjected to a 30-minute occlusion of the left anterior descending coronary artery followed by reperfusion, with or without a protective IPC protocol prior to sustained ischemia. We found that IPC had no effect on ischemic succinate accumulation with both control and IPC mice having profound increases in succinate compared to normoxia. Furthermore, after only 1-minute reperfusion succinate was rapidly metabolised returning to near pre-ischemic levels in both groups. We conclude that IPC does not affect ischemic succinate accumulation, or its oxidation at reperfusion.


Asunto(s)
Precondicionamiento Isquémico Miocárdico , Daño por Reperfusión Miocárdica/metabolismo , Oxidación-Reducción , Ácido Succínico/metabolismo , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Metabolismo Energético , Masculino , Metaboloma , Metabolómica/métodos , Ratones , Mitocondrias/metabolismo , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Miocardio/patología , Especies Reactivas de Oxígeno/metabolismo
6.
J Am Heart Assoc ; 7(8)2018 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-29626151

RESUMEN

BACKGROUND: Ischemia-reperfusion injury following ST-segment-elevation myocardial infarction (STEMI) is a leading determinant of clinical outcome. In experimental models of myocardial ischemia, succinate accumulation leading to mitochondrial dysfunction is a major cause of ischemia-reperfusion injury; however, the potential importance and specificity of myocardial succinate accumulation in human STEMI is unknown. We sought to identify the metabolites released from the heart in patients undergoing primary percutaneous coronary intervention for emergency treatment of STEMI. METHODS AND RESULTS: Blood samples were obtained from the coronary artery, coronary sinus, and peripheral vein in patients undergoing primary percutaneous coronary intervention for acute STEMI and in control patients undergoing nonemergency coronary angiography or percutaneous coronary intervention for stable angina or non-STEMI. Plasma metabolites were analyzed by targeted liquid chromatography and mass spectrometry. Metabolite levels for coronary artery, coronary sinus, and peripheral vein were compared to derive cardiac and systemic release ratios. In STEMI patients, cardiac magnetic resonance imaging was performed 2 days and 6 months after primary percutaneous coronary intervention to quantify acute myocardial edema and final infarct size, respectively. In total, 115 patients undergoing acute STEMI and 26 control patients were included. Succinate was the only metabolite significantly increased in coronary sinus blood compared with venous blood in STEMI patients, indicating cardiac release of succinate. STEMI patients had higher succinate concentrations in arterial, coronary sinus, and peripheral venous blood than patients with non-STEMI or stable angina. Furthermore, cardiac succinate release in STEMI correlated with the extent of acute myocardial injury, quantified by cardiac magnetic resonance imaging. CONCLUSION: Succinate release by the myocardium correlates with the extent of ischemia.


Asunto(s)
Daño por Reperfusión Miocárdica/sangre , Infarto del Miocardio con Elevación del ST/sangre , Ácido Succínico/sangre , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Angiografía Coronaria , Femenino , Estudios de Seguimiento , Humanos , Imagen por Resonancia Cinemagnética , Masculino , Persona de Mediana Edad , Daño por Reperfusión Miocárdica/diagnóstico , Daño por Reperfusión Miocárdica/etiología , Miocardio/metabolismo , Miocardio/patología , Intervención Coronaria Percutánea/métodos , Pronóstico , Estudios Retrospectivos , Infarto del Miocardio con Elevación del ST/complicaciones , Infarto del Miocardio con Elevación del ST/cirugía , Factores de Tiempo
7.
Artículo en Inglés | MEDLINE | ID: mdl-29515752

RESUMEN

PURPOSE OF REVIEW: In this review, we outline the potential for hypoxia imaging as a diagnostic and prognostic tool in cardiology. We describe the lead hypoxia PET radiotracers currently in development and propose a rationale for how they should most appropriately be screened and validated. RECENT FINDINGS: While the majority of hypoxia imaging agents has been developed for oncology, the requirements for hypoxia imaging in cardiology are different. Recent work suggests that the bis(thiosemicarbazone) family of compounds may be capable of detecting the subtle degrees of hypoxia associated with cardiovascular syndromes, and that they have the potential to be "tuned" to provide different tracers for different applications. SUMMARY: New tracers currently in development show significant promise for imaging evolving cardiovascular disease. Fundamental to their exploitation is their careful, considered validation and characterization so that the information they provide delivers the greatest prognostic insight achievable.

8.
Cell Chem Biol ; 24(10): 1285-1298.e12, 2017 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-28890317

RESUMEN

Mitochondrial superoxide (O2⋅-) underlies much oxidative damage and redox signaling. Fluorescent probes can detect O2⋅-, but are of limited applicability in vivo, while in cells their usefulness is constrained by side reactions and DNA intercalation. To overcome these limitations, we developed a dual-purpose mitochondrial O2⋅- probe, MitoNeoD, which can assess O2⋅- changes in vivo by mass spectrometry and in vitro by fluorescence. MitoNeoD comprises a O2⋅--sensitive reduced phenanthridinium moiety modified to prevent DNA intercalation, as well as a carbon-deuterium bond to enhance its selectivity for O2⋅- over non-specific oxidation, and a triphenylphosphonium lipophilic cation moiety leading to the rapid accumulation within mitochondria. We demonstrated that MitoNeoD was a versatile and robust probe to assess changes in mitochondrial O2⋅- from isolated mitochondria to animal models, thus offering a way to examine the many roles of mitochondrial O2⋅- production in health and disease.


Asunto(s)
Mitocondrias/metabolismo , Sondas Moleculares/metabolismo , Superóxidos/metabolismo , Animales , Transporte Biológico , Línea Celular , ADN/química , ADN/metabolismo , Masculino , Espectrometría de Masas , Ratones , Modelos Moleculares , Sondas Moleculares/química , Conformación de Ácido Nucleico , Oxidación-Reducción
9.
J Biol Chem ; 292(35): 14486-14495, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28710281

RESUMEN

Nitrate (NO3-) and nitrite (NO2-) are known to be cardioprotective and to alter energy metabolism in vivo NO3- action results from its conversion to NO2- by salivary bacteria, but the mechanism(s) by which NO2- affects metabolism remains obscure. NO2- may act by S-nitrosating protein thiols, thereby altering protein activity. But how this occurs, and the functional importance of S-nitrosation sites across the mammalian proteome, remain largely uncharacterized. Here we analyzed protein thiols within mouse hearts in vivo using quantitative proteomics to determine S-nitrosation site occupancy. We extended the thiol-redox proteomic technique, isotope-coded affinity tag labeling, to quantify the extent of NO2--dependent S-nitrosation of proteins thiols in vivo Using this approach, called SNOxICAT (S-nitrosothiol redox isotope-coded affinity tag), we found that exposure to NO2- under normoxic conditions or exposure to ischemia alone results in minimal S-nitrosation of protein thiols. However, exposure to NO2- in conjunction with ischemia led to extensive S-nitrosation of protein thiols across all cellular compartments. Several mitochondrial protein thiols exposed to the mitochondrial matrix were selectively S-nitrosated under these conditions, potentially contributing to the beneficial effects of NO2- on mitochondrial metabolism. The permeability of the mitochondrial inner membrane to HNO2, but not to NO2-, combined with the lack of S-nitrosation during anoxia alone or by NO2- during normoxia places constraints on how S-nitrosation occurs in vivo and on its mechanisms of cardioprotection and modulation of energy metabolism. Quantifying S-nitrosated protein thiols now allows determination of modified cysteines across the proteome and identification of those most likely responsible for the functional consequences of NO2- exposure.


Asunto(s)
Modelos Animales de Enfermedad , Mitocondrias Cardíacas/metabolismo , Isquemia Miocárdica/metabolismo , Miocardio/metabolismo , Nitritos/metabolismo , Procesamiento Proteico-Postraduccional , Regulación hacia Arriba , Marcadores de Afinidad/metabolismo , Animales , Cardiotónicos/farmacología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Cisteína/metabolismo , Femenino , Corazón/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Dilatación Mitocondrial/efectos de los fármacos , Isquemia Miocárdica/tratamiento farmacológico , Nitratos/farmacología , Nitritos/farmacología , Nitrosación/efectos de los fármacos , Compuestos de Potasio/farmacología , Proteómica/métodos , Ratas Wistar , Regulación hacia Arriba/efectos de los fármacos
10.
Cardiovasc Res ; 111(2): 134-41, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27194563

RESUMEN

Myocardial ischaemia/reperfusion (IR) injury is a major cause of death worldwide and remains a disease for which current clinical therapies are strikingly deficient. While the production of mitochondrial reactive oxygen species (ROS) is a critical driver of tissue damage upon reperfusion, the precise mechanisms underlying ROS production have remained elusive. More recently, it has been demonstrated that a specific metabolic mechanism occurs during ischaemia that underlies elevated ROS at reperfusion, suggesting a unifying model as to why so many different compounds have been found to be cardioprotective against IR injury. This review will discuss the role of the citric acid cycle intermediate succinate in IR pathology focusing on the mechanism by which this metabolite accumulates during ischaemia and how it can drive ROS production at Complex I via reverse electron transport. We will then examine the potential for manipulating succinate accumulation and metabolism during IR injury in order to protect the heart against IR damage and discuss targets for novel therapeutics designed to reduce reperfusion injury in patients.


Asunto(s)
Metabolismo Energético , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Ácido Succínico/metabolismo , Animales , Fármacos Cardiovasculares/farmacología , Transporte de Electrón , Complejo I de Transporte de Electrón/metabolismo , Complejo II de Transporte de Electrones/metabolismo , Metabolismo Energético/efectos de los fármacos , Humanos , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/patología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
11.
Circ Res ; 118(5): 898-906, 2016 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-26941425

RESUMEN

Mitochondrial reactive oxygen species production has emerged as an important pathological mechanism in myocardial ischemia/reperfusion injury. Attempts at targeting reactive oxygen species by scavenging using antioxidants have, however, been clinically disappointing. This review will provide an overview of the current understanding of mitochondrial reactive oxygen species in ischemia/reperfusion injury. We will outline novel therapeutic approaches designed to directly target the mitochondrial respiratory chain and prevent excessive reactive oxygen species production and its associated pathology. This approach could lead to more effective interventions in an area where there is an urgent need for new treatments.


Asunto(s)
Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/metabolismo , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Fármacos Cardiovasculares/administración & dosificación , Sistemas de Liberación de Medicamentos/tendencias , Humanos , Especies Reactivas de Oxígeno/antagonistas & inhibidores
12.
Cell Metab ; 23(2): 254-63, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26777689

RESUMEN

Ischemia-reperfusion (IR) injury occurs when blood supply to an organ is disrupted--ischemia--and then restored--reperfusion--leading to a burst of reactive oxygen species (ROS) from mitochondria. It has been tacitly assumed that ROS production during IR is a non-specific consequence of oxygen interacting with dysfunctional mitochondria upon reperfusion. Recently, this view has changed, suggesting that ROS production during IR occurs by a defined mechanism. Here we survey the metabolic factors underlying IR injury and propose a unifying mechanism for its causes that makes sense of the huge amount of disparate data in this area and provides testable hypotheses and new directions for therapies.


Asunto(s)
Mitocondrias/metabolismo , Daño por Reperfusión/metabolismo , Superóxidos/metabolismo , Animales , Transporte de Electrón , Humanos , Modelos Biológicos , Compuestos de Sulfhidrilo/metabolismo
13.
Cell Metab ; 23(2): 379-85, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26712463

RESUMEN

The mitochondrial membrane potential (Δψm) is a major determinant and indicator of cell fate, but it is not possible to assess small changes in Δψm within cells or in vivo. To overcome this, we developed an approach that utilizes two mitochondria-targeted probes each containing a triphenylphosphonium (TPP) lipophilic cation that drives their accumulation in response to Δψm and the plasma membrane potential (Δψp). One probe contains an azido moiety and the other a cyclooctyne, which react together in a concentration-dependent manner by "click" chemistry to form MitoClick. As the mitochondrial accumulation of both probes depends exponentially on Δψm and Δψp, the rate of MitoClick formation is exquisitely sensitive to small changes in these potentials. MitoClick accumulation can then be quantified by liquid chromatography-tandem mass spectrometry (LC-MS/MS). This approach enables assessment of subtle changes in membrane potentials within cells and in the mouse heart in vivo.


Asunto(s)
Química Clic/métodos , Potencial de la Membrana Mitocondrial , Espectrometría de Masas en Tándem/métodos , Animales , Línea Celular , Ratones Endogámicos C57BL , Sondas Moleculares/metabolismo
14.
Nature ; 515(7527): 431-435, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25383517

RESUMEN

Ischaemia-reperfusion injury occurs when the blood supply to an organ is disrupted and then restored, and underlies many disorders, notably heart attack and stroke. While reperfusion of ischaemic tissue is essential for survival, it also initiates oxidative damage, cell death and aberrant immune responses through the generation of mitochondrial reactive oxygen species (ROS). Although mitochondrial ROS production in ischaemia reperfusion is established, it has generally been considered a nonspecific response to reperfusion. Here we develop a comparative in vivo metabolomic analysis, and unexpectedly identify widely conserved metabolic pathways responsible for mitochondrial ROS production during ischaemia reperfusion. We show that selective accumulation of the citric acid cycle intermediate succinate is a universal metabolic signature of ischaemia in a range of tissues and is responsible for mitochondrial ROS production during reperfusion. Ischaemic succinate accumulation arises from reversal of succinate dehydrogenase, which in turn is driven by fumarate overflow from purine nucleotide breakdown and partial reversal of the malate/aspartate shuttle. After reperfusion, the accumulated succinate is rapidly re-oxidized by succinate dehydrogenase, driving extensive ROS generation by reverse electron transport at mitochondrial complex I. Decreasing ischaemic succinate accumulation by pharmacological inhibition is sufficient to ameliorate in vivo ischaemia-reperfusion injury in murine models of heart attack and stroke. Thus, we have identified a conserved metabolic response of tissues to ischaemia and reperfusion that unifies many hitherto unconnected aspects of ischaemia-reperfusion injury. Furthermore, these findings reveal a new pathway for metabolic control of ROS production in vivo, while demonstrating that inhibition of ischaemic succinate accumulation and its oxidation after subsequent reperfusion is a potential therapeutic target to decrease ischaemia-reperfusion injury in a range of pathologies.


Asunto(s)
Isquemia/metabolismo , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/metabolismo , Ácido Succínico/metabolismo , Adenosina Monofosfato/metabolismo , Animales , Ácido Aspártico/metabolismo , Ciclo del Ácido Cítrico , Modelos Animales de Enfermedad , Transporte de Electrón , Complejo I de Transporte de Electrón/metabolismo , Fumaratos/metabolismo , Isquemia/enzimología , Malatos/metabolismo , Masculino , Metabolómica , Ratones , Mitocondrias/enzimología , Infarto del Miocardio/enzimología , Infarto del Miocardio/metabolismo , Miocardio/citología , Miocardio/enzimología , Miocardio/metabolismo , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/metabolismo , NAD/metabolismo , Daño por Reperfusión/enzimología , Accidente Cerebrovascular/enzimología , Accidente Cerebrovascular/metabolismo , Succinato Deshidrogenasa/metabolismo
15.
Eur J Heart Fail ; 16(7): 712-7, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24891297

RESUMEN

AIMS: Recently it has been shown that the mitochondria-targeted S-nitrosothiol MitoSNO protects against acute ischaemia/reperfusion (IR) injury by inhibiting the reactivation of mitochondrial complex I in the first minutes of reperfusion of ischaemic tissue, thereby preventing free radical formation that underlies IR injury. However, it remains unclear how this transient inhibition of mitochondrial complex I-mediated free radicals at reperfusion affects the long-term recovery of the heart following IR injury. Here we determined whether the acute protection by MitoSNO at reperfusion prevented the subsequent development of post-myocardial infarction heart failure. METHODS AND RESULTS: Mice were subjected to 30 min left coronary artery occlusion followed by reperfusion and recovery over 28 days. MitoSNO (100 ng/kg) was applied 5 min before the onset of reperfusion followed by 20 min infusion (1 ng/kg/min). Infarct size and cardiac function were measured by magnetic resonance imaging (MRI) 24 h after infarction. MitoSNO-treated mice exhibited reduced infarct size and preserved function. In addition, MitoSNO at reperfusion improved outcome measures 28 days post-IR, including preserved systolic function (63.7 ±1.8% LVEF vs. 53.7 ± 2.1% in controls, P = 0.01) and tissue fibrosis. CONCLUSIONS: MitoSNO action acutely at reperfusion reduces infarct size and protects from post-myocardial infarction heart failure. Therefore, targeted inhibition of mitochondrial complex I in the first minutes of reperfusion by MitoSNO is a rational therapeutic strategy for preventing subsequent heart failure in patients undergoing IR injury.


Asunto(s)
Oclusión Coronaria , Radicales Libres/metabolismo , Insuficiencia Cardíaca/metabolismo , Corazón/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Infarto del Miocardio/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Donantes de Óxido Nítrico/farmacología , S-Nitrosotioles/farmacología , Animales , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/patología , Imagen por Resonancia Cinemagnética , Ratones , Mitocondrias/metabolismo , Proteínas Mitocondriales/efectos de los fármacos , Proteínas Mitocondriales/metabolismo , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Miocitos Cardíacos/metabolismo , Nitrosación
16.
Biochim Biophys Acta ; 1840(2): 923-30, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23726990

RESUMEN

BACKGROUND: The ability to measure the concentrations of small damaging and signalling molecules such as reactive oxygen species (ROS) in vivo is essential to understanding their biological roles. While a range of methods can be applied to in vitro systems, measuring the levels and relative changes in reactive species in vivo is challenging. SCOPE OF REVIEW: One approach towards achieving this goal is the use of exomarkers. In this, exogenous probe compounds are administered to the intact organism and are then transformed by the reactive molecules in vivo to produce a diagnostic exomarker. The exomarker and the precursor probe can be analysed ex vivo to infer the identity and amounts of the reactive species present in vivo. This is akin to the measurement of biomarkers produced by the interaction of reactive species with endogenous biomolecules. MAJOR CONCLUSIONS AND GENERAL SIGNIFICANCE: Our laboratories have developed mitochondria-targeted probes that generate exomarkers that can be analysed ex vivo by mass spectrometry to assess levels of reactive species within mitochondria in vivo. We have used one of these compounds, MitoB, to infer the levels of mitochondrial hydrogen peroxide within flies and mice. Here we describe the development of MitoB and expand on this example to discuss how better probes and exomarkers can be developed. 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)
Biomarcadores/análisis , Mitocondrias/metabolismo , Modelos Biológicos , Sondas Moleculares , Especies Reactivas de Oxígeno/análisis , Animales , Ratones , Estrés Oxidativo
17.
Nat Med ; 19(6): 753-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23708290

RESUMEN

Oxidative damage from elevated production of reactive oxygen species (ROS) contributes to ischemia-reperfusion injury in myocardial infarction and stroke. The mechanism by which the increase in ROS occurs is not known, and it is unclear how this increase can be prevented. A wide variety of nitric oxide donors and S-nitrosating agents protect the ischemic myocardium from infarction, but the responsible mechanisms are unclear. Here we used a mitochondria-selective S-nitrosating agent, MitoSNO, to determine how mitochondrial S-nitrosation at the reperfusion phase of myocardial infarction is cardioprotective in vivo in mice. We found that protection is due to the S-nitrosation of mitochondrial complex I, which is the entry point for electrons from NADH into the respiratory chain. Reversible S-nitrosation of complex I slows the reactivation of mitochondria during the crucial first minutes of the reperfusion of ischemic tissue, thereby decreasing ROS production, oxidative damage and tissue necrosis. Inhibition of complex I is afforded by the selective S-nitrosation of Cys39 on the ND3 subunit, which becomes susceptible to modification only after ischemia. Our results identify rapid complex I reactivation as a central pathological feature of ischemia-reperfusion injury and show that preventing this reactivation by modification of a cysteine switch is a robust cardioprotective mechanism and hence a rational therapeutic strategy.


Asunto(s)
Cisteína/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Mitocondrias Cardíacas/metabolismo , Proteínas Mitocondriales/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Nitrosación , Subunidades de Proteína , Ratas , Especies Reactivas de Oxígeno/metabolismo
18.
Life Sci ; 91(13-14): 544-9, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22480514

RESUMEN

AIMS: To determine the pharmacology of ET(A)- and ET(B)-mediated ß-arrestin recruitment and compare this to established human pharmacology of these receptors to identify evidence for endothelin receptor biased signalling and pathway specific blockade by antagonists. MAIN METHODS: The ability of ET-1, ET-2, ET-3, sarafotoxin 6b and sarafotoxin 6c to activate ET(A) and ET(B)-mediated ß-arrestin recruitment was determined in CHO-K1 cells. Affinities were obtained for ET(A) selective (BQ123, sitaxentan, ambrisentan), ET(B) selective (BQ788) and mixed (bosentan) antagonists using ET-1 and compared to affinities obtained in competition experiments in human heart and by Schild analysis in human saphenous vein. Agonist dependence of affinities was compared for BQ123 and BQ788 in the ET(A) and ET(B) ß-arrestin assays respectively. KEY FINDINGS: For ß-arrestin recruitment, order of potency was as expected for the ET(A) (ET-1≥ET-2>>ET-3) and ET(B) (ET-1=ET-2=ET-3) receptors. However, at the ET(A) receptor sarafotoxin 6b and ET-3 were partial agonists. Antagonism of ET peptides by selective and mixed antagonists appeared non-competitive. BQ123, but not BQ788, exhibited agonist-dependent affinities. Bosentan was significantly more effective an inhibitor of ß-arrestin recruitment mediated by ET(A) compared to the ET(B) receptor. In the ET(A) vasoconstrictor assay, ET-1, ET-2 and S6b were equipotent, full agonists and antagonists tested behaved in a competitive manner, although affinities were lower than predicted from the competition binding experiments in left ventricle. SIGNIFICANCE: These data suggest that the pharmacology of ET(A) and ET(B) receptors linked to G-protein- and ß-arrestin mediated responses was different and bosentan appeared to show bias, preferentially blocking ET(A) mediated ß-arrestin recruitment.


Asunto(s)
Arrestinas/metabolismo , Proteínas de Unión al GTP/metabolismo , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Transducción de Señal , Animales , Bosentán , Células CHO , Cricetinae , Cricetulus , Antagonistas de los Receptores de la Endotelina A , Antagonistas de los Receptores de la Endotelina B , Endotelina-1/farmacología , Endotelina-2/farmacología , Endotelina-3/farmacología , Humanos , Receptor de Endotelina A/agonistas , Receptor de Endotelina B/agonistas , Sulfonamidas/metabolismo , Vasoconstrictores/farmacología , Venenos de Víboras/farmacología , beta-Arrestinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...