Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 131(6): 528-541, 2022 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-35959683

RESUMEN

BACKGROUND: Inhibiting SDH (succinate dehydrogenase), with the competitive inhibitor malonate, has shown promise in ameliorating ischemia/reperfusion injury. However, key for translation to the clinic is understanding the mechanism of malonate entry into cells to enable inhibition of SDH, its mitochondrial target, as malonate itself poorly permeates cellular membranes. The possibility of malonate selectively entering the at-risk heart tissue on reperfusion, however, remains unexplored. METHODS: C57BL/6J mice, C2C12 and H9c2 myoblasts, and HeLa cells were used to elucidate the mechanism of selective malonate uptake into the ischemic heart upon reperfusion. Cells were treated with malonate while varying pH or together with transport inhibitors. Mouse hearts were either perfused ex vivo (Langendorff) or subjected to in vivo left anterior descending coronary artery ligation as models of ischemia/reperfusion injury. Succinate and malonate levels were assessed by liquid chromatography-tandem mass spectrometry LC-MS/MS, in vivo by mass spectrometry imaging, and infarct size by TTC (2,3,5-triphenyl-2H-tetrazolium chloride) staining. RESULTS: Malonate was robustly protective against cardiac ischemia/reperfusion injury, but only if administered at reperfusion and not when infused before ischemia. The extent of malonate uptake into the heart was proportional to the duration of ischemia. Malonate entry into cardiomyocytes in vivo and in vitro was dramatically increased at the low pH (≈6.5) associated with ischemia. This increased uptake of malonate was blocked by selective inhibition of MCT1 (monocarboxylate transporter 1). Reperfusion of the ischemic heart region with malonate led to selective SDH inhibition in the at-risk region. Acid-formulation greatly enhances the cardioprotective potency of malonate. CONCLUSIONS: Cardioprotection by malonate is dependent on its entry into cardiomyocytes. This is facilitated by the local decrease in pH that occurs during ischemia, leading to its selective uptake upon reperfusion into the at-risk tissue, via MCT1. Thus, malonate's preferential uptake in reperfused tissue means it is an at-risk tissue-selective drug that protects against cardiac ischemia/reperfusion injury.


Asunto(s)
Daño por Reperfusión Miocárdica , Animales , Cromatografía Liquida , Células HeLa , Humanos , Isquemia , Malonatos/farmacología , Malonatos/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos , Espectrometría de Masas en Tándem
2.
Diabet Med ; 40(5): e15064, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36782075

RESUMEN

AIMS: The aim of this scoping review is to evaluate the current biomarkers used in the assessment of adverse cardiac remodelling in people with diabetes mellitus (DM) and in the diagnosis and prognosis of subsequent cardiovascular disease. We aim to discuss the biomarkers' pathophysiological roles as a reflection of the cardiac remodelling mechanisms in the presence of DM. METHODS: We performed the literature search to include studies from 2003 to 2021 using the following databases: MEDLINE, Scopus, Web of Science, PubMed, and Cochrane library. Articles that met our inclusion criteria were screened and appraised before being included in this review. The PRISMA guidelines for Scoping Reviews were followed. RESULTS: Our literature search identified a total of 43 eligible articles, which were included in this scoping review. We identified 15 different biomarkers, each described by at least two studies, that were used to determine signs of cardiac remodelling in cardiovascular disease (CVD) and people with DM. NT-proBNP was identified as the most frequently employed biomarker in this context; however, we also identified emerging biomarkers including hs-CRP, hs-cTnT, and Galectin-3. CONCLUSION: There is a complex relationship between DM and cardiovascular health, where more research is needed. Current biomarkers reflective of adverse cardiac remodelling in DM are often used to diagnose other CVDs, such as NT-proBNP for heart failure. Hence there is a need for identification of specific biomarkers that can detect early signs of cardiac remodelling in the presence of DM. Further research into these biomarkers and mechanisms can deepen our understanding of their role in DM-associated CVD and lead to better preventative therapies.


Asunto(s)
Enfermedades Cardiovasculares , Diabetes Mellitus , Humanos , Pronóstico , Remodelación Ventricular , Biomarcadores
3.
J Mol Cell Cardiol ; 161: 106-115, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34371034

RESUMEN

Elevated intracellular Na (Nai) and metabolic impairment are interrelated pathophysiological features of the failing heart (HF). There have been a number of studies showing that myocardial sodium elevation subtly affects mitochondrial function. During contraction, mitochondrial calcium (Camito) stimulates a variety of TCA cycle enzymes, thereby providing reducing equivalents to maintain ATP supply. Nai elevation has been shown to impact Camito; however, whether metabolic remodelling in HF is caused by increased Nai has only been recently demonstrated. This novel insight may help to elucidate the contribution of metabolic remodelling in the pathophysiology of HF, the lack of efficacy of current HF therapies and a rationale for the development of future metabolism-targeting treatments. Here we review the relationship between Na pump inhibition, elevated Nai, and altered metabolic profile in the context of HF and their link to metabolic (in)flexibility and mitochondrial reprogramming.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Miocardio/metabolismo , Sodio/metabolismo , Animales , Compuestos Epoxi/farmacología , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/fisiopatología , Humanos
4.
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
5.
Biol Lett ; 15(11): 20190710, 2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31771414

RESUMEN

The African naked mole-rat (Heterocephalus glaber) is unique among mammals, displaying extreme longevity, resistance to cardiovascular disease and an ability to survive long periods of extreme hypoxia. The metabolic adaptations required for resistance to hypoxia are hotly debated and a recent report provides evidence that they are able to switch from glucose to fructose driven glycolysis in the brain. However, other systemic alterations in their metabolism are largely unknown. In the current study, a semi-targeted high resolution 1H magnetic resonance spectroscopy (MRS) metabolomics investigation was performed on cardiac tissue from the naked mole-rat (NMR) and wild-type C57/BL6 mice to better understand these adaptations. A range of metabolic differences was observed in the NMR including increased lactate, consistent with enhanced rates of glycolysis previously reported, increased glutathione, suggesting increased resistance to oxidative stress and decreased succinate/fumarate ratio suggesting reduced oxidative phosphorylation and ROS production. Surprisingly, the most significant difference was an elevation of glycogen stores and glucose-1-phosphate resulting from glycogen turnover, that were completely absent in the mouse heart and above the levels found in the mouse liver. Thus, we identified a range of metabolic adaptations in the NMR heart that are relevant to their ability to survive extreme environmental pressures and metabolic stress. Our study underscores the plasticity of energetic pathways and the need for compensatory strategies to adapt in response to the physiological and pathological stress including ageing and ischaemic heart pathologies.


Asunto(s)
Glucógeno , Ratas Topo , Adaptación Fisiológica , Animales , Longevidad , Metabolómica , Ratones
6.
Biochem Soc Trans ; 46(4): 817-827, 2018 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-29970448

RESUMEN

Alterations in excitation-contraction coupling and elevated intracellular sodium (Nai) are hallmarks of pathological cardiac remodelling that underline contractile dysfunction. In addition, changes in cardiac metabolism are observed in cardiac hypertrophy and heart failure (HF) that lead to a mismatch in ATP supply and demand, contributing to poor prognosis. A link between Nai and altered metabolism has been proposed but is not well understood. Many mitochondrial enzymes are stimulated by mitochondrial calcium (Camito) during contraction, thereby sustaining production of reducing equivalents to maintain ATP supply. This stimulation is thought to be perturbed when cytosolic Nai is high due to increased Camito efflux, potentially compromising ATPmito production and leading to metabolic dysregulation. Increased Nai has been previously shown to affect Camito; however, whether Nai elevation plays a causative role in energetic mismatching in the hypertrophied and failing heart remains unknown. In this review, we discuss the relationship between elevated Nai, NaK ATPase dysregulation and the metabolic phenotype in the contexts of pathological hypertrophy and HF and their link to metabolic flexibility, capacity (reserve) and efficiency that are governed by intracellular ion homeostasis. The development of non-invasive analytical techniques using nuclear magnetic resonance able to probe metabolism in situ in the functioning heart will enable a better understanding of the underlying mechanisms of Nai overload in cardiac pathophysiology. They will lead to novel insights that help to explain the metabolic contribution towards these diseases, the incomplete rescue observed with current therapies and a rationale for future energy-targeted therapies.


Asunto(s)
Cardiomegalia/metabolismo , Sodio/metabolismo , Remodelación Ventricular/fisiología , Adenosina Trifosfato/biosíntesis , Adenosina Trifosfato/metabolismo , Calcio/metabolismo , Cardiomegalia/complicaciones , Metabolismo Energético , Insuficiencia Cardíaca/metabolismo , Homeostasis , Humanos , Mitocondrias Cardíacas/enzimología , Mitocondrias Cardíacas/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
7.
FASEB J ; 30(8): 2684-97, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27103577

RESUMEN

The role of peroxisome proliferator-activated receptor α (PPARα)-mediated metabolic remodeling in cardiac adaptation to hypoxia has yet to be defined. Here, mice were housed in hypoxia for 3 wk before in vivo contractile function was measured using cine MRI. In isolated, perfused hearts, energetics were measured using (31)P magnetic resonance spectroscopy (MRS), and glycolysis and fatty acid oxidation were measured using [(3)H] labeling. Compared with a normoxic, chow-fed control mouse heart, hypoxia decreased PPARα expression, fatty acid oxidation, and mitochondrial uncoupling protein 3 (UCP3) levels, while increasing glycolysis, all of which served to maintain normal ATP concentrations ([ATP]) and thereby, ejection fractions. A high-fat diet increased cardiac PPARα expression, fatty acid oxidation, and UCP3 levels with decreased glycolysis. Hypoxia was unable to alter the high PPARα expression or reverse the metabolic changes caused by the high-fat diet, with the result that [ATP] and contractile function decreased significantly. The adaptive metabolic changes caused by hypoxia in control mouse hearts were found to have occurred already in PPARα-deficient (PPARα(-/-)) mouse hearts and sustained function in hypoxia despite an inability for further metabolic remodeling. We conclude that decreased cardiac PPARα expression is essential for adaptive metabolic remodeling in hypoxia, but is prevented by dietary fat.-Cole, M. A., Abd Jamil, A. H., Heather, L. C., Murray, A. J., Sutton, E. R., Slingo, M., Sebag-Montefiore, L., Tan, S. C., Aksentijevic, D., Gildea, O. S., Stuckey, D. J., Yeoh, K. K., Carr, C. A., Evans, R. D., Aasum, E., Schofield, C. J., Ratcliffe, P. J., Neubauer, S., Robbins, P. A., Clarke, K. On the pivotal role of PPARα in adaptation of the heart to hypoxia and why fat in the diet increases hypoxic injury.


Asunto(s)
Adaptación Fisiológica , Grasas de la Dieta/efectos adversos , Corazón/efectos de los fármacos , Miocardio/metabolismo , Consumo de Oxígeno/fisiología , PPAR alfa/metabolismo , Alimentación Animal/análisis , Animales , Línea Celular , Grasas de la Dieta/administración & dosificación , Regulación de la Expresión Génica/fisiología , Corazón/fisiología , Masculino , Ratones , Miocitos Cardíacos/metabolismo , PPAR alfa/genética
8.
J Physiol ; 594(2): 307-20, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26574233

RESUMEN

KEY POINTS: Adaptation to hypoxia makes the heart more oxygen efficient, by metabolising more glucose. In contrast, type 2 diabetes makes the heart metabolise more fatty acids. Diabetes increases the chances of the heart being exposed to hypoxia, but whether the diabetic heart can adapt and respond is unknown. In this study we show that diabetic hearts retain the ability to adapt their metabolism in response to hypoxia, with functional hypoxia signalling pathways. However, the hypoxia-induced changes in metabolism are additive to abnormal baseline metabolism, resulting in hypoxic diabetic hearts metabolising more fat and less glucose than controls. This stops the diabetic heart being able to recover its function when stressed. These results demonstrate that the diabetic heart retains metabolic flexibility to adapt to hypoxia, but is hindered by the baseline effects of the disease. This increases our understanding of how the diabetic heart is affected by hypoxia-associated complications of the disease. ABSTRACT: Hypoxia activates the hypoxia-inducible factor (HIF), promoting glycolysis and suppressing mitochondrial respiration. In the type 2 diabetic heart, glycolysis is suppressed whereas fatty acid metabolism is promoted. The diabetic heart experiences chronic hypoxia as a consequence of increased obstructive sleep apnoea and cardiovascular disease. Given the opposing metabolic effects of hypoxia and diabetes, we questioned whether diabetes affects cardiac metabolic adaptation to hypoxia. Control and type 2 diabetic rats were housed for 3 weeks in normoxia or 11% oxygen. Metabolism and function were measured in the isolated perfused heart using radiolabelled substrates. Following chronic hypoxia, both control and diabetic hearts upregulated glycolysis, lactate efflux and glycogen content and decreased fatty acid oxidation rates, with similar activation of HIF signalling pathways. However, hypoxia-induced changes were superimposed on diabetic hearts that were metabolically abnormal in normoxia, resulting in glycolytic rates 30% lower, and fatty acid oxidation 36% higher, in hypoxic diabetic hearts than hypoxic controls. Peroxisome proliferator-activated receptor α target proteins were suppressed by hypoxia, but activated by diabetes. Mitochondrial respiration in diabetic hearts was divergently activated following hypoxia compared with controls. These differences in metabolism were associated with decreased contractile recovery of the hypoxic diabetic heart following an acute hypoxic insult. In conclusion, type 2 diabetic hearts retain metabolic flexibility to adapt to hypoxia, with normal HIF signalling pathways. However, they are more dependent on oxidative metabolism following hypoxia due to abnormal normoxic metabolism, which was associated with a functional deficit in response to stress.


Asunto(s)
Adaptación Fisiológica , Diabetes Mellitus Tipo 2/metabolismo , Cardiomiopatías Diabéticas/metabolismo , Miocardio/metabolismo , Estrés Oxidativo , Oxígeno/metabolismo , Animales , Hipoxia de la Célula , Glucógeno/metabolismo , Glucólisis , Ácido Láctico/metabolismo , Masculino , Mitocondrias Musculares/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Ratas , Ratas Wistar , Transducción de Señal
9.
Exp Physiol ; 101(2): 282-94, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26585840

RESUMEN

NEW FINDINGS: What is the central question of this study? Rate-pressure product (RPP) is commonly used as an index of cardiac 'effort'. In canine and human hearts (which have a positive force-frequency relationship), RPP is linearly correlated with oxygen consumption and has therefore been widely adopted as a species-independent index of cardiac work. However, given that isolated rodent hearts demonstrate a negative force-frequency relationship, its use in this model requires validation. What is the main finding and its importance? Despite its widespread use, RPP is not correlated with oxygen consumption (or cardiac 'effort') in the Langendorff-perfused isolated rat heart. This lack of correlation was also evident when perfusions included a range of metabolic substrates, insulin or ß-adrenoceptor stimulation. Langendorff perfusion of hearts isolated from rats and mice has been used extensively for physiological, pharmacological and biochemical studies. The ability to phenotype these hearts reliably is, therefore, essential. One of the commonly used indices of function is rate-pressure product (RPP); a rather ill-defined index of 'work' or, more correctly, 'effort'. Rate-pressure product, as originally described in dog or human hearts, was shown to be correlated with myocardial oxygen consumption (MV̇O2). Despite its widespread use, the application of this index to rat or mouse hearts (which, unlike the dog or human, have a negative force-frequency relationship) has not been characterized. The aim of this study was to examine the relationship between RPP and MV̇O2 in Langendorff-perfused rat hearts. Paced hearts (300-750 beats min(-1)) were perfused either with Krebs-Henseleit (KH) buffer (11 mm glucose) or with buffer supplemented with metabolic substrates and insulin. The arteriovenous oxygen consumption (MV̇O2) was recorded. Metabolic status was assessed using (31) P magnetic resonance spectroscopy and lactate efflux. Experiments were repeated in the presence of isoprenaline and in unpaced hearts where heart rate was increased by cumulative isoprenaline challenge. In KH buffer-perfused hearts, MV̇O2 increased with increasing heart rate, but given that left ventricular developed pressure decreased with increases in rate, RPP was not correlated with MV̇O2, lactate production or phosphocreatine/ATP ratio. Although the provision of substrates or ß-adrenoceptor stimulation changed the shape of the RPP-MV̇O2 relationship, neither intervention resulted in a positive correlation between RPP and oxygen consumption. Rate-pressure product is therefore an unreliable index of oxygen consumption or 'cardiac effort' in the isolated rat heart.


Asunto(s)
Corazón/fisiología , Consumo de Oxígeno/fisiología , Oxígeno/metabolismo , Animales , Frecuencia Cardíaca/fisiología , Preparación de Corazón Aislado/métodos , Ácido Láctico/metabolismo , Masculino , Miocardio/metabolismo , Perfusión/métodos , Fosfocreatina/metabolismo , Presión , Ratas , Ratas Wistar , Receptores Adrenérgicos beta/metabolismo
10.
J Mol Cell Cardiol ; 86: 95-101, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26196304

RESUMEN

We investigate the potential of multiple quantum filtered (MQF) (23)Na NMR to probe intracellular [Na]i in the Langendorff perfused mouse heart. In the presence of Tm(DOTP) shift reagent the triple quantum filtered (TQF) signal originated largely from the intracellular sodium pool with a 32±6% contribution of the total TQF signal arising from extracellular sodium, whilst the rank 2 double-quantum filtered signal (DQF), acquired with a 54.7° flip-angle pulse, originated exclusively from the extracellular sodium pool. Given the different cellular origins of the (23)Na MQF signals we propose that the TQF/DQF ratio can be used as a semi-quantitative measure of [Na]i in the mouse heart. We demonstrate a good correlation of this ratio with [Na]i measured with shift reagent at baseline and under conditions of elevated [Na]i. We compare the measurements of [Na]i using both shift reagent and TQF/DQF ratio in a cohort of wild type mouse hearts and in a transgenic PLM(3SA) mouse expressing a non-phosphorylatable form of phospholemman, showing a modest but measurable elevation of baseline [Na]i. MQF filtered (23)Na NMR is a potentially useful tool for studying normal and pathophysiological changes in [Na]i, particularly in transgenic mouse models with altered Na regulation.


Asunto(s)
Corazón/fisiopatología , Preparación de Corazón Aislado , Miocardio/metabolismo , Animales , Corazón/diagnóstico por imagen , Imagen por Resonancia Magnética , Ratones , Radiografía , Sodio/metabolismo , Radioisótopos de Sodio/administración & dosificación , Radioisótopos de Sodio/metabolismo
11.
Circ Res ; 112(6): 945-55, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23325497

RESUMEN

RATIONALE: Creatine is thought to be involved in the spatial and temporal buffering of ATP in energetic organs such as heart and skeletal muscle. Creatine depletion affects force generation during maximal stimulation, while reduced levels of myocardial creatine are a hallmark of the failing heart, leading to the widely held view that creatine is important at high workloads and under conditions of pathological stress. OBJECTIVE: We therefore hypothesised that the consequences of creatine-deficiency in mice would be impaired running capacity, and exacerbation of heart failure following myocardial infarction. METHODS AND RESULTS: Surprisingly, mice with whole-body creatine deficiency due to knockout of the biosynthetic enzyme (guanidinoacetate N-methyltransferase [GAMT]) voluntarily ran just as fast and as far as controls (>10 km/night) and performed the same level of work when tested to exhaustion on a treadmill. Furthermore, survival following myocardial infarction was not altered, nor was subsequent left ventricular (LV) remodelling and development of chronic heart failure exacerbated, as measured by 3D-echocardiography and invasive hemodynamics. These findings could not be accounted for by compensatory adaptations, with no differences detected between WT and GAMT(-/-) proteomes. Alternative phosphotransfer mechanisms were explored; adenylate kinase activity was unaltered, and although GAMT(-/-) hearts accumulated the creatine precursor guanidinoacetate, this had negligible energy-transfer activity, while mitochondria retained near normal function. CONCLUSIONS: Creatine-deficient mice show unaltered maximal exercise capacity and response to chronic myocardial infarction, and no obvious metabolic adaptations. Our results question the paradigm that creatine is essential for high workload and chronic stress responses in heart and skeletal muscle.


Asunto(s)
Creatina/deficiencia , Tolerancia al Ejercicio/fisiología , Infarto del Miocardio/fisiopatología , Esfuerzo Físico/fisiología , Adenilato Quinasa/metabolismo , Animales , Femenino , Glicina/análogos & derivados , Glicina/metabolismo , Guanidinoacetato N-Metiltransferasa/genética , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Cardíacas/fisiología , Infarto del Miocardio/complicaciones , Infarto del Miocardio/metabolismo , Consumo de Oxígeno/fisiología , Condicionamiento Físico Animal , Remodelación Ventricular/fisiología
12.
J Mol Cell Cardiol ; 75: 76-87, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25066696

RESUMEN

UNLABELLED: Inhibition of malonyl-coenzyme A decarboxylase (MCD) shifts metabolism from fatty acid towards glucose oxidation, which has therapeutic potential for obesity and myocardial ischemic injury. However, ~40% of patients with MCD deficiency are diagnosed with cardiomyopathy during infancy. AIM: To clarify the link between MCD deficiency and cardiac dysfunction in early life and to determine the contributing systemic and cardiac metabolic perturbations. METHODS AND RESULTS: MCD knockout mice ((-/-)) exhibited non-Mendelian genotype ratios (31% fewer MCD(-/-)) with deaths clustered around weaning. Immediately prior to weaning (18days) MCD(-/-) mice had lower body weights, elevated body fat, hepatic steatosis and glycogen depletion compared to wild-type littermates. MCD(-/-) plasma was hyperketonemic, hyperlipidemic, had 60% lower lactate levels and markers of cellular damage were elevated. MCD(-/-) hearts exhibited hypertrophy, impaired ejection fraction and were energetically compromised (32% lower total adenine nucleotide pool). However differences between WT and MCD(-/-) converged with age, suggesting that, in surviving MCD(-/-) mice, early cardiac dysfunction resolves over time. These observations were corroborated by in silico modelling of cardiomyocyte metabolism, which indicated improvement of the MCD(-/-) metabolic phenotype and improved cardiac efficiency when switched from a high-fat diet (representative of suckling) to a standard post-weaning diet, independent of any developmental changes. CONCLUSIONS: MCD(-/-) mice consistently exhibited cardiac dysfunction and severe metabolic perturbations while on a high-fat, low carbohydrate diet of maternal milk and these gradually resolved post-weaning. This suggests that dysfunction is a common feature of MCD deficiency during early development, but that severity is dependent on composition of dietary substrates.


Asunto(s)
Carboxiliasas/deficiencia , Corazón/fisiopatología , Destete , Envejecimiento/patología , Animales , Simulación por Computador , Dieta Alta en Grasa , Femenino , Eliminación de Gen , Genotipo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , Fenotipo , Especificidad por Sustrato , Análisis de Supervivencia
13.
Nat Commun ; 15(1): 2204, 2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38538579

RESUMEN

The naked mole-rat Heterocephalus glaber is a eusocial mammal exhibiting extreme longevity (37-year lifespan), extraordinary resistance to hypoxia and absence of cardiovascular disease. To identify the mechanisms behind these exceptional traits, metabolomics and RNAseq of cardiac tissue from naked mole-rats was compared to other African mole-rat genera (Cape, Cape dune, Common, Natal, Mahali, Highveld and Damaraland mole-rats) and evolutionarily divergent mammals (Hottentot golden mole and C57/BL6 mouse). We identify metabolic and genetic adaptations unique to naked mole-rats including elevated glycogen, thus enabling glycolytic ATP generation during cardiac ischemia. Elevated normoxic expression of HIF-1α is observed while downstream hypoxia responsive-genes are down-regulated, suggesting adaptation to low oxygen environments. Naked mole-rat hearts show reduced succinate levels during ischemia compared to C57/BL6 mouse and negligible tissue damage following ischemia-reperfusion injury. These evolutionary traits reflect adaptation to a unique hypoxic and eusocial lifestyle that collectively may contribute to their longevity and health span.


Asunto(s)
Longevidad , Oxígeno , Animales , Ratones , Longevidad/genética , Hipoxia/genética , Ratas Topo/genética , Isquemia
14.
Am J Physiol Heart Circ Physiol ; 305(4): H506-20, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23792673

RESUMEN

Disruption of the creatine kinase (CK) system in hearts of CK-deficient mice leads to changes in the ultrastructure and regulation of mitochondrial respiration. We expected to see similar changes in creatine-deficient mice, which lack the enzyme guanidinoacetate methyltransferase (GAMT) to produce creatine. The aim of this study was to characterize the changes in cardiomyocyte mitochondrial organization, regulation of respiration, and intracellular compartmentation associated with GAMT deficiency. Three-dimensional mitochondrial organization was assessed by confocal microscopy. On populations of permeabilized cardiomyocytes, we recorded ADP and ATP kinetics of respiration, competition between mitochondria and pyruvate kinase for ADP produced by ATPases, ADP kinetics of endogenous pyruvate kinase, and ATP kinetics of ATPases. These data were analyzed by mathematical models to estimate intracellular compartmentation. Quantitative analysis of morphological and kinetic data as well as derived model fits showed no difference between GAMT-deficient and wild-type mice. We conclude that inactivation of the CK system by GAMT deficiency does not alter mitochondrial organization and intracellular compartmentation in relaxed cardiomyocytes. Thus, our results suggest that the healthy heart is able to preserve cardiac function at a basal level in the absence of CK-facilitated energy transfer without compromising intracellular organization and the regulation of mitochondrial energy homeostasis. This raises questions on the importance of the CK system as a spatial energy buffer in unstressed cardiomyocytes.


Asunto(s)
Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Creatina/deficiencia , Metabolismo Energético , Guanidinoacetato N-Metiltransferasa/deficiencia , Trastornos del Desarrollo del Lenguaje/enzimología , Mitocondrias Cardíacas/enzimología , Trastornos del Movimiento/congénito , Miocitos Cardíacos/enzimología , Adenosina Trifosfatasas/metabolismo , Animales , Creatina Quinasa/metabolismo , Modelos Animales de Enfermedad , Femenino , Genotipo , Guanidinoacetato N-Metiltransferasa/genética , Homeostasis , Cinética , Trastornos del Desarrollo del Lenguaje/genética , Trastornos del Desarrollo del Lenguaje/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Microscopía Fluorescente , Mitocondrias Cardíacas/patología , Modelos Cardiovasculares , Trastornos del Movimiento/enzimología , Trastornos del Movimiento/genética , Trastornos del Movimiento/patología , Miocitos Cardíacos/patología , Fenotipo , Piruvato Quinasa/metabolismo
15.
J Infect Public Health ; 16(8): 1322-1331, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37343504

RESUMEN

BACKGROUND: Research concerned with attitudes towards COVID-19 vaccination in upper middle-income countries such as Bosnia and Herzegovina (B&H) is scarce. Currently, B&H has the lowest number of fully vaccinated adults in Europe, and the highest cumulative number of COVID-19 deaths and SARS-CoV-2 infected individuals. The aim of our study was to examine the factors associated with COVID-19 vaccination status in B&H. METHODS: An online survey among 1304 B&H adults was conducted in October 2021 evaluating vaccine acceptance, together with socio-demographic variables, attitudes and beliefs related to COVID-19 vaccination. RESULTS: The results from a binary logistic regression indicate that those who believed that the COVID-19 vaccine was effective were 45 times more likely to be vaccinated compared to those who did not. We also show that those who had received childhood immunisations were 41 times more likely to be vaccinated against COVID-19 compared to those who had never been previously immunised. Other significant factors were related to respondents' trust in government institutions and healthcare policymakers as well as trust in public healthcare workers. CONCLUSION: We suggest that future vaccination campaigns should be aimed at educating the public regarding the importance and safety of vaccines, together with strengthening trust in the public health system.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Adulto , Humanos , Niño , COVID-19/prevención & control , SARS-CoV-2 , Vacunación , Inmunización
16.
Basic Res Cardiol ; 107(5): 276, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22760499

RESUMEN

The creatine kinase (CK) energy transport and buffering system supports cardiac function at times of high demand and is impaired in the failing heart. Mice deficient in muscle- and mitochondrial-CK (M/Mt-CK(-/-)) have previously been described, but exhibit an unexpectedly mild phenotype of compensated left ventricular (LV) hypertrophy. We hypothesised that heart failure would develop with age and performed echocardiography and LV haemodynamics at 1 year. Since all previous studies have utilised mice with a mixed genetic background, we backcrossed for >10 generations on to C57BL/6, and repeated the in vivo investigations. Male M/Mt-CK(-/-) mice on the mixed genetic background developed congestive heart failure as evidenced by significantly elevated end-diastolic pressure, impaired contractility, LV dilatation, hypertrophy and pulmonary congestion. Female mice were less severely affected, only showing trends for these parameters. After backcrossing, M/Mt-CK(-/-) mice had LV dysfunction consisting of impaired isovolumetric pressure changes and reduced contractile reserve, but did not develop congestive heart failure. Body weight was lower in knockout mice as a consequence of reduced total body fat. LV weight was not significantly elevated in relation to other internal organs and gene expression of LVH markers was normal, suggesting an absence of hypertrophy. In conclusion, the consequences of CK deficiency are highly dependent on genetic modifiers, gender and age. However, the observation that a primary defect in CK can, under the right conditions, result in heart failure suggests that impaired CK activity in the failing heart could contribute to disease progression.


Asunto(s)
Creatina Quinasa/deficiencia , Insuficiencia Cardíaca/etiología , Factores de Edad , Animales , Composición Corporal , Enfermedad Crónica , Femenino , Insuficiencia Cardíaca/genética , Hemodinámica , Hipertrofia Ventricular Izquierda/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Factores Sexuales
17.
Nat Rev Nephrol ; 18(8): 524-537, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35637381

RESUMEN

Chronic kidney disease (CKD) affects millions of people globally and, for most patients, the risk of developing cardiovascular disease is higher than that of progression to kidney failure. Moreover, mortality owing to cardiovascular complications in patients with CKD is markedly higher than in matched individuals from the general population. This mortality was traditionally thought to be driven by coronary heart disease but >75% of patients with CKD have left ventricular hypertrophy, which contributes to mortality, particularly sudden cardiac death. The aetiology of cardiac complications in CKD is multifactorial. In addition to haemodynamic overload, uraemic toxin accumulation and altered ion homeostasis, which are known to underlie left ventricular hypertrophy in CKD and drive cardiac dysfunction, we examine the role of myocardial metabolic remodelling in CKD. Uraemic cardiomyopathy is characterized by myriad cardiac metabolic maladaptations, including altered mitochondrial function, changes in myocardial substrate utilization, altered metabolic transporter function and expression, and impaired insulin response and phosphoinositide-3 kinase-AKT signalling, which collectively lead to impaired cardiac energetics. Interestingly, none of the standard treatments used to treat CKD target the metabolism of the uraemic heart directly. An improved understanding of the cardiac metabolic perturbations that occur in CKD might allow the development of novel treatments for uraemic cardiomyopathy.


Asunto(s)
Cardiomiopatías , Insuficiencia Renal Crónica , Cardiomiopatías/etiología , Corazón , Humanos , Hipertrofia Ventricular Izquierda/complicaciones , Miocardio , Insuficiencia Renal Crónica/complicaciones
18.
Front Cardiovasc Med ; 9: 945726, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36312266

RESUMEN

Cardiovascular disease (CVD) is the principal cause of mortality and morbidity globally. With the pressures for improved care and translation of the latest medical advances and knowledge to an actionable plan, clinical decision-making for cardiologists is challenging. Artificial Intelligence (AI) is a field in computer science that studies the design of intelligent agents which take the best feasible action in a situation. It incorporates the use of computational algorithms which simulate and perform tasks that traditionally require human intelligence such as problem solving and learning. Whilst medicine is arguably the last to apply AI in its everyday routine, cardiology is at the forefront of AI revolution in the medical field. The development of AI methods for accurate prediction of CVD outcomes, non-invasive diagnosis of coronary artery disease (CAD), detection of malignant arrythmias through wearables, and diagnosis, treatment strategies and prediction of outcomes for heart failure (HF) patients, demonstrates the potential of AI in future cardiology. With the advancements of AI, Internet of Things (IoT) and the promotion of precision medicine, the future of cardiology will be heavily based on these innovative digital technologies. Despite this, ethical dilemmas regarding the implementation of AI technologies in real-world are still unaddressed.

19.
Sci Rep ; 12(1): 1111, 2022 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-35064159

RESUMEN

Preeclampsia is a cardiovascular pregnancy complication characterised by new onset hypertension and organ damage or intrauterine growth restriction. It is one of the leading causes of maternal and fetal mortality in pregnancy globally. Short of pre-term delivery of the fetus and placenta, treatment options are limited. Consequently, preeclampsia leads to increased cardiovascular disease risk in both mothers and offspring later in life. Here we aim to examine the impact of the reduced uterine perfusion pressure (RUPP) rat model of preeclampsia on the maternal cardiovascular system, placental and fetal heart metabolism. The surgical RUPP model was induced in pregnant rats by applying silver clips around the aorta and uterine arteries on gestational day 14, resulting in ~ 40% uterine blood flow reduction. The experiment was terminated on gestational day 19 and metabolomic profile of placentae, maternal and fetal hearts analysed using high-resolution 1H NMR spectroscopy. Impairment of uterine perfusion in RUPP rats caused placental and cardiac hypoxia and a series of metabolic adaptations: altered energetics, carbohydrate, lipid and amino acid metabolism of placentae and maternal hearts. Comparatively, the fetal metabolic phenotype was mildly affected. Nevertheless, long-term effects of these changes in both mothers and the offspring should be investigated further in the future.


Asunto(s)
Hipoxia/metabolismo , Placenta/metabolismo , Preeclampsia/metabolismo , Útero/irrigación sanguínea , Animales , Presión Sanguínea/fisiología , Simulación por Computador , Modelos Animales de Enfermedad , Femenino , Corazón Fetal/metabolismo , Humanos , Hipoxia/fisiopatología , Metabolómica , Modelos Biológicos , Placenta/irrigación sanguínea , Circulación Placentaria/fisiología , Preeclampsia/fisiopatología , Embarazo , Espectroscopía de Protones por Resonancia Magnética , Ratas , Útero/fisiología
20.
JCI Insight ; 7(10)2022 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-35472029

RESUMEN

Voltage-gated hydrogen channel 1 (Hvcn1) is a voltage-gated proton channel, which reduces cytosol acidification and facilitates the production of ROS. The increased expression of this channel in some cancers has led to proposing Hvcn1 antagonists as potential therapeutics. While its role in most leukocytes has been studied in depth, the function of Hvcn1 in T cells remains poorly defined. We show that Hvcn1 plays a nonredundant role in protecting naive T cells from intracellular acidification during priming. Despite sharing overall functional impairment in vivo and in vitro, Hvcn1-deficient CD4+ and CD8+ T cells display profound differences during the transition from naive to primed T cells, including in the preservation of T cell receptor (TCR) signaling, cellular division, and death. These selective features result, at least in part, from a substantially different metabolic response to intracellular acidification associated with priming. While Hvcn1-deficient naive CD4+ T cells reprogram to rescue the glycolytic pathway, naive CD8+ T cells, which express high levels of this channel in the mitochondria, respond by metabolically compensating mitochondrial dysfunction, at least in part via AMPK activation. These observations imply heterogeneity between adaptation of naive CD4+ and CD8+ T cells to intracellular acidification during activation.


Asunto(s)
Hidrógeno , Protones , Concentración de Iones de Hidrógeno , Recuento de Linfocitos , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA