Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.953
Filtrar
1.
PeerJ ; 12: e17333, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38948204

RESUMO

Acute heart attack is the primary cause of cardiovascular-related death worldwide. A common treatment is reperfusion of ischemic tissue, which can cause irreversible damage to the myocardium. The number of mitochondria in cardiomyocytes is large, which generate adenosine triphosphate (ATP) to sustain proper cardiac contractile function, and mitochondrial dysfunction plays a crucial role in cell death during myocardial ischemia-reperfusion, leading to an increasing number of studies investigating the impact of mitochondria on ischemia-reperfusion injury. The disarray of mitochondrial dynamics, excessive Ca2+ accumulation, activation of mitochondrial permeable transition pores, swelling of mitochondria, ultimately the death of cardiomyocyte are the consequences of ischemia-reperfusion injury. κ-opioid receptors can alleviate mitochondrial dysfunction, regulate mitochondrial dynamics, mitigate myocardial ischemia-reperfusion injury, exert protective effects on myocardium. The mechanism of κ-OR activation during myocardial ischemia-reperfusion to regulate mitochondrial dynamics and reduce myocardial ischemia-reperfusion injury will be discussed, so as to provide theoretical basis for the protection of ischemic myocardium.


Assuntos
Traumatismo por Reperfusão Miocárdica , Miócitos Cardíacos , Receptores Opioides kappa , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Receptores Opioides kappa/metabolismo , Humanos , Animais , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/patologia , Dinâmica Mitocondrial/fisiologia , Cálcio/metabolismo
2.
Circ Res ; 135(2): 372-396, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38963864

RESUMO

Despite clinical and scientific advancements, heart failure is the major cause of morbidity and mortality worldwide. Both mitochondrial dysfunction and inflammation contribute to the development and progression of heart failure. Although inflammation is crucial to reparative healing following acute cardiomyocyte injury, chronic inflammation damages the heart, impairs function, and decreases cardiac output. Mitochondria, which comprise one third of cardiomyocyte volume, may prove a potential therapeutic target for heart failure. Known primarily for energy production, mitochondria are also involved in other processes including calcium homeostasis and the regulation of cellular apoptosis. Mitochondrial function is closely related to morphology, which alters through mitochondrial dynamics, thus ensuring that the energy needs of the cell are met. However, in heart failure, changes in substrate use lead to mitochondrial dysfunction and impaired myocyte function. This review discusses mitochondrial and cristae dynamics, including the role of the mitochondria contact site and cristae organizing system complex in mitochondrial ultrastructure changes. Additionally, this review covers the role of mitochondria-endoplasmic reticulum contact sites, mitochondrial communication via nanotunnels, and altered metabolite production during heart failure. We highlight these often-neglected factors and promising clinical mitochondrial targets for heart failure.


Assuntos
Insuficiência Cardíaca , Mitocôndrias Cardíacas , Humanos , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/patologia , Animais , Dinâmica Mitocondrial , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Metabolismo Energético , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/patologia
3.
BMC Cardiovasc Disord ; 24(1): 350, 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38987722

RESUMO

BACKGROUND: Antineoplastic medications, including doxorubicin, idarubicin, and epirubicin, have been found to adversely affect the heart due to oxidative stress - mitochondrial dysfunction - ferroptosis (ORMFs), which act as contributing attributes to anthracycline-induced cardiotoxicity. To better understand this phenomenon, the time-resolved measurements of ORMFS genes were analyzed in this study. METHODS: The effect of three anthracycline drugs on ORMFs genes was studied using a human 3D cardiac microtissue cell model. Transcriptome data was collected over 14 days at two doses (therapeutic and toxic). WGCNA identified key module-related genes, and functional enrichment analysis investigated the biological processes quantified by ssGSEA, such as immune cell infiltration and angiogenesis. Biopsies were collected from heart failure patients and control subjects. GSE59672 and GSE2965 were collected for validation. Molecular docking was used to identify anthracyclines's interaction with key genes. RESULTS: The ORMFs genes were screened in vivo or in vitro. Using WGCNA, six co-expressed gene modules were grouped, with MEblue emerging as the most significant module. Eight key genes intersecting the blue module with the dynamic response genes were obtained: CD36, CDH5, CHI3L1, HBA2, HSD11B1, OGN, RPL8, and VWF. Compared with control samples, all key genes except RPL8 were down-regulated in vitro ANT treatment settings, and their expression levels varied over time. According to functional analyses, the key module-related genes were engaged in angiogenesis and the immune system pathways. In all ANT-treated settings, ssGSEA demonstrated a significant down-regulation of angiogenesis score and immune cell activity, including Activated CD4 T cell, Immature B cell, Memory B cell, Natural killer cell, Type 1 T helper cell, and Type 2 T helper cell. Molecular docking revealed that RPL8 and CHI3L1 show significant binding affinity for anthracyclines. CONCLUSION: This study focuses on the dynamic characteristics of ORMFs genes in both human cardiac microtissues and cardiac biopsies from ANT-treated patients. It has been highlighted that ORMFs genes may contribute to immune infiltration and angiogenesis in cases of anthracycline-induced cardiotoxicity. A thorough understanding of these genes could potentially lead to improved diagnosis and treatment of the disease.


Assuntos
Cardiotoxicidade , Ferroptose , Simulação de Acoplamento Molecular , Estresse Oxidativo , Humanos , Estresse Oxidativo/efeitos dos fármacos , Ferroptose/efeitos dos fármacos , Ferroptose/genética , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/patologia , Mitocôndrias Cardíacas/genética , Redes Reguladoras de Genes , Fatores de Tempo , Transcriptoma , Epirubicina/efeitos adversos , Doxorrubicina , Antibióticos Antineoplásicos/efeitos adversos , Estudos de Casos e Controles , Idarubicina , Insuficiência Cardíaca/induzido quimicamente , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Perfilação da Expressão Gênica , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Estudos Longitudinais , Antraciclinas/efeitos adversos , Regulação da Expressão Gênica , Transdução de Sinais
4.
Bull Exp Biol Med ; 177(1): 22-25, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38954297

RESUMO

We studied the respiratory activity of mitochondria in peripheral blood leukocytes from 36 patients with coronary heart disease (CHD) and a history of ventricular tachyarrhythmias required cardioverter-defibrillator implantation. The measurements were carried out in incubation buffers with different oxidation substrates (succinate and pyruvate-malate mixture). In pyruvate-malate incubation buffer, oxygen consumption rate and respiratory control coefficients in patients with triggered device did not differ significantly from those in patients without cardioverter-defibrillator triggering. At the same time, respiratory control coefficients were below the reference values. In succinate buffer, values of mitochondrial parameters were significantly lower in patients with triggered devices. Our findings indicate that mitochondria of patients with non-triggered cardioverters-defibrillators have better functional and metabolic plasticity. It was concluded that activity of respiratory processes in mitochondria could be an indicator that should be taken into the account when assessing the risk of developing ventricular tachyarrhythmias.


Assuntos
Doença das Coronárias , Desfibriladores Implantáveis , Consumo de Oxigênio , Humanos , Masculino , Pessoa de Meia-Idade , Doença das Coronárias/fisiopatologia , Doença das Coronárias/terapia , Consumo de Oxigênio/fisiologia , Feminino , Mitocôndrias/metabolismo , Idoso , Taquicardia Ventricular/fisiopatologia , Taquicardia Ventricular/terapia , Ácido Pirúvico/metabolismo , Ácido Succínico/metabolismo , Malatos/metabolismo , Mitocôndrias Cardíacas/metabolismo
5.
Cardiovasc Diabetol ; 23(1): 261, 2024 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-39026280

RESUMO

Mitochondria play a central role in cellular energy metabolism, and their dysfunction is increasingly recognized as a critical factor in the pathogenesis of diabetes-related cardiac pathophysiology, including vulnerability to ischemic events that culminate in myocardial infarction on the one hand and ventricular arrhythmias on the other. In diabetes, hyperglycemia and altered metabolic substrates lead to excessive production of reactive oxygen species (ROS) by mitochondria, initiating a cascade of oxidative stress that damages mitochondrial DNA, proteins, and lipids. This mitochondrial injury compromises the efficiency of oxidative phosphorylation, leading to impaired ATP production. The resulting energy deficit and oxidative damage contribute to functional abnormalities in cardiac cells, placing the heart at an increased risk of electromechanical dysfunction and irreversible cell death in response to ischemic insults. While cardiac mitochondria are often considered to be relatively autonomous entities in their capacity to produce energy and ROS, their highly dynamic nature within an elaborate network of closely-coupled organelles that occupies 30-40% of the cardiomyocyte volume is fundamental to their ability to exert intricate regulation over global cardiac function. In this article, we review evidence linking the dynamic properties of the mitochondrial network to overall cardiac function and its response to injury. We then highlight select studies linking mitochondrial ultrastructural alterations driven by changes in mitochondrial fission, fusion and mitophagy in promoting cardiac ischemic injury to the diabetic heart.


Assuntos
Cardiomiopatias Diabéticas , Metabolismo Energético , Mitocôndrias Cardíacas , Isquemia Miocárdica , Estresse Oxidativo , Humanos , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/patologia , Animais , Cardiomiopatias Diabéticas/metabolismo , Cardiomiopatias Diabéticas/fisiopatologia , Cardiomiopatias Diabéticas/patologia , Cardiomiopatias Diabéticas/etiologia , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/fisiopatologia , Isquemia Miocárdica/patologia , Dinâmica Mitocondrial , Mitofagia , Espécies Reativas de Oxigênio/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Transdução de Sinais
7.
Vascul Pharmacol ; 155: 107324, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38985581

RESUMO

Doxorubicin (DOX) is a highly effective chemotherapeutic agent whose clinical use is hindered by the onset of cardiotoxic effects, resulting in reduced ejection fraction within the first year from treatment initiation. Recently it has been demonstrated that DOX accumulates within mitochondria, leading to disruption of metabolic processes and energetic imbalance. We previously described that phosphoinositide 3-kinase γ (PI3Kγ) contributes to DOX-induced cardiotoxicity, causing autophagy inhibition and accumulation of damaged mitochondria. Here we intend to describe the maladaptive metabolic rewiring occurring in DOX-treated hearts and the contribution of PI3Kγ signalling to this process. Metabolomic analysis of DOX-treated WT hearts revealed an accumulation of TCA cycle metabolites due to a cycle slowdown, with reduced levels of pyruvate, unchanged abundance of lactate and increased Acetyl-CoA production. Moreover, the activity of glycolytic enzymes was upregulated, and fatty acid oxidation downregulated, after DOX, indicative of increased glucose oxidation. In agreement, oxygen consumption was increased in after pyruvate supplementation, with the formation of cytotoxic ROS rather than energy production. These metabolic changes were fully prevented in KD hearts. Interestingly, they failed to increase glucose oxidation in response to DOX even with autophagy inhibition, indicating that PI3Kγ likely controls the fuel preference after DOX through an autophagy-independent mechanism. In vitro experiments showed that inhibition of PI3Kγ inhibits pyruvate dehydrogenase (PDH), the key enzyme of Randle cycle regulating the switch from fatty acids to glucose usage, while decreasing DOX-induced mobilization of GLUT-4-carrying vesicles to the plasma membrane and limiting the ensuing glucose uptake. These results demonstrate that PI3Kγ promotes a maladaptive metabolic rewiring in DOX-treated hearts, through a two-pronged mechanism controlling PDH activation and GLUT-4-mediated glucose uptake.


Assuntos
Cardiotoxicidade , Doxorrubicina , Metabolismo Energético , Ácidos Graxos , Glucose , Oxirredução , Animais , Doxorrubicina/toxicidade , Glucose/metabolismo , Ácidos Graxos/metabolismo , Metabolismo Energético/efeitos dos fármacos , Classe Ib de Fosfatidilinositol 3-Quinase/metabolismo , Glicólise/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Masculino , Transdução de Sinais/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Ciclo do Ácido Cítrico/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Cardiopatias/induzido quimicamente , Cardiopatias/metabolismo , Cardiopatias/patologia , Cardiopatias/prevenção & controle , Cardiopatias/fisiopatologia , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/patologia , Mitocôndrias Cardíacas/enzimologia , Camundongos Knockout , Modelos Animais de Doenças , Espécies Reativas de Oxigênio/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Antibióticos Antineoplásicos/toxicidade , Antibióticos Antineoplásicos/efeitos adversos
14.
Cardiovasc Diabetol ; 23(1): 239, 2024 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-38978010

RESUMO

BACKGROUND: Type 2 diabetes (T2D) is a frequent comorbidity encountered in patients with severe aortic stenosis (AS), leading to an adverse left ventricular (LV) remodeling and dysfunction. Metabolic alterations have been suggested as contributors of the deleterious effect of T2D on LV remodeling and function in patients with severe AS, but so far, the underlying mechanisms remain unclear. Mitochondria play a central role in the regulation of cardiac energy metabolism. OBJECTIVES: We aimed to explore the mitochondrial alterations associated with the deleterious effect of T2D on LV remodeling and function in patients with AS, preserved ejection fraction, and no additional heart disease. METHODS: We combined an in-depth clinical, biological and echocardiography phenotype of patients with severe AS, with (n = 34) or without (n = 50) T2D, referred for a valve replacement, with transcriptomic and histological analyses of an intra-operative myocardial LV biopsy. RESULTS: T2D patients had similar AS severity but displayed worse cardiac remodeling, systolic and diastolic function than non-diabetics. RNAseq analysis identified 1029 significantly differentially expressed genes. Functional enrichment analysis revealed several T2D-specific upregulated pathways despite comorbidity adjustment, gathering regulation of inflammation, extracellular matrix organization, endothelial function/angiogenesis, and adaptation to cardiac hypertrophy. Downregulated gene sets independently associated with T2D were related to mitochondrial respiratory chain organization/function and mitochondrial organization. Generation of causal networks suggested a reduced Ca2+ signaling up to the mitochondria, with the measured gene remodeling of the mitochondrial Ca2+ uniporter in favor of enhanced uptake. Histological analyses supported a greater cardiomyocyte hypertrophy and a decreased proximity between the mitochondrial VDAC porin and the reticular IP3-receptor in T2D. CONCLUSIONS: Our data support a crucial role for mitochondrial Ca2+ signaling in T2D-induced cardiac dysfunction in severe AS patients, from a structural reticulum-mitochondria Ca2+ uncoupling to a mitochondrial gene remodeling. Thus, our findings open a new therapeutic avenue to be tested in animal models and further human cardiac biopsies in order to propose new treatments for T2D patients suffering from AS. TRIAL REGISTRATION: URL: https://www. CLINICALTRIALS: gov ; Unique Identifier: NCT01862237.


Assuntos
Estenose da Valva Aórtica , Sinalização do Cálcio , Diabetes Mellitus Tipo 2 , Perfilação da Expressão Gênica , Mitocôndrias Cardíacas , Índice de Gravidade de Doença , Transcriptoma , Função Ventricular Esquerda , Remodelação Ventricular , Humanos , Estenose da Valva Aórtica/metabolismo , Estenose da Valva Aórtica/genética , Estenose da Valva Aórtica/fisiopatologia , Estenose da Valva Aórtica/diagnóstico por imagem , Estenose da Valva Aórtica/cirurgia , Estenose da Valva Aórtica/patologia , Masculino , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/patologia , Feminino , Idoso , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/complicações , Pessoa de Meia-Idade , Idoso de 80 Anos ou mais , Disfunção Ventricular Esquerda/fisiopatologia , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/metabolismo , Disfunção Ventricular Esquerda/diagnóstico por imagem
15.
Life Sci ; 351: 122760, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-38823506

RESUMO

Photobiomodulation (PBM) represents a promising and powerful approach for non-invasive therapeutic interventions. This emerging field of research has gained a considerable attention due to its potential for multiple disciplines, including medicine, neuroscience, and sports medicine. While PBM has shown the ability to stimulate various cellular processes in numerous medical applications, the fine-tuning of treatment parameters, such as wavelength, irradiance, treatment duration, and illumination geometry, remains an ongoing challenge. Furthermore, additional research is necessary to unveil the specific mechanisms of action and establish standardized protocols for diverse clinical applications. Given the widely accepted understanding that mitochondria play a pivotal role in the PBM mechanisms, our study delves into a multitude of PBM illumination parameters while assessing the PBM's effects on the basis of endpoints reflecting the mitochondrial metabolism of human cardiac myocytes (HCM), that are known for their high mitochondrial density. These endpoints include: i) the endogenous production of protoporphyrin IX (PpIX), ii) changes in mitochondrial potential monitored by Rhodamine 123 (Rhod 123), iii) changes in the HCM's oxygen consumption, iv) the fluorescence lifetime of Rhod 123 in mitochondria, and v) alterations of the mitochondrial morphology. The good correlation observed between these different methods to assess PBM effects underscores that monitoring the endogenous PpIX production offers interesting indirect insights into the mitochondrial metabolic activity. This conclusion is important since many approved therapeutics and cancer detection approaches are based on the use of PpIX. Finally, this correlation strongly suggests that the PBM effects mentioned above have a common "fundamental" mechanistic origin.


Assuntos
Terapia com Luz de Baixa Intensidade , Miócitos Cardíacos , Humanos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/efeitos da radiação , Terapia com Luz de Baixa Intensidade/métodos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos da radiação , Consumo de Oxigênio/efeitos da radiação , Protoporfirinas/metabolismo , Células Cultivadas , Potencial da Membrana Mitocondrial/efeitos da radiação , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/efeitos da radiação
16.
Eur J Pharmacol ; 977: 176736, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-38878877

RESUMO

Mitochondrial dysfunction and the activation of multiple programmed cell death (PCD) have been shown to aggravate the severity and mortality associated with the progression of myocardial infarction (MI). Although pharmacological modulation of mitochondrial dynamics, including treatment with the fusion promoter (M1) and the fission inhibitor (Mdivi-1), exerted cardioprotection against several cardiac complications, their roles in the post-MI model have never been investigated. Using a MI rat model instigated by permanent left-anterior descending (LAD) coronary artery occlusion, post-MI rats were randomly assigned to receive one of 4 treatments (n = 10/group): vehicle (DMSO 3%V/V), enalapril (10 mg/kg), Mdivi-1 (1.2 mg/kg) and M1 (2 mg/kg), while a control group of sham operated rats underwent surgery without LAD occlusion (n = 10). After 32-day treatment, cardiac and mitochondrial function, and histopathological morphology were investigated and molecular analysis was performed. Treatment with enalapril, Mdivi-1, and M1 significantly mitigated cardiac pathological remodeling, reduced myocardial injury, and improved left ventricular (LV) function in post-MI rats. Importantly, all interventions also attenuated mitochondrial dynamic imbalance and mitigated activation of apoptosis, necroptosis, and pyroptosis after MI. This investigation demonstrated for the first time that chronic mitochondrial dynamic-targeted therapy mitigated mitochondrial dysfunction and activation of PCD, leading to improved LV function in post-MI rats.


Assuntos
Apoptose , Enalapril , Dinâmica Mitocondrial , Infarto do Miocárdio , Disfunção Ventricular Esquerda , Animais , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Dinâmica Mitocondrial/efeitos dos fármacos , Masculino , Disfunção Ventricular Esquerda/tratamento farmacológico , Disfunção Ventricular Esquerda/fisiopatologia , Ratos , Enalapril/farmacologia , Enalapril/uso terapêutico , Apoptose/efeitos dos fármacos , Ratos Sprague-Dawley , Quinazolinonas/farmacologia , Quinazolinonas/uso terapêutico , Modelos Animais de Doenças , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/patologia
17.
Biomed Khim ; 70(3): 135-144, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38940202

RESUMO

Type 1 diabetes mellitus (T1DM) is the most severe form of diabetes, which is characterized by absolute insulin deficiency induced by the destruction of pancreatic beta cells. The aim of this study was to evaluate the effect of a structural analogue of apelin-12 ((NαMe)Arg-Pro-Arg-Leu-Ser-His-Lys-Gly-Pro-Nle-Pro-Phe-OH, metilin) on hyperglycemia, mitochondrial (MCh) respiration in permeabilized cardiac left ventricular (LV) fibers, the myocardial energy state, and cardiomyocyte membranes damage in a model of streptozotocin (STZ) diabetes in rats. Metilin was prepared by solid-phase synthesis using the Fmoc strategy and purified using HPLC. Four groups of animals were used: initial state (IS); control (C), diabetic control (D) and diabetic animals additionally treated with metilin (DM). The following parameters have been studied: blood glucose, MCh respiration in LV fibers, the content of cardiac ATP, ADP, AMP, phosphocreatine (PCr) and creatine (Cr), the activity of creatine kinase-MB (CK-MB) and lactate dehydrogenase (LDH) in blood plasma. Administration of metilin to STZ-treated rats decreased blood glucose, increased state 3 oxygen consumption, the respiratory control ratio in MCh of permeabilized LV fibers, and increased the functional coupling of mitochondrial CK (mt-CK) to oxidative phosphorylation compared with these parameters in group D. In STZ-treated animals metilin administration caused an increase in the PCr content and prevention of the loss of total creatine (ΣCr=PCr+Cr) in the diabetic hearts, as well as restoration of the PCr/ATP ratio in the myocardium and a decrease in the activity of CK-MB and LDH in plasma to initial values. Thus, metilin prevented energy disorders disturbances in cardiomyocytes of animals with experimental T1DM.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Metabolismo Energético , Peptídeos e Proteínas de Sinalização Intercelular , Ratos Wistar , Animais , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/tratamento farmacológico , Ratos , Masculino , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/tratamento farmacológico , Metabolismo Energético/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/efeitos dos fármacos , Glicemia/metabolismo , Miocárdio/metabolismo , Estreptozocina
18.
Biomolecules ; 14(6)2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38927133

RESUMO

Lipid peroxidation plays an important role in various pathologies and aging, at least partially mediated by ferroptosis. The role of mitochondrial lipid peroxidation during ferroptosis remains poorly understood. We show that supplementation of exogenous iron in the form of ferric ammonium citrate at submillimolar doses induces production of reactive oxygen species (ROS) and lipid peroxidation in mitochondria that precede ferroptosis in H9c2 cardiomyocytes. The mitochondria-targeted antioxidant SkQ1 and the redox mediator methylene blue, which inhibits the production of ROS in complex I of the mitochondrial electron transport chain, prevent both mitochondrial lipid peroxidation and ferroptosis. SkQ1 and methylene blue also prevented accumulation of lipofuscin observed after 24 h incubation of cardiomyocytes with ferric ammonium citrate. Using isolated cardiac mitochondria as an in vitro ferroptosis model, it was shown that rotenone (complex I inhibitor) in the presence of ferrous iron stimulates lipid peroxidation and lipofuscin accumulation. Our data indicate that ROS generated in complex I stimulate mitochondrial lipid peroxidation, lipofuscin accumulation, and ferroptosis induced by exogenous iron.


Assuntos
Ferroptose , Ferro , Peroxidação de Lipídeos , Lipofuscina , Miócitos Cardíacos , Espécies Reativas de Oxigênio , Peroxidação de Lipídeos/efeitos dos fármacos , Ferroptose/efeitos dos fármacos , Lipofuscina/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Animais , Ratos , Espécies Reativas de Oxigênio/metabolismo , Ferro/metabolismo , Linhagem Celular , Compostos de Amônio Quaternário/farmacologia , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Azul de Metileno/farmacologia , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/efeitos dos fármacos , Compostos Férricos , Plastoquinona/análogos & derivados
19.
J Physiol ; 602(14): 3423-3448, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38885335

RESUMO

Chronic coronary artery stenosis can lead to regional myocardial dysfunction in the absence of myocardial infarction by repetitive stunning, hibernation or both. The molecular mechanisms underlying repetitive stunning-associated myocardial dysfunction are not clear. We used non-targeted metabolomics to elucidate responses to chronically stunned myocardium in a canine model with and without ß-adrenergic blockade treatment. After development of left ventricular systolic dysfunction induced by ameroid constrictors on the coronary arteries, animals were randomized to 3 months of placebo, metoprolol or carvedilol. We compared these two ß-blockers with their different ß-adrenergic selectivities on myocardial function, perfusion and metabolic pathways involved in tissue undergoing chronic stunning. Control animals underwent sham surgery. Dysfunction in stunned myocardium was associated with reduced fatty acid oxidation and enhanced ketogenic amino acid metabolism, together with alterations in mitochondrial membrane phospholipid composition. These changes were consistent with impaired mitochondrial function and were linked to reduced nitric oxide and peroxisome proliferator-activated receptor signalling, resulting in a decline in adenosine monophosphate-activated protein kinase. Mitochondrial changes were ameliorated by carvedilol more than metoprolol, and improvement was linked to nitric oxide and possibly hydrogen sulphide signalling. In summary, repetitive myocardial stunning commonly seen in chronic multivessel coronary artery disease is associated with adverse metabolic remodelling linked to mitochondrial dysfunction and specific signalling pathways. These changes are reversed by ß-blockers, with the non-selective inhibitor having a more favourable impact. This is the first investigation to demonstrate that ß-blockade-associated improvement of ventricular function in chronic myocardial stunning is associated with restoration of mitochondrial function. KEY POINTS: The mechanisms responsible for the metabolic changes associated with repetitive myocardial stunning seen in chronic multivessel coronary artery disease have not been fully investigated. In a canine model of repetitive myocardial stunning, we showed that carvedilol, a non-selective ß-receptor blocker, ameliorated adverse metabolic remodelling compared to metoprolol, a selective ß1-receptor blocker, by improving nitric oxide synthase and adenosine monophosphate protein kinase function, enhancing calcium/calmodulin-dependent protein kinase, probably increasing hydrogen sulphide, and suppressing cyclic-adenosine monophosphate signalling. Mitochondrial fatty acid oxidation alterations were ameliorated by carvedilol to a larger extent than metoprolol; this improvement was linked to nitric oxide and possibly hydrogen sulphide signalling. Both ß-blockers improved the cardiac energy imbalance by reducing metabolites in ketogenic amino acid and nucleotide metabolism. These results elucidated why metabolic remodelling with carvedilol is preferable to metoprolol when treating chronic ischaemic left ventricular systolic dysfunction caused by repetitive myocardial stunning.


Assuntos
Antagonistas de Receptores Adrenérgicos beta 1 , Estenose Coronária , Metabolômica , Metoprolol , Miocárdio Atordoado , Animais , Miocárdio Atordoado/tratamento farmacológico , Miocárdio Atordoado/metabolismo , Miocárdio Atordoado/etiologia , Cães , Metoprolol/farmacologia , Estenose Coronária/tratamento farmacológico , Estenose Coronária/metabolismo , Antagonistas de Receptores Adrenérgicos beta 1/farmacologia , Antagonistas de Receptores Adrenérgicos beta 1/uso terapêutico , Carvedilol/farmacologia , Masculino , Propanolaminas/farmacologia , Carbazóis/farmacologia , Miocárdio/metabolismo , Miocárdio/patologia , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo
20.
Cell Physiol Biochem ; 58(3): 226-249, 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38857359

RESUMO

BACKGROUND/AIMS: Important benefits of intermittent hypoxic training (IHT) have emerged as an effective tool for enhancing adaptive potential in different pathological states, among which acute hypoxia dominates. Therefore, the aim of our study was to evaluate the mechanisms related to the effects of the nitric oxide system (nitrites, nitrates, carbamide, and total polyamine content) on ADP-stimulated oxygen consumption and oxidative phosphorylation in heart and liver mitochondria and biomarkers of oxidative stress in the blood, heart, and liver of rats exposed to the IHT method and acute hypoxia and treated with the amino acid L-arginine (600 mg/kg, 30 min) or the NO synthase inhibitor L-NNA (35 mg/kg, 30 min) prior to each IHT session. METHODS: We analysed the modulation of the system of oxygen-dependent processes (mitochondrial respiration with the oxygraphic method, microsomal oxidation, and lipoperoxidation processes using biochemical methods) in tissues during IHT in the formation of short-term and long-term effects (30, 60, and 180 days after the last IHT session) with simultaneous administration of L-arginine. In particular, we investigated how mitochondrial functions are modulated during intermittent hypoxia with the use of oxidation substrates (succinate or α-ketoglutarate) in bioenergetic mechanisms of cellular stability and adaptation. RESULTS: The IHT method is associated with a significant increase in the production of endogenous nitric oxide measured by the levels of its stable metabolite, nitrite anion, in both plasma (almost 7-fold) and erythrocytes (more than 7-fold) of rats. The intensification of nitric oxide-dependent pathways of metabolic transformations in the energy supply processes in the heart and liver, accompanied by oscillatory mechanisms of adaptation in the interval mode, causes a probable decrease in the production of urea and polyamines in plasma and liver, but not in erythrocytes. The administration of L-arginine prior to the IHT sessions increased the level of the nitrite-reducing component of the nitric oxide cycle, which persisted for up to 180 days of the experiment. CONCLUSION: Thus, the efficacy of IHT and its nitrite-dependent component shown in this study is associated with the formation of long-term adaptive responses by preventing the intensification of lipoperoxidation processes in tissues due to pronounced changes in the main enzymes of antioxidant defence and stabilisation of erythrocyte membranes, which has a pronounced protective effect on the system of regulation of oxygen-dependent processes as a whole.


Assuntos
Arginina , Hipóxia , Consumo de Oxigênio , Ratos Wistar , Animais , Masculino , Hipóxia/metabolismo , Ratos , Arginina/farmacologia , Arginina/análogos & derivados , Arginina/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Óxido Nítrico/metabolismo , Oxigênio/metabolismo , Adaptação Fisiológica , Mitocôndrias Hepáticas/metabolismo , Mitocôndrias Hepáticas/efeitos dos fármacos , Fosforilação Oxidativa/efeitos dos fármacos , Fígado/metabolismo , Fígado/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos dos fármacos , Nitritos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...