Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
1.
Cells ; 13(10)2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38786104

RESUMO

Radiation-induced heart disease (RIHD), a common side effect of chest irradiation, is a primary cause of mortality among patients surviving thoracic cancer. Thus, the development of novel, clinically applicable cardioprotective agents which can alleviate the harmful effects of irradiation on the heart is of great importance in the field of experimental oncocardiology. Biglycan and decorin are structurally related small leucine-rich proteoglycans which have been reported to exert cardioprotective properties in certain cardiovascular pathologies. Therefore, in the present study we aimed to examine if biglycan or decorin can reduce radiation-induced damage of cardiomyocytes. A single dose of 10 Gray irradiation was applied to induce radiation-induced cell damage in H9c2 cardiomyoblasts, followed by treatment with either biglycan or decorin at various concentrations. Measurement of cell viability revealed that both proteoglycans improved the survival of cardiac cells post-irradiation. The cardiocytoprotective effect of both biglycan and decorin involved the alleviation of radiation-induced proapoptotic mechanisms by retaining the progression of apoptotic membrane blebbing and lowering the number of apoptotic cell nuclei and DNA double-strand breaks. Our findings provide evidence that these natural proteoglycans may exert protection against radiation-induced damage of cardiac cells.


Assuntos
Apoptose , Biglicano , Decorina , Miócitos Cardíacos , Decorina/metabolismo , Biglicano/metabolismo , Apoptose/efeitos da radiação , Apoptose/efeitos dos fármacos , Animais , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/efeitos da radiação , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Ratos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Humanos
2.
Molecules ; 26(19)2021 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-34641361

RESUMO

Prunus mume blossom is an edible flower that has been used in traditional Chinese medicine for thousands of years. Flavonoids are one of the most active substances in Prunus mume blossoms. The optimal ultrasonic-assisted enzymatic extraction of flavonoids from Prunus mume blossom (FPMB), the components of FPMB, and its protective effect on injured cardiomyocytes were investigated in this study. According to our results, the optimal extraction process for FPMB is as follows: cellulase at 2.0%, ultrasonic power at 300 W, ultrasonic enzymolysis for 30 min, and an enzymolysis temperature of 40 °C. FPMB significantly promoted the survival rate of cardiomyocytes and reduced the concentration of reactive oxygen species (ROS). FPMB also improved the activities of proteases caspase-3, caspase-8, and caspase-9 in cardiomyocytes. The cardiomyocyte apoptosis rate in mice was significantly reduced by exposure to FPMB. These results suggest that the extraction rate of FPMB may be improved by an ultrasonic-assisted enzymatic method. FPMB has a protective effect on the injured cardiomyocytes.


Assuntos
Enzimas/metabolismo , Flavonoides/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Extratos Vegetais/farmacologia , Substâncias Protetoras/farmacologia , Prunus/química , Ultrassom/métodos , Animais , Masculino , Camundongos , Miócitos Cardíacos/patologia , Miócitos Cardíacos/efeitos da radiação
3.
Nat Commun ; 12(1): 3279, 2021 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-34078883

RESUMO

Targeting the molecular pathways underlying the cardiotoxicity associated with thoracic irradiation and doxorubicin (Dox) could reduce the morbidity and mortality associated with these anticancer treatments. Here, we find that vascular endothelial cells (ECs) with persistent DNA damage induced by irradiation and Dox treatment exhibit a fibrotic phenotype (endothelial-mesenchymal transition, EndMT) correlating with the colocalization of L1CAM and persistent DNA damage foci. We demonstrate that treatment with the anti-L1CAM antibody Ab417 decreases L1CAM overexpression and nuclear translocation and persistent DNA damage foci. We show that in whole-heart-irradiated mice, EC-specific p53 deletion increases vascular fibrosis and the colocalization of L1CAM and DNA damage foci, while Ab417 attenuates these effects. We also demonstrate that Ab417 prevents cardiac dysfunction-related decrease in fractional shortening and prolongs survival after whole-heart irradiation or Dox treatment. We show that cardiomyopathy patient-derived cardiovascular ECs with persistent DNA damage show upregulated L1CAM and EndMT, indicating clinical applicability of Ab417. We conclude that controlling vascular DNA damage by inhibiting nuclear L1CAM translocation might effectively prevent anticancer therapy-associated cardiotoxicity.


Assuntos
Anticorpos Neutralizantes/farmacologia , Cardiomiopatias/prevenção & controle , Cardiotoxicidade/prevenção & controle , Doxorrubicina/toxicidade , Raios gama/efeitos adversos , Molécula L1 de Adesão de Célula Nervosa/genética , Animais , Antibióticos Antineoplásicos/toxicidade , Cardiomiopatias/etiologia , Cardiomiopatias/genética , Cardiomiopatias/metabolismo , Cardiotoxicidade/etiologia , Cardiotoxicidade/genética , Cardiotoxicidade/metabolismo , Estudos de Casos e Controles , Técnicas de Cocultura , Dano ao DNA , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Células Endoteliais/efeitos da radiação , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos da radiação , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Miócitos Cardíacos/efeitos da radiação , Molécula L1 de Adesão de Célula Nervosa/antagonistas & inibidores , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
4.
PLoS One ; 16(6): e0252346, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34086732

RESUMO

Whereas it is evident that a well aligned and regular sarcomeric structure in cardiomyocytes is vital for heart function, considerably less is known about the contribution of individual elements to the mechanics of the entire cell. For instance, it is unclear whether altered Z-disc elements are the reason or the outcome of related cardiomyopathies. Therefore, it is crucial to gain more insight into this cellular organization. This study utilizes femtosecond laser-based nanosurgery to better understand sarcomeres and their repair upon damage. We investigated the influence of the extent and the location of the Z-disc damage. A single, three, five or ten Z-disc ablations were performed in neonatal rat cardiomyocytes. We employed image-based analysis using a self-written software together with different already published algorithms. We observed that cardiomyocyte survival associated with the damage extent, but not with the cell area or the total number of Z-discs per cell. The cell survival is independent of the damage position and can be compensated. However, the sarcomere alignment/orientation is changing over time after ablation. The contraction time is also independent of the extent of damage for the tested parameters. Additionally, we observed shortening rates between 6-7% of the initial sarcomere length in laser treated cardiomyocytes. This rate is an important indicator for force generation in myocytes. In conclusion, femtosecond laser-based nanosurgery together with image-based sarcomere tracking is a powerful tool to better understand the Z-disc complex and its force propagation function and role in cellular mechanisms.


Assuntos
Lasers/efeitos adversos , Miócitos Cardíacos/efeitos da radiação , Sarcômeros/efeitos da radiação , Algoritmos , Animais , Diferenciação Celular , Células Cultivadas , Processamento de Imagem Assistida por Computador/métodos , Contração Miocárdica/efeitos da radiação , Ratos , Ratos Sprague-Dawley
5.
Biomed Res Int ; 2021: 8880179, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33532500

RESUMO

Regulated necrosis (necroptosis) is crucially involved in cardiac ischaemia-reperfusion injury (MIRI). The aim of our study is to investigate whether shock wave therapy (SWT) is capable of exerting protective effects by inhibiting necroptosis during myocardial ischaemia-reperfusion (I/R) injury and the possible role of autophagy in this process. We established a hypoxia/reoxygenation (H/R) model in vitro using HL-1 cells to simulate MIRI. MTS assays and LDH cytotoxicity assay were performed to measure cell viability and cell damage. Annexin V/PI staining was used to determine apoptosis and necrosis. Western blotting was performed to assess the changes in cell signaling pathways associated with autophagy, necroptosis, and apoptosis. Reactive oxygen species (ROS) production was detected using DHE staining. Autophagosome generation and degradation (autophagic flux) were analysed using GFP and RFP tandemly tagged LC3 (tfLC3). HL-1 cells were then transfected with p62/SQSTM1 siRNA in order to analyse its role in cardioprotection. Our results revealed that SWT increased cell viability in the H/R model and decreased receptor-interacting serine/threonine-protein kinase 1 (RIPK1) and RIPK3 expression. ROS production was also inhibited by SWT. Moreover, SWT decreased Beclin1 expression and the ratio of LC3-II/LC3-I following H/R. Simultaneously, in the tfLC3 assay, the SWT provoked a decrease in the cumulative autophagosome abundance. siRNA-mediated knockdown of p62 attenuated H/R-induced necroptosis, and SWT did not exert additive effects. Taken together, SWT ameliorated H/R injury by inhibiting necroptosis. SWT also relieved the blockade of autophagic flux in response to H/R injury. The restoration of autophagic flux by SWT might contribute to its cardioprotective effect on necroptosis following H/R injury.


Assuntos
Autofagia/efeitos da radiação , Hipóxia Celular/efeitos da radiação , Tratamento por Ondas de Choque Extracorpóreas , Miócitos Cardíacos , Necroptose/efeitos da radiação , Animais , Linhagem Celular , Sobrevivência Celular/efeitos da radiação , Coração/efeitos da radiação , Camundongos , Modelos Biológicos , Traumatismo por Reperfusão Miocárdica , Miocárdio/citologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos da radiação
6.
Int J Mol Sci ; 23(1)2021 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-35008778

RESUMO

Cardiac radioablation is emerging as an alternative option for refractory ventricular arrhythmias. However, the immediate acute effect of high-dose irradiation on human cardiomyocytes remains poorly known. We measured the electrical activities of human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) upon irradiation with 0, 20, 25, 30, 40, and 50 Gy using a multi-electrode array, and cardiomyocyte function gene levels were evaluated. iPSC-CMs showed to recover their electrophysiological activities (total active electrode, spike amplitude and slope, and corrected field potential duration) within 3-6 h from the acute effects of high-dose irradiation. The beat rate immediately increased until 3 h after irradiation, but it steadily decreased afterward. Conduction velocity slowed in cells irradiated with ≥25 Gy until 6-12 h and recovered within 24 h; notably, 20 and 25 Gy-treated groups showed subsequent continuous increase. At day 7 post-irradiation, except for cTnT, cardiomyocyte function gene levels increased with increasing irradiation dose, but uniquely peaked at 25-30 Gy. Altogether, high-dose irradiation immediately and reversibly modifies the electrical conduction of cardiomyocytes. Thus, compensatory mechanisms at the cellular level may be activated after the high-dose irradiation acute effects, thereby, contributing to the immediate antiarrhythmic outcome of cardiac radioablation for refractory ventricular arrhythmias.


Assuntos
Arritmias Cardíacas/terapia , Células-Tronco Pluripotentes Induzidas/citologia , Miócitos Cardíacos/efeitos da radiação , Ablação por Radiofrequência , Arritmias Cardíacas/fisiopatologia , Relação Dose-Resposta à Radiação , Eletrodos , Fenômenos Eletrofisiológicos/efeitos da radiação , Regulação da Expressão Gênica/efeitos da radiação , Humanos , Fatores de Tempo
7.
Chin J Nat Med ; 18(6): 436-445, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32503735

RESUMO

This study investigated the effects of X-ray irradiation on primary rat cardiac fibroblasts (CFs) and its potential mechanism, as well as whether sodium tanshinone IIA sulfonate (STS) has protective effect on CFs and its possible mechanism. Our data demonstrated that X-rays inhibited cell growth and increased oxidative stress in CFs, and STS mitigated X-ray-induced injury. Enzyme-linked immuno-sorbent assay showed that X-rays increased the levels of secreted angiotensin II (Ang II) and brain natriuretic peptide (BNP). STS inhibited the X-ray-induced increases in Ang II and BNP release. Apoptosis and cell cycle of CFs were analyzed using flow cytometry. X-rays induced apoptosis in CFs, whereas STS inhibited apoptosis in CFs after X-ray irradiation. X-rays induced S-phase cell cycle arrest in CFs, which could be reversed by STS. X-rays increased the expression of phosphorylated-P38/P38, cleaved caspase-3 and caspase-3 as well as decreased the expression of phosphorylated extracellular signal-regulated kinase 1/2 (ERK 1/2)/ERK 1/2 and B cell lymphoma 2 (Bcl-2)/Bcl-2 associated X protein (BAX) in CFs, as shown by Western blotting. STS mitigated the X-ray radiation-induced expression changes of these proteins. In conclusion, our results demonstrated that STS may potentially be developed as a medical countermeasure to mitigate radiation-induced cardiac damage.


Assuntos
Fibroblastos/efeitos dos fármacos , Fibroblastos/efeitos da radiação , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/efeitos da radiação , Fenantrenos/farmacologia , Lesões por Radiação/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
8.
FASEB J ; 34(2): 3347-3358, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31919902

RESUMO

Radiation therapy (RT) is commonly used to treat solid tumors of the breast, lung, and esophagus; however, the heart is an unintentional target of ionizing radiation (IR). IR exposure to the heart results in chronic toxicities including heart failure. We hypothesize that the circadian system plays regulatory roles in minimizing the IR-induced cardiotoxicity. We treated mice in control (Day Shift), environmentally disrupted (Rotating Shift), and genetically disrupted (Per 1/2 mutant) circadian conditions with 18 Gy of IR to the heart. Compared to control mice, circadian clock disruption significantly exacerbated post-IR systolic dysfunction (by ultrasound echocardiography) and increased fibrosis in mice. At the cellular level, Bmal1 protein bound to Atm, Brca1, and Brca2 promoter regions and its expression level was inversely correlated with the DNA damage levels based on the state of the clock. Further studies with circadian synchronized cardiomyocytes revealed that Bmal1 depletion increased the IR-induced DNA damage and apoptosis. Collectively, these findings suggest that the circadian clock protects from IR-induced toxicity and potentially impacts RT treatment outcome in cancer patients through IR-induced DNA damage responses.


Assuntos
Miócitos Cardíacos/metabolismo , Proteínas Circadianas Period/genética , Lesões Experimentais por Radiação/genética , Animais , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Linhagem Celular , Dano ao DNA , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Miócitos Cardíacos/fisiologia , Miócitos Cardíacos/efeitos da radiação , Regiões Promotoras Genéticas , Lesões Experimentais por Radiação/metabolismo , Radiação Ionizante , Ratos , Sístole
9.
Prog Biophys Mol Biol ; 154: 51-61, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-31738979

RESUMO

Optogenetics enables cell-type specific monitoring and actuation via light-activated proteins. In cardiac research, expressing light-activated depolarising ion channels in cardiomyocytes allows optical pacing and defibrillation. Previous studies largely relied on epicardial illumination. Light penetration through the myocardium is however problematic when moving to larger animals and humans. To overcome this limitation, we assessed the utility of an implantable multi light-emitting diode (LED) optical probe (IMLOP) for intramural pacing of mouse hearts expressing cardiac-specific channelrhodopsin-2 (ChR2). Here we demonstrated that IMLOP insertion needs approximately 20 mN of force, limiting possible damage from excessive loads applied during implantation. Histological sections confirmed the confined nature of tissue damage during acute use. The temperature change of the surrounding tissue was below 1 K during LED operation, rendering the probe safe for use in situ. This was confirmed in control experiments where no effect on cardiac action potential conduction was observed even when using stimulation parameters twenty-fold greater than required for pacing. In situ experiments on ChR2-expressing mouse hearts demonstrated that optical stimulation is possible with light intensities as low as 700 µW/mm2; although stable pacing requires higher intensities. When pacing with a single LED, rheobase and chronaxie values were 13.3 mW/mm2 ± 0.9 mW/mm2 and 3 ms ± 0.6 ms, respectively. When doubling the stimulated volume the rheobase decreased significantly (6.5 mW/mm2 ± 0.9 mW/mm2). We have demonstrated IMLOP-based intramural optical pacing of the heart. Probes cause locally constrained tissue damage in the acute setting and require low light intensities for pacing. Further development is necessary to assess effects of chronic implantation.


Assuntos
Channelrhodopsins/metabolismo , Regulação da Expressão Gênica , Audição/fisiologia , Dispositivos Ópticos , Potenciais de Ação/efeitos da radiação , Animais , Regulação da Expressão Gênica/efeitos da radiação , Audição/efeitos da radiação , Camundongos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/efeitos da radiação , Temperatura
10.
Int J Mol Sci ; 20(20)2019 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-31652604

RESUMO

Chronic exposure to low-dose ionizing radiation is associated with an increased risk of cardiovascular disease. Alteration in energy metabolism has been suggested to contribute to radiation-induced heart pathology, mitochondrial dysfunction being a hallmark of this disease. The goal of this study was to investigate the regulatory role of acetylation in heart mitochondria in the long-term response to chronic radiation. ApoE-deficient C57Bl/6J mice were exposed to low-dose-rate (20 mGy/day) gamma radiation for 300 days, resulting in a cumulative total body dose of 6.0 Gy. Heart mitochondria were isolated and analyzed using quantitative proteomics. Radiation-induced proteome and acetylome alterations were further validated using immunoblotting, enzyme activity assays, and ELISA. In total, 71 proteins showed peptides with a changed acetylation status following irradiation. The great majority (94%) of the hyperacetylated proteins were involved in the TCA cycle, fatty acid oxidation, oxidative stress response and sirtuin pathway. The elevated acetylation patterns coincided with reduced activity of mitochondrial sirtuins, increased the level of Acetyl-CoA, and were accompanied by inactivation of major cardiac metabolic regulators PGC-1 alpha and PPAR alpha. These observations suggest that the changes in mitochondrial acetylation after irradiation is associated with impairment of heart metabolism. We propose a novel mechanism involved in the development of late cardiac damage following chronic irradiation.


Assuntos
Proteínas Mitocondriais/metabolismo , Miócitos Cardíacos/metabolismo , Processamento de Proteína Pós-Traducional , Sirtuínas/genética , Irradiação Corporal Total/efeitos adversos , Acetilação , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Regulação para Baixo , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/efeitos da radiação , Proteínas Mitocondriais/efeitos da radiação , Miócitos Cardíacos/efeitos da radiação , PPAR alfa/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo
11.
Biofactors ; 45(6): 983-990, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31509323

RESUMO

Cardiac optogenetics is an emergent research area and refers to the delivery of light-activated proteins to excitable heart tissue and the subsequent use of light for controlling the electrical function with high spatial and temporal resolution. Channelrhodopsin-2 (ChR2) is a light-sensitive ion channel with the chromophore, all trans retinal, derived from vitamin A (all-trans-retinol; retinol). In this study, we explored whether exogenous vitamin A can be a limiting factor in the light responsiveness of cardiomyocytes-expressing ChR2. We showed that in cardiomyocytes virally transduced with ChR2 (H134R)-enhanced yellow fluorescent protein, vitamin A supplements lower than 10 µM significantly increased ChR2 expression. Adding 1 µM vitamin A changed light-induced transmembrane potential difference significantly, whereas 5 µM dramatically induced membrane depolarization and triggered intracellular calcium elevation. We concluded that vitamin A supplementation can modulate the efficiency of ChR2 and provide a complementary strategy for improving the performance of optogenetic tools.


Assuntos
Proteínas de Transporte/genética , Miocárdio/metabolismo , Optogenética , Vitamina A/farmacologia , Animais , Animais Recém-Nascidos , Cálcio/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Transdução de Sinal Luminoso/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/efeitos da radiação , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/efeitos da radiação , Ratos
12.
Adv Healthc Mater ; 8(13): e1900198, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31066237

RESUMO

The use of light for triggering skeletal and cardiac muscles allows lower invasiveness higher selectivity and unprecedented possibility to target individual cells or even subcellular compartments in a temporally and spatially precise manner. Because cells are in general transparent, this requires the development of suitable interfaces that bestow light sensitivity to living matter. In the present work, successfully demonstrated is the use of conjugated polymer films as transducer to optically enhance the contraction rate of a human and patient-specific cardiac in vitro cell model. By different experimental approaches, the coupling mechanism to the photothermal effect is assigned. This work extends the range of application of the polymer-mediated cell photostimulation phenomenon to cardiac muscle cells, opening up possible applications in cardiac therapy and for implementation of in vitro studies.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Miócitos Cardíacos/metabolismo , Polímeros/química , Materiais Biocompatíveis/química , Diferenciação Celular , Sobrevivência Celular/efeitos dos fármacos , Reprogramação Celular , Vidro/química , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Luz , Contração Muscular/efeitos dos fármacos , Contração Muscular/efeitos da radiação , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos da radiação , Polímeros/farmacologia , Temperatura
13.
Nat Commun ; 10(1): 1281, 2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30894542

RESUMO

The standard technique for investigating adrenergic effects on heart function is perfusion with pharmaceutical agonists, which does not provide high temporal or spatial precision. Herein we demonstrate that the light sensitive Gs-protein coupled receptor JellyOp enables optogenetic stimulation of Gs-signaling in cardiomyocytes and the whole heart. Illumination of transgenic embryonic stem cell-derived cardiomyocytes or of the right atrium of mice expressing JellyOp elevates cAMP levels and instantaneously accelerates spontaneous beating rates similar to pharmacological ß-adrenergic stimulation. Light application to the dorsal left atrium instead leads to supraventricular extrabeats, indicating adverse effects of localized Gs-signaling. In isolated ventricular cardiomyocytes from JellyOp mice, we find increased Ca2+ currents, fractional cell shortening and relaxation rates after illumination enabling the analysis of differential Gs-signaling with high temporal precision. Thus, JellyOp expression allows localized and time-restricted Gs stimulation and will provide mechanistic insights into different effects of site-specific, long-lasting and pulsatile Gs activation.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Átrios do Coração/metabolismo , Ventrículos do Coração/metabolismo , Transdução de Sinal Luminoso , Miócitos Cardíacos/metabolismo , Optogenética/métodos , Animais , Cálcio/metabolismo , Cátions Bivalentes , Diferenciação Celular , AMP Cíclico/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Átrios do Coração/citologia , Átrios do Coração/efeitos da radiação , Ventrículos do Coração/citologia , Ventrículos do Coração/efeitos da radiação , Luz , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Embrionárias Murinas/efeitos da radiação , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos da radiação
14.
Sci Rep ; 9(1): 5000, 2019 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-30899027

RESUMO

Noninvasive X-ray stereotactic treatment is considered a promising alternative to catheter ablation in patients affected by severe heart arrhythmia. High-energy heavy ions can deliver high radiation doses in small targets with reduced damage to the normal tissue compared to conventional X-rays. For this reason, charged particle therapy, widely used in oncology, can be a powerful tool for radiosurgery in cardiac diseases. We have recently performed a feasibility study in a swine model using high doses of high-energy C-ions to target specific cardiac structures. Interruption of cardiac conduction was observed in some animals. Here we report the biological effects measured in the pig heart tissue of the same animals six months after the treatment. Immunohistological analysis of the target tissue showed (1.) long-lasting vascular damage, i.e. persistent hemorrhage, loss of microvessels, and occurrence of siderophages, (2.) fibrosis and (3.) loss of polarity of targeted cardiomyocytes and wavy fibers with vacuolization. We conclude that the observed physiological changes in heart function are produced by radiation-induced fibrosis and cardiomyocyte functional inactivation. No effects were observed in the normal tissue traversed by the particle beam, suggesting that charged particles have the potential to produce ablation of specific heart targets with minimal side effects.


Assuntos
Arritmias Cardíacas/radioterapia , Miocárdio/patologia , Miócitos Cardíacos/efeitos da radiação , Terapia por Raios X/efeitos adversos , Animais , Arritmias Cardíacas/patologia , Ablação por Cateter/efeitos adversos , Coração/efeitos da radiação , Frequência Cardíaca/efeitos da radiação , Íons Pesados/efeitos adversos , Humanos , Miócitos Cardíacos/patologia , Radiação não Ionizante/efeitos adversos , Radiocirurgia/efeitos adversos , Técnicas Estereotáxicas/tendências , Suínos
15.
Int J Mol Sci ; 20(3)2019 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-30717456

RESUMO

Space radiation has recently been considered a risk factor for astronauts' cardiac health. As an example, for the case of how to query and identify datasets within NASA's GeneLab database and demonstrate the database utility, we used an unbiased systems biology method for identifying key genes/drivers for the contribution of space radiation on the cardiovascular system. This knowledge can contribute to designing appropriate experiments targeting these specific pathways. Microarray data from cardiomyocytes of male C57BL/6 mice followed-up for 28 days after exposure to 900 mGy of 1 GeV proton or 150 mGy of 1 GeV/n 56Fe were compared to human endothelial cells (HUVECs) cultured for 7 days on the International Space Station (ISS). We observed common molecular pathways between simulated space radiation and HUVECs flown on the ISS. The analysis suggests FYN is the central driver/hub for the cardiovascular response to space radiation: the known oxidative stress induced immediately following radiation would only be transient and would upregulate FYN, which in turn would reduce reactive oxygen species (ROS) levels, protecting the cardiovascular system. The transcriptomic signature of exposure to protons was also much closer to the spaceflight signature than 56Fe's signature. To our knowledge, this is the first time GeneLab datasets were utilized to provide potential biological indications that the majority of ions on the ISS are protons, clearly illustrating the power of omics analysis. More generally, this work also demonstrates how to combine animal radiation studies done on the ground and spaceflight studies to evaluate human risk in space.


Assuntos
Sistema Cardiovascular/efeitos da radiação , Miócitos Cardíacos/efeitos da radiação , Proteínas Proto-Oncogênicas c-fyn/genética , Radiação Ionizante , Voo Espacial , Transcriptoma , Animais , Sistema Cardiovascular/metabolismo , Células Cultivadas , Radiação Cósmica , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/metabolismo , Estresse Oxidativo , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Prótons , Espécies Reativas de Oxigênio/metabolismo
16.
J Cell Mol Med ; 23(3): 1963-1975, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30592150

RESUMO

The aggressive immunological activity elicited by acute viral myocarditis contributes to a large amount of cardiomyocytes loss and poor prognosis of patients in clinic. Low-intensity pulsed ultrasound (LIPUS), which is an effective treatment modality for osteoarthropathy, has been recently illustrated regulating the overactive inflammatory response in various diseases. Here, we aimed to investigate whether LIPUS could attenuate coxsackievirus B3 (CVB3) infection-induced injury by coordinating the inflammatory response. Male BALB/c mice were inoculated intraperitoneally with CVB3 to establish the model of acute viral myocarditis. LIPUS treatment was given on Day 1, Day 1, 3 and Day 1, 3, 5 post-inoculation, respectively. All mice were followed up for 14 days. Day 1, 3, 5 LIPUS treatment significantly improved the survival rate, attenuated the ventricular dysfunction and ameliorated the cardiac histopathological injury of CVB3-infected mice. Western blotting analysis showed Day 1, 3, 5 LIPUS treatment decreased pro-inflammatory cytokines, increased the activation of caveolin-1 and suppressed p38 mitogen-activated protein kinase (MAPK) and extracellular signal-regulated kinase (ERK) signallings in heart tissue. RAW264.7 cells were treated with lipopolysaccharides (LPS) to simulate the augmented inflammatory response in vivo. LIPUS treatment on RAW264.7 inhibited the expression of pro-inflammatory cytokines, activated caveolin-1 and suppressed p38 MAPK and ERK signallings. Transfecting RAW264.7 with caveolin-1 siRNA blunted the suppression of pro-inflammatory cytokines and MAPK signallings by LIPUS treatment. Taken together, we demonstrated for the first time that LIPUS treatment attenuated the aggressive inflammatory response during acute viral myocarditis. The underlying mechanism may be activating caveolin-1 and suppressing MAPK signallings.


Assuntos
Infecções por Coxsackievirus/terapia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Coração/efeitos da radiação , Inflamação/terapia , Miocardite/terapia , Transdução de Sinais/efeitos da radiação , Animais , Caveolina 1/metabolismo , Infecções por Coxsackievirus/metabolismo , Infecções por Coxsackievirus/virologia , Citocinas/metabolismo , Enterovirus/patogenicidade , Humanos , Inflamação/virologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Miocardite/virologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/efeitos da radiação , Miócitos Cardíacos/virologia , Células RAW 264.7 , Terapia por Ultrassom/métodos , Ondas Ultrassônicas
17.
Int J Radiat Biol ; 94(12): 1095-1103, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30247079

RESUMO

Purpose: Radiation-induced heart disease caused by cardiac exposure to ionizing radiation comprises a variety of cardiovascular effects. Research in this field has been hampered by limited availability of clinical samples and appropriate test models. In this study, we wanted to elucidate the molecular mechanisms underlying electrophysiological changes, which we have observed in a previous study. Materials and methods: We employed RNA deep-sequencing of human-induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) 48 h after 5 Gy X-ray irradiation. By comparison to public data from hiPSC-CMs and human myocardium, we verified the expression of cardiac-specific genes in hiPSC-CMs. Results were validated by qRT-PCR. Results: Differentially gene expression analysis identified 39 and 481 significantly up- and down-regulated genes after irradiation, respectively. Besides, a large fraction of genes associated with cell cycle processes, we identified genes implicated in cardiac calcium homeostasis (PDE3B), oxidative stress response (FDXR and SPATA18) and the etiology of cardiomyopathy (SGCD, BBC3 and GDF15). Conclusions: Notably, observed gene expression characteristics specific to hiPSC-CMs might be relevant regarding further investigations of the response to external stressors like radiation. The genes and biological processes highlighted in our study present promising starting points for functional follow-up studies for which hiPSC-CMs could pose an appropriate cell model when cell type specific peculiarities are taken into account.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Miócitos Cardíacos/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/fisiologia , Expressão Gênica/efeitos da radiação , Fator 15 de Diferenciação de Crescimento/fisiologia , Humanos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Análise de Sequência de RNA , Raios X
18.
Biomed Environ Sci ; 31(8): 561-571, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30231960

RESUMO

OBJECTIVE: To detect the effects of microwave on calcium levels in primary hippocampal neurons and primary cardiomyocytes by the real-time microwave exposure combined with laser scanning confocal microscopy. METHODS: The primary hippocampal neurons and primary cardiomyocytes were cultured and labeled with probes, including Fluo-4 AM, Mag-Fluo-AM, and Rhod-2, to reflect the levels of whole calcium [Ca2+], endoplasmic reticulum calcium [Ca2+]ER, and mitochondrial calcium [Ca2+]MIT, respectively. Then, the cells were exposed to a pulsed microwave of 2.856 GHz with specific absorption rate (SAR) values of 0, 4, and 40 W/kg for 6 min to observe the changes in calcium levels. RESULTS: The results showed that the 4 and 40 W/kg microwave radiation caused a significant decrease in the levels of [Ca2+], [Ca2+]ER, and [Ca2+]MIT in primary hippocampal neurons. In the primary cardiomyocytes, only the 40 W/kg microwave radiation caused the decrease in the levels of [Ca2+], [Ca2+]ER, and [Ca2+]MIT. Primary hippocampal neurons were more sensitive to microwave exposure than primary cardiomyocytes. The mitochondria were more sensitive to microwave exposure than the endoplasmic reticulum. CONCLUSION: The calcium efflux was occurred during microwave exposure in primary hippocampal neurons and primary cardiomyocytes. Additionally, neurons and mitochondria were sensitive cells and organelle respectively.


Assuntos
Cálcio/metabolismo , Micro-Ondas , Miócitos Cardíacos/efeitos da radiação , Neurônios/efeitos da radiação , Animais , Células Cultivadas , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/efeitos da radiação , Hipocampo/citologia , Mitocôndrias/metabolismo , Mitocôndrias/efeitos da radiação , Miócitos Cardíacos/metabolismo , Neurônios/metabolismo , Ratos Wistar
19.
Int J Mol Med ; 42(5): 2849-2858, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30226567

RESUMO

Radiotherapy significantly increases survival innumerous cancer patients, although it may have delayed adverse effects, including significant short­ and long­term effects on cardiovascular function, leading to significant morbidity and mortality. However, the mechanisms underlying these effects remain unclear. Cardiomyocyte senescence contributes to cardiovascular disease via impaired cardiac function. MicroRNA­34a (miR­34a) is a senescence­associated miR involved in the pathology of cardiovascular diseases, while macrophage migration inhibitory factor (MIF) is a cardioprotective cytokine with an important role in cardiovascular diseases. The present study aimed to determine whether MIF has a cytoprotective effect in cardiomyocytes exposed to radiation through modulating miR­34a. Human cardiomyocytes (HCMs) were incubated with MIF and then exposed to radiation. Cellular proliferation was measured using a Cell Counting Kit­8, while cellular senescence was evaluated based on the senescence­associated ß­galactosidase activity and the gene expression levels of cyclin­dependent kinase inhibitor 1a (Cdkn1a) and Cdkn2c. Oxidative stress was evaluated by measuring the generation of reactive oxygen species and malondialdehyde, as well as the expression of antioxidant genes. In addition, HCMs were treated with small interfering RNA against sirtuin 1 (SIRT1) to examine the role of this gene in MIF­associated rejuvenation following radiation­associated senescence. miR­34a was significantly increased in HCMs exposed to radiation, while MIF inhibited senescence by suppressing miR­34a. SIRT1 was identified as a target gene of miR­34a, mediating the anti­senescence effect induced by MIF. Furthermore, MIF rejuvenation involved rebalancing the oxidation process disturbed by radiation. These results provided direct evidence that inhibition of miR­34a by MIF protected against radiation­induced cardiomyocyte senescence via targeting SIRT1. Inhibition of miR­34a by MIF may thus be a novel strategy for combating cardiac radiation­associated damage.


Assuntos
Senescência Celular/efeitos da radiação , Oxirredutases Intramoleculares/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , MicroRNAs/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos da radiação , Sirtuína 1/metabolismo , Linhagem Celular , Humanos , MicroRNAs/genética , Miócitos Cardíacos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos da radiação , Regulação para Cima/efeitos da radiação
20.
Acta Biochim Pol ; 65(2): 309-318, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29906297

RESUMO

Several lines of evidence indicate that exposure of heart to ionizing radiation increases the risk of cardiotoxicity manifested by heart dysfunction and cardiovascular diseases. It was initially believed that the heart is an organ relatively resistant to radiation. Currently, however, it is suspected that even low doses of radiation (< 2 Gy) may have a negative impact on the cardiovascular system. Cardiotoxicity of ionizing radiation is associated with metabolic changes observed in cardiac cells injured by radiation. In this study, we used human cardiomyocytes as a model system, and studied their metabolic response to radiation using high-resolution magic angle spinning nuclear magnetic resonance techniques (HR-MAS NMR). Human cardiomyocytes cultured in vitro were exposed to ionizing radiation and their survival was assessed by clonogenic assay. Changes in apoptosis intensity and cell cycle distribution after the irradiation were measured as well. NMR spectra of cardiomyocytes were acquired using Bruker Avance 400 MHz spectrometer at a spinning rate of 3200 Hz. Survival of cardiomyocytes after NMR experiments was assessed by the Trypan blue exclusion assay. Exposure of cardiomyocytes to small doses of ionizing radiation had no effect on cell proliferation potential and intensity of cell death. However, analysis of metabolic profiles revealed changes in lipids, threonine, glycine, glycerophosphocholine, choline, valine, isoleucine, glutamate, reduced glutathione and taurine metabolism. The results of this study showed that ionizing radiation affects metabolic profiles of cardiomyocytes even at low doses, which potentially have no effect on cell viability.


Assuntos
Metaboloma/efeitos da radiação , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/efeitos da radiação , Radiação Ionizante , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Células Cultivadas , Relação Dose-Resposta à Radiação , Humanos , Espectroscopia de Ressonância Magnética , Metabolômica/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...