Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
Add more filters

Publication year range
1.
BMC Cardiovasc Disord ; 24(1): 323, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38918713

ABSTRACT

BACKGROUND: Radiotherapy is a primary local treatment for tumors, yet it may lead to complications such as radiation-induced heart disease (RIHD). Currently, there is no standardized approach for preventing RIHD. Dexmedetomidine (Dex) is reported to have cardio-protection effects, while its role in radiation-induced myocardial injury is unknown. In the current study, we aimed to evaluate the radioprotective effect of dexmedetomidine in X-ray radiation-treated mice. METHODS: 18 male mice were randomized into 3 groups: control, 16 Gy, and 16 Gy + Dex. The 16 Gy group received a single dose of 16 Gy X-ray radiation. The 16 Gy + Dex group was pretreated with dexmedetomidine (30 µg/kg, intraperitoneal injection) 30 min before X-ray radiation. The control group was treated with saline and did not receive X-ray radiation. Myocardial tissues were collected 16 weeks after X-ray radiation. Hematoxylin-eosin staining was performed for histopathological examination. Terminal deoxynucleotidyl transferase dUTP nick-end labeling staining was performed to assess the state of apoptotic cells. Immunohistochemistry staining was performed to examine the expression of CD34 molecule and von Willebrand factor. Besides, western blot assay was employed for the detection of apoptosis-related proteins (BCL2 apoptosis regulator and BCL2-associated X) as well as autophagy-related proteins (microtubule-associated protein 1 light chain 3, beclin 1, and sequestosome 1). RESULTS: The findings demonstrated that 16 Gy X-ray radiation resulted in significant changes in myocardial tissues, increased myocardial apoptosis, and activated autophagy. Pretreatment with dexmedetomidine significantly protects mice against 16 Gy X-ray radiation-induced myocardial injury by inhibiting apoptosis and autophagy. CONCLUSION: In summary, our study confirmed the radioprotective effect of dexmedetomidine in mitigating cardiomyocyte apoptosis and autophagy induced by 16 Gy X-ray radiation.


Subject(s)
Apoptosis , Autophagy , Dexmedetomidine , Myocytes, Cardiac , Radiation Injuries, Experimental , Animals , Autophagy/drug effects , Autophagy/radiation effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Myocytes, Cardiac/radiation effects , Myocytes, Cardiac/metabolism , Apoptosis/drug effects , Male , Dexmedetomidine/pharmacology , Radiation Injuries, Experimental/prevention & control , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/drug therapy , Radiation-Protective Agents/pharmacology , Disease Models, Animal , Signal Transduction/drug effects , Mice , Autophagy-Related Proteins/metabolism , Mice, Inbred C57BL , Apoptosis Regulatory Proteins/metabolism
2.
Int Heart J ; 65(4): 730-737, 2024.
Article in English | MEDLINE | ID: mdl-39085112

ABSTRACT

Sodium tanshinone IIA sulfonate (STS), which is extracted from a Chinese medicinal herb, possesses many pharmacologic functions, such as coronary dilation, anti-inflammatory properties, and antiapoptotic and antioxidant effects. It remains unknown whether STS can protect cardiomyocytes injured after radiation therapy. An in vitro Sprague-Dawley (SD) rat neonatal cardiomyocyte system was established. Primary cardiomyocytes (PCMs) from neonatal SD rats were isolated under sterile conditions. PCM cells were divided into a control group (0 Gy/hour) and 5 experimental radiation therapy groups (0.25 Gy/hour, 0.5 Gy/hour, 1 Gy/hour, 2 Gy/hour, and 4 Gy/hour). Cell viability, the content of malondialdehyde (MDA), the lactate dehydrogenase (LDH) leakage rate, and superoxide dismutase (SOD) and glutathione (GSH) activities were recorded separately in each group after 7 days of culture. Western blot was used to detect the levels of p38, caspase-3 protein, and X protein (BAX) associated with B-cell lymphoma 2 (Bcl-2) in PCMs. X-rays inhibited cell growth, decreased cell viability, and induced an oxidative stress response in PCMs. STS and SB203580 (the inhibitor of P38 mitogen-activated protein kinase pathway) alleviated X-ray-induced damage to PCMs. An enzyme-linked immunosorbent assay showed that X-rays increased the cTnT level. STS and SB203580 ameliorated the X-ray-induced increase in cTnT leakage. X-rays enhanced the expression of p38/p-p38 and caspase-3 while reducing the expression of Bcl-2/BAX in PCMs, as demonstrated by western blotting. STS and SB203580 mitigated the changes in protein expression triggered by X-ray radiation. In conclusions, STS was shown to exert significant cardioprotective, anti-inflammatory, and antioxidant effects in PCMs by inhibiting the p38 mitogen-activated protein kinase pathway.


Subject(s)
Myocytes, Cardiac , Phenanthrenes , Rats, Sprague-Dawley , p38 Mitogen-Activated Protein Kinases , Animals , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/radiation effects , Rats , Phenanthrenes/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Cells, Cultured , Animals, Newborn , Cell Survival/drug effects , Oxidative Stress/drug effects , Apoptosis/drug effects , MAP Kinase Signaling System/drug effects , Antioxidants/pharmacology
3.
FASEB J ; 34(2): 3347-3358, 2020 02.
Article in English | MEDLINE | ID: mdl-31919902

ABSTRACT

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.


Subject(s)
Myocytes, Cardiac/metabolism , Period Circadian Proteins/genetics , Radiation Injuries, Experimental/genetics , Animals , Apoptosis , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Cell Line , DNA Damage , Mice , Mice, Inbred C57BL , Mutation , Myocytes, Cardiac/physiology , Myocytes, Cardiac/radiation effects , Promoter Regions, Genetic , Radiation Injuries, Experimental/metabolism , Radiation, Ionizing , Rats , Systole
4.
Int J Mol Sci ; 23(1)2021 Dec 29.
Article in English | MEDLINE | ID: mdl-35008778

ABSTRACT

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.


Subject(s)
Arrhythmias, Cardiac/therapy , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/radiation effects , Radiofrequency Ablation , Arrhythmias, Cardiac/physiopathology , Dose-Response Relationship, Radiation , Electrodes , Electrophysiological Phenomena/radiation effects , Gene Expression Regulation/radiation effects , Humans , Time Factors
5.
Molecules ; 26(19)2021 Sep 25.
Article in English | MEDLINE | ID: mdl-34641361

ABSTRACT

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.


Subject(s)
Enzymes/metabolism , Flavonoids/pharmacology , Myocytes, Cardiac/drug effects , Plant Extracts/pharmacology , Protective Agents/pharmacology , Prunus/chemistry , Ultrasonics/methods , Animals , Male , Mice , Myocytes, Cardiac/pathology , Myocytes, Cardiac/radiation effects
6.
J Cell Mol Med ; 23(3): 1963-1975, 2019 03.
Article in English | MEDLINE | ID: mdl-30592150

ABSTRACT

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.


Subject(s)
Coxsackievirus Infections/therapy , Extracellular Signal-Regulated MAP Kinases/metabolism , Heart/radiation effects , Inflammation/therapy , Myocarditis/therapy , Signal Transduction/radiation effects , Animals , Caveolin 1/metabolism , Coxsackievirus Infections/metabolism , Coxsackievirus Infections/virology , Cytokines/metabolism , Enterovirus/pathogenicity , Humans , Inflammation/virology , Male , Mice , Mice, Inbred BALB C , Myocarditis/virology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/radiation effects , Myocytes, Cardiac/virology , RAW 264.7 Cells , Ultrasonic Therapy/methods , Ultrasonic Waves
7.
Int J Mol Sci ; 20(3)2019 Feb 03.
Article in English | MEDLINE | ID: mdl-30717456

ABSTRACT

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.


Subject(s)
Cardiovascular System/radiation effects , Myocytes, Cardiac/radiation effects , Proto-Oncogene Proteins c-fyn/genetics , Radiation, Ionizing , Space Flight , Transcriptome , Animals , Cardiovascular System/metabolism , Cells, Cultured , Cosmic Radiation , Gene Expression Regulation , Humans , Male , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Oxidative Stress , Proto-Oncogene Proteins c-fyn/metabolism , Protons , Reactive Oxygen Species/metabolism
8.
Int J Mol Sci ; 20(20)2019 Oct 22.
Article in English | MEDLINE | ID: mdl-31652604

ABSTRACT

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.


Subject(s)
Mitochondrial Proteins/metabolism , Myocytes, Cardiac/metabolism , Protein Processing, Post-Translational , Sirtuins/genetics , Whole-Body Irradiation/adverse effects , Acetylation , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Down-Regulation , Female , Mice , Mice, Inbred C57BL , Mitochondria, Heart/metabolism , Mitochondria, Heart/radiation effects , Mitochondrial Proteins/radiation effects , Myocytes, Cardiac/radiation effects , PPAR alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
9.
Physiology (Bethesda) ; 32(1): 33-41, 2017 01.
Article in English | MEDLINE | ID: mdl-27927803

ABSTRACT

Regenerative mechanisms reported in the hearts of lower vertebrates have been recapitulated in the mammalian milieu, and recent studies have provided strong evidence for cardiomyocyte turnover in humans. These findings speak to an emerging consensus that adult mammalian cardiomyocytes do have the ability to divide, and it stands to reason that enrichment of this innate proliferative capacity should prove essential for complete cardiac regeneration.


Subject(s)
Heart/physiology , Myocytes, Cardiac/physiology , Regeneration , Animals , Carbon Radioisotopes , Cell Proliferation/radiation effects , Disease Models, Animal , Heart/radiation effects , Heart Failure/physiopathology , Humans , Mice , Myocytes, Cardiac/radiation effects , Radiometric Dating , Regeneration/radiation effects
10.
Biomed Environ Sci ; 31(8): 561-571, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30231960

ABSTRACT

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.


Subject(s)
Calcium/metabolism , Microwaves , Myocytes, Cardiac/radiation effects , Neurons/radiation effects , Animals , Cells, Cultured , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/radiation effects , Hippocampus/cytology , Mitochondria/metabolism , Mitochondria/radiation effects , Myocytes, Cardiac/metabolism , Neurons/metabolism , Rats, Wistar
11.
Cell Physiol Biochem ; 42(2): 673-684, 2017.
Article in English | MEDLINE | ID: mdl-28618416

ABSTRACT

BACKGROUND: Autophagy plays an important role in cardiovascular disease. Controversy still exists regarding the effect of autophagy on ischemic/hypoxic myocardium. Cardiac shock wave therapy (CSWT) is an effective alternative treatment for refractory ischemic heart disease. Whether CSWT can regulate cardiomyocyte autophagy under hypoxic conditions is not clear. We established a myocardial hypoxia model using the H9c2 cell line and performed shock waves (SWs) treatment to evaluate the effect of SW on autophagy. METHODS: The H9c2 cells were incubated under hypoxic conditions, and SW treatment was then performed at energies of 0.02, 0.05, or 0.10 mJ/mm2. The cell viability and intracellular ATP level were examined. Western blot analysis was used to assess the expression of LC3B, AMPK, mTOR, Beclin-1, Sirt1, and HIF-1α. Autophagic vacuoles were visualized by monodansylcadaverine staining. RESULTS: After the 24-hour hypoxic period, cardiomyocyte viability and ATP levels were decreased and autophagy was significantly increased in H9c2 cells. SW treatment with an energy of 0.05 mJ/mm2 significantly increased the cellular viability, ATP level, LC3B-II/I, and number of autophagic vacuoles. In addition, phosphorylated AMPK and Sirt1 were increased and phosphorylated mTOR and HIF-1α were decreased after SW treatment. CONCLUSION: SW treatment can potentially promote cardiomyocyte autophagy during hypoxia and protect cardiomyocyte function by regulating the AMPK/mTOR pathway.


Subject(s)
Autophagy/radiation effects , High-Energy Shock Waves/therapeutic use , Myocardial Ischemia/therapy , Myocytes, Cardiac/radiation effects , Animals , Apoptosis/radiation effects , Cell Hypoxia/genetics , Cell Line , Cell Survival/radiation effects , Disease Models, Animal , Humans , Myocardial Ischemia/physiopathology , Myocytes, Cardiac/pathology , Phagosomes/metabolism , Phagosomes/radiation effects , Phosphorylation , Rats , Signal Transduction/radiation effects
12.
Can J Physiol Pharmacol ; 95(10): 1190-1203, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28750189

ABSTRACT

Irradiation of normal tissues leads to acute increase in reactive oxygen/nitrogen species that serve as intra- and inter-cellular signaling to alter cell and tissue function. In the case of chest irradiation, it can affect the heart, blood vessels, and lungs, with consequent tissue remodelation and adverse side effects and symptoms. This complex process is orchestrated by a large number of interacting molecular signals, including cytokines, chemokines, and growth factors. Inflammation, endothelial cell dysfunction, thrombogenesis, organ dysfunction, and ultimate failing of the heart occur as a pathological entity - "radiation-induced heart disease" (RIHD) that is major source of morbidity and mortality. The purpose of this review is to bring insights into the basic mechanisms of RIHD that may lead to the identification of targets for intervention in the radiotherapy side effect. Studies of authors also provide knowledge about how to select targeted drugs or biological molecules to modify the progression of radiation damage in the heart. New prospective studies are needed to validate that assessed factors and changes are useful as early markers of cardiac damage.


Subject(s)
Coronary Vessels/radiation effects , Heart Diseases/etiology , Inflammation Mediators/metabolism , Myocytes, Cardiac/radiation effects , Radiation Injuries/etiology , Reactive Oxygen Species/metabolism , Animals , Apoptosis/radiation effects , Biomarkers/metabolism , Coronary Vessels/metabolism , Coronary Vessels/pathology , DNA Damage , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelial Cells/radiation effects , Heart Diseases/metabolism , Heart Diseases/pathology , Humans , Lipid Peroxidation/radiation effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress/radiation effects , Radiation Injuries/metabolism , Radiation Injuries/pathology , Signal Transduction/radiation effects
13.
J Mol Cell Cardiol ; 98: 146-58, 2016 09.
Article in English | MEDLINE | ID: mdl-27418252

ABSTRACT

Proper ß-adrenergic signaling is indispensable for modulating heart frequency. Studies on extremely-low-frequency pulsed electromagnetic field (ELF-PEMF) effects in the heart beat function are contradictory and no definitive conclusions were obtained so far. To investigate the interplay between ELF-PEMF exposure and ß-adrenergic signaling, cultures of primary murine neonatal cardiomyocytes and of sinoatrial node were exposed to ELF-PEMF and short and long-term effects were evaluated. The ELF-PEMF generated a variable magnetic induction field of 0-6mT at a frequency of 75Hz. Exposure to 3mT ELF-PEMF induced a decrease of contraction rate, Ca(2+) transients, contraction force, and energy consumption both under basal conditions and after ß-adrenergic stimulation in neonatal cardiomyocytes. ELF-PEMF exposure inhibited ß-adrenergic response in sinoatrial node (SAN) region. ELF-PEMF specifically modulated ß2 adrenergic receptor response and the exposure did not modify the increase of contraction rate after adenylate cyclase stimulation by forskolin. In HEK293T cells transfected with ß1 or ß2 adrenergic receptors, ELF-PEMF exposure induced a rapid and selective internalization of ß2 adrenergic receptor. The ß-adrenergic signaling, was reduced trough Gi protein by ELF-PEMF exposure since the phosphorylation level of phospholamban and the PI3K pathway were impaired after isoproterenol stimulation in neonatal cardiomyocytes. Long term effects of ELF-PEMF exposure were assessed in cultures of isolated cardiomyocytes. ELF-PEMF counteracts cell size increase, the generation of binucleated of cardiomyocytes and prevents the up-regulation of hypertrophic markers after ß-adrenergic stimulation, indicating an inhibition of cell growth and maturation. These data show that short and long term exposure to ELF-PEMF induces a reduction of cardiac ß-adrenergic response at molecular, functional and adaptative levels.


Subject(s)
Electromagnetic Fields , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/radiation effects , Receptors, Adrenergic, beta/metabolism , Adrenergic beta-Agonists/pharmacology , Algorithms , Animals , Calcium/metabolism , Calcium Signaling , Energy Metabolism/drug effects , Energy Metabolism/radiation effects , Mice , Models, Biological , Myocardial Contraction/drug effects , Myocardial Contraction/radiation effects , Myocytes, Cardiac/drug effects , Receptors, Adrenergic, beta/genetics , Signal Transduction/drug effects , Signal Transduction/radiation effects , Sinoatrial Node/drug effects , Sinoatrial Node/physiology , Sinoatrial Node/radiation effects
14.
J Cell Mol Med ; 20(7): 1352-66, 2016 07.
Article in English | MEDLINE | ID: mdl-26991817

ABSTRACT

We investigated whether low-dose radiation (LDR) can prevent late-stage diabetic cardiomyopathy and whether this protection is because of the induction of anti-apoptotic and anti-oxidant pathways. Streptozotocin-induced diabetic C57BL/6J mice were treated with/without whole-body LDR (12.5, 25, or 50 mGy) every 2 days. Twelve weeks after onset of diabetes, cardiomyopathy was diagnosed characterized by significant cardiac dysfunction, hypertrophy and histopathological abnormalities associated with increased oxidative stress and apoptosis, which was prevented by LDR (25 or 50 mGy only). Low-dose radiation-induced cardiac protection also associated with P53 inactivation, enhanced Nrf2 function and improved Akt activation. Next, for the mechanistic study, mouse primary cardiomyocytes were treated with high glucose (33 mmol/l) for 24 hrs and during the last 15 hrs bovine serum albumin-conjugated palmitate (62.5 µmol/l) was added into the medium to mimic diabetes, and cells were treated with LDR (25 mGy) every 6 hrs during the whole process of HG/Pal treatment. Data show that blocking Akt/MDM2/P53 or Akt/Nrf2 pathways with small interfering RNA of akt, mdm2 and nrf2 not only prevented LDR-induced anti-apoptotic and anti-oxidant effects but also prevented LDR-induced suppression on cardiomyocyte hypertrophy and fibrosis against HG/Pal. Low-dose radiation prevented diabetic cardiomyopathy by improving cardiac function and hypertrophic remodelling attributed to Akt/MDM2/P53-mediated anti-apoptotic and Akt/Nrf2-mediated anti-oxidant pathways simultaneously.


Subject(s)
Antioxidants/therapeutic use , Apoptosis/radiation effects , Diabetes Mellitus, Type 1/complications , Diabetic Cardiomyopathies/prevention & control , Diabetic Cardiomyopathies/radiotherapy , Proto-Oncogene Proteins c-akt/metabolism , Animals , Antioxidants/pharmacology , Biomarkers, Tumor/blood , Cardiomegaly/blood , Cardiomegaly/complications , Cardiomegaly/drug therapy , Cardiomegaly/pathology , Diabetes Mellitus, Type 1/blood , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/pathology , Dose-Response Relationship, Radiation , Fibrosis , Glucose/toxicity , Glycogen Synthase Kinase 3 beta/metabolism , Hyperglycemia/complications , Hyperglycemia/drug therapy , Hyperglycemia/radiotherapy , Male , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/radiation effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/radiation effects , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Palmitates/toxicity , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects , Tumor Suppressor Protein p53/metabolism , X-Rays
15.
Biochim Biophys Acta ; 1848(10 Pt A): 2118-25, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26112464

ABSTRACT

Ca2+ activation and membrane electroporation by 10-ns and 4-ms electric pulses (nsEP and msEP) were compared in rat embryonic cardiomyocytes. The lowest electric field which triggered Ca2+ transients was expectedly higher for nsEP (36 kV/cm) than for msEP (0.09 kV/cm) but the respective doses were similar (190 and 460 mJ/g). At higher intensities, both stimuli triggered prolonged firing in quiescent cells. An increase of basal Ca2+ level by >10 nM in cells with blocked voltage-gated Ca2+ channels and depleted Ca2+ depot occurred at 63 kV/cm (nsEP) or 0.14 kV/cm (msEP) and was regarded as electroporation threshold. These electric field values were at 150-230% of stimulation thresholds for both msEP and nsEP, notwithstanding a 400,000-fold difference in pulse duration. For comparable levels of electroporative Ca2+ uptake, msEP caused at least 10-fold greater uptake of propidium than nsEP, suggesting increased yield of larger pores. Electroporation by msEP started Ca2+ entry abruptly and locally at the electrode-facing poles of cell, followed by a slow diffusion to the center. In a stark contrast, nsEP evoked a "supra-electroporation" pattern of slower but spatially uniform Ca2+ entry. Thus nsEP and msEP had comparable dose efficiency, but differed profoundly in the size and localization of electropores.


Subject(s)
Cell Membrane Permeability/physiology , Electroporation/methods , Myocytes, Cardiac/physiology , Myocytes, Cardiac/radiation effects , Propidium/pharmacokinetics , Animals , Cell Membrane Permeability/radiation effects , Cells, Cultured , Dose-Response Relationship, Radiation , Metabolic Clearance Rate/radiation effects , Radiation Dosage , Rats , Static Electricity
16.
Pharmacol Res ; 111: 859-866, 2016 09.
Article in English | MEDLINE | ID: mdl-27480202

ABSTRACT

Despite the great clinical significance of radiation-induced cardiac damage, experimental investigation of its mechanisms is an unmet need in medicine. Beneficial effects of growth hormone-releasing hormone (GHRH) agonists in regeneration of the heart have been demonstrated. The aim of this study was the evaluation of the potential of modern GHRH agonistic analogs in prevention of radiation damage in an in vitro cardiac myocyte-based model. Cultures of cardiac myocytes isolated from newborn rats (NRVM) were exposed to a radiation dose of 10Gy. The effects of the agonistic analogs, JI-34 and MR-356, of human GHRH on cell viability, proliferation, their mechanism of action and the protein expression of the GHRH/SV1 receptors were studied. JI-34 and MR-356, had no effect on cell viability or proliferation in unirradiated cultures. However, in irradiated cells JI-34 showed protective effects on cell viability at concentrations of 10 and 100nM, and MR-356 at 500nM; but no such protective effect was detected on cell proliferation. Both agonistic analogs decreased radiation-induced ROS level and JI-34 interfered with the activation of SAFE/RISK pathways. Using Western blot analysis, a 52kDa protein isoform of GHRHR was detected in the samples in both irradiated and unirradiated cells. Since GHRH agonistic analogs, JI-34 and MR-356 alleviated radiation-induced damage of cardiac myocytes, they should be tested in vivo as potential protective agents against radiogenic heart damage.


Subject(s)
Alprostadil/analogs & derivatives , Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone-Releasing Hormone/agonists , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/radiation effects , Peptide Fragments/pharmacology , Radiation-Protective Agents/pharmacology , Alprostadil/pharmacology , Animals , Animals, Newborn , Cardiotoxicity , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Cells, Cultured , Cytoprotection , Dose-Response Relationship, Drug , Growth Hormone-Releasing Hormone/metabolism , Growth Hormone-Releasing Hormone/pharmacology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats, Wistar , Reactive Oxygen Species/metabolism , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/agonists , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects
17.
Can J Physiol Pharmacol ; 94(8): 849-57, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27203380

ABSTRACT

Microwaves may exert adverse biological effects on the cardiovascular system at the integrated system and cellular levels. However, the mechanism underlying such effects remains poorly understood. Here, we report a previously uncharacterized mechanism through which microwaves damage myocardial cells. Rats were treated with 2450 MHz microwave radiation at 50, 100, 150, or 200 mW/cm(2) for 6 min. Microwave treatment significantly enhanced the levels of various enzymes in serum. In addition, it increased the malondialdehyde content while decreasing the levels of antioxidative stress enzymes, activities of enzyme complexes I-IV, and ATP in myocardial tissues. Notably, irradiated myocardial cells exhibited structural damage and underwent apoptosis. Furthermore, Western blot analysis revealed significant changes in expression levels of proteins involved in oxidative stress regulation and apoptotic signaling pathways, indicating that microwave irradiation could induce myocardial cell apoptosis by interfering with oxidative stress and cardiac energy metabolism. Our findings provide useful insights into the mechanism of microwave-induced damage to the cardiovascular system.


Subject(s)
Apoptosis/physiology , Apoptosis/radiation effects , Microwaves/adverse effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/radiation effects , Animals , Antioxidants/metabolism , Antioxidants/radiation effects , Dose-Response Relationship, Radiation , Male , Malondialdehyde/metabolism , Malondialdehyde/radiation effects , Myocytes, Cardiac/pathology , Oxidative Stress/physiology , Oxidative Stress/radiation effects , Rats , Rats, Wistar
18.
Gen Physiol Biophys ; 35(2): 121-30, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26830130

ABSTRACT

Although high-intensity electric fields (HEF) application is currently the only effective therapy available to terminate ventricular fibrillation, it may cause injury to cardiac cells. In this study we determined the relation between HEF pulse length and cardiomyocyte lethal injury. We obtained lethality curves by survival analysis, which were used to determine the value of HEF necessary to kill 50% of cells (E50) and plotted a strength-duration (SxD) curve for lethality with 10 different durations: 0.1, 0.2, 0.5, 1, 3, 5, 10, 20, 35 and 70 ms. For the same durations we also obtained an SxD curve for excitation and established an indicator for stimulatory safeness (stimulation safety factor - SSF) as the ratio between the SxD curve for lethality and one for excitation. We found that the lower the pulse duration, the higher the HEF intensity required to cell death. Contrary to expectations, the highest SSF value does not correspond to the lowest pulse duration but to the one of 0.5 ms. As defibrillation threshold has been described as duration-dependent, our results imply that the use of shorter stimulus duration - instead of the one typically used in the clinic (10 ms) - might increase defibrillation safeness.


Subject(s)
Apoptosis/physiology , Cell Fractionation/methods , Electric Stimulation/methods , Heart Ventricles/cytology , Myocytes, Cardiac/physiology , Myocytes, Cardiac/radiation effects , Animals , Apoptosis/radiation effects , Cells, Cultured , Dose-Response Relationship, Radiation , Electromagnetic Fields , Electroporation , Heart Ventricles/radiation effects , Male , Myocytes, Cardiac/cytology , Radiation Dosage , Rats , Rats, Wistar
19.
Radiats Biol Radioecol ; 56(2): 171-6, 2016.
Article in Russian | MEDLINE | ID: mdl-27534068

ABSTRACT

It has been shown that a single exposure to 171 MHz electromagnetic field with 180 V/m electric field strength and 0.04 mW/kg specific absorption rate significantly alters the Na+/Ca2+ exchange in the isolated rat heart. It is assumed that enhancement of the Na+/Ca2+ exchange towards removing Ca2+ from the cardiomyocytes electromagnetic field exposure is a result of Ca2+ extraction from the sarcoplasmic reticulum and the increase of its intracellular level.


Subject(s)
Electromagnetic Fields , Heart/radiation effects , Myocytes, Cardiac/radiation effects , Radio Waves , Animals , Calcium/metabolism , Myocytes, Cardiac/metabolism , Organ Culture Techniques , Rats , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/radiation effects , Sodium/metabolism
20.
Am J Physiol Heart Circ Physiol ; 309(11): H1947-63, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26408534

ABSTRACT

There are 160,000 cancer patients worldwide treated with particle radiotherapy (RT). With the advent of proton, and high (H) charge (Z) and energy (E) HZE ionizing particle RT, the cardiovascular diseases risk estimates are uncertain. In addition, future deep space exploratory-type missions will expose humans to unknown but low doses of particle irradiation (IR). We examined molecular responses using transcriptome profiling in left ventricular murine cardiomyocytes isolated from mice that were exposed to 90 cGy, 1 GeV proton ((1)H) and 15 cGy, 1 GeV/nucleon iron ((56)Fe) over 28 days after exposure. Unsupervised clustering analysis of gene expression segregated samples according to the IR response and time after exposure, with (56)Fe-IR showing the greatest level of gene modulation. (1)H-IR showed little differential transcript modulation. Network analysis categorized the major differentially expressed genes into cell cycle, oxidative responses, and transcriptional regulation functional groups. Transcriptional networks identified key nodes regulating expression. Validation of the signal transduction network by protein analysis and gel shift assay showed that particle IR clearly regulates a long-lived signaling mechanism for ERK1/2, p38 MAPK signaling and identified NFATc4, GATA4, STAT3, and NF-κB as regulators of the response at specific time points. These data suggest that the molecular responses and gene expression to (56)Fe-IR in cardiomyocytes are unique and long-lasting. Our study may have significant implications for the efforts of National Aeronautics and Space Administration to develop heart disease risk estimates for astronauts and for patients receiving conventional and particle RT via identification of specific HZE-IR molecular markers.


Subject(s)
Gene Regulatory Networks/radiation effects , Iron Radioisotopes/toxicity , Myocytes, Cardiac/radiation effects , Radiotherapy, High-Energy/adverse effects , Signal Transduction/radiation effects , Animals , Cells, Cultured , Cluster Analysis , Enzyme Activation , Fibrosis , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation/radiation effects , Male , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Risk Assessment , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Time Factors , Transcription, Genetic/radiation effects , Transcriptome/radiation effects , Whole-Body Irradiation
SELECTION OF CITATIONS
SEARCH DETAIL