Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.124
Filter
1.
Biosensors (Basel) ; 14(5)2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38785693

ABSTRACT

Zebrafish larvae have emerged as a valuable model for studying heart physiology and pathophysiology, as well as for drug discovery, in part thanks to its transparency, which simplifies microscopy. However, in fluorescence-based optical mapping, the beating of the heart results in motion artifacts. Two approaches have been employed to eliminate heart motion during calcium or voltage mapping in zebrafish larvae: the knockdown of cardiac troponin T2A and the use of myosin inhibitors. However, these methods disrupt the mechano-electric and mechano-mechanic coupling mechanisms. We have used ratiometric genetically encoded biosensors to image calcium in the beating heart of intact zebrafish larvae because ratiometric quantification corrects for motion artifacts. In this study, we found that halting heart motion by genetic means (injection of tnnt2a morpholino) or chemical tools (incubation with para-aminoblebbistatin) leads to bradycardia, and increases calcium levels and the size of the calcium transients, likely by abolishing a feedback mechanism that connects contraction with calcium regulation. These outcomes were not influenced by the calcium-binding domain of the gene-encoded biosensors employed, as biosensors with a modified troponin C (Twitch-4), calmodulin (mCyRFP1-GCaMP6f), or the photoprotein aequorin (GFP-aequorin) all yielded similar results. Cardiac contraction appears to be an important regulator of systolic and diastolic Ca2+ levels, and of the heart rate.


Subject(s)
Biosensing Techniques , Calcium , Larva , Myocardial Contraction , Zebrafish , Animals , Calcium/metabolism , Myocardial Contraction/physiology , Heart/physiology , Troponin T/metabolism , Zebrafish Proteins/metabolism , Troponin C/metabolism
2.
Sci Rep ; 14(1): 11081, 2024 05 15.
Article in English | MEDLINE | ID: mdl-38744867

ABSTRACT

Despite progress in generating cardiomyocytes from pluripotent stem cells, these populations often include non-contractile cells, necessitating cardiomyocyte selection for experimental purpose. This study explores a novel cardiomyocyte enrichment mechanism: low-adhesion culture selection. The cardiac cells derived from human induced pluripotent stem cells were subjected to a coating-free low-adhesion culture using bovine serum albumin and high molecular weight dextran sulfate. This approach effectively increased the population of cardiac troponin T-positive cardiomyocytes. Similar results were obtained with commercially available low-adhesion culture dishes. Subsequently, we accessed the practicality of selection of cardiomyocytes using this phenomenon by comparing it with established methods such as glucose-free culture and selection based on puromycin resistance genes. The cardiomyocytes enriched through low-adhesion culture selection maintained autonomous pulsation and responsiveness to beta-stimuli. Moreover, no significant differences were observed in the expression of genes related to subtype commitment and maturation when compared to other selection methods. In conclusion, cardiomyocytes derived from pluripotent stem cells were more low-adhesion culture resistant than their accompanying non-contractile cells, and low-adhesion culture is an alternative method for selection of pluripotent stem cell-derived cardiomyocytes.


Subject(s)
Cell Adhesion , Cell Culture Techniques , Cell Differentiation , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Cell Culture Techniques/methods , Cells, Cultured , Troponin T/metabolism , Troponin T/genetics
3.
Med ; 5(4): 335-347.e3, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38521068

ABSTRACT

BACKGROUND: Vaccine-related acute myocarditis is recognized as a rare and specific vaccine complication following mRNA-based COVID-19 vaccinations. The precise mechanisms remain unclear. We hypothesized that natural killer (NK) cells play a central role in its pathogenesis. METHODS: Samples from 60 adolescents with vaccine-related myocarditis were analyzed, including pro-inflammatory cytokines, cardiac troponin T, genotyping, and immunophenotyping of the corresponding activation subsets of NK cells, monocytes, and T cells. Results were compared with samples from 10 vaccinated individuals without myocarditis and 10 healthy controls. FINDINGS: Phenotypically, high levels of serum cytokines pivotal for NK cells, including interleukin-1ß (IL-1ß), interferon α2 (IFN-α2), IL-12, and IFN-γ, were observed in post-vaccination patients with myocarditis, who also had high percentage of CD57+ NK cells in blood, which in turn correlated positively with elevated levels of cardiac troponin T. Abundance of the CD57+ NK subset was particularly prominent in males and in those after the second dose of vaccination. Genotypically, killer cell immunoglobulin-like receptor (KIR) KIR2DL5B(-)/KIR2DS3(+)/KIR2DS5(-)/KIR2DS4del(+) was a risk haplotype, in addition to single-nucleotide polymorphisms related to the NK cell-specific expression quantitative trait loci DNAM-1 and FuT11, which also correlated with cardiac troponin T levels in post-vaccination patients with myocarditis. CONCLUSION: Collectively, these data suggest that NK cell activation by mRNA COVID-19 vaccine contributed to the pathogenesis of acute myocarditis in genetically and epidemiologically vulnerable subjects. FUNDING: This work was funded by the Hong Kong Collaborative Research Fund (CRF) 2020/21 and the CRF Coronavirus and Novel Infectious Diseases Research Exercises (reference no. C7149-20G).


Subject(s)
COVID-19 , Myocarditis , Male , Adolescent , Humans , Myocarditis/etiology , Myocarditis/metabolism , COVID-19 Vaccines/adverse effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Troponin T/metabolism , Interferon-gamma/metabolism , COVID-19/prevention & control , Killer Cells, Natural/metabolism , Cytokines/metabolism , Vaccination/adverse effects , Receptors, KIR2DL5/metabolism
4.
Environ Toxicol ; 39(4): 2064-2076, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38095131

ABSTRACT

OBJECTIVE: We aimed to determine the role of Troponin T1 (TNNT1) in paclitaxel (PTX) resistance and tumor progression in breast cancer (BC). METHODS: Differentially expressed genes were obtained from the GSE4298 and GSE90564 datasets. Hub genes were isolated from protein-protein interaction networks and further validated by real-time quantitative polymerase chain reaction. The effect of TNNT1 on PTX resistance was determined using cell counting kit-8, 5-ethynyl-2'-deoxyuridine, wound healing, transwell, flow cytometry assays, and subcutaneous xenografted tumor model. Western blotting was used to detect proteins associated with PTX resistance, apoptosis, migration, invasion, and other key pathways. Hematoxylin-eosin and immunohistochemical staining were used to evaluate the role of TNNT1 in tumors. RESULTS: After comprehensive bioinformatic analysis, we identified CCND1, IGF1, SFN, INHBA, TNNT1, and TNFSF11 as hub genes for PTX resistance in BC. TNNT1 plays a key role in BC and is upregulated in PTX-resistant BC cells. TNNT1 silencing inhibited PTX resistance, proliferation, migration, and invasion while promoting apoptosis of PTX-resistant BC cells. Tumor xenograft experiments revealed that TNNT1 silencing suppresses PTX resistance and tumor development in vivo. In addition, TNNT1 silencing inhibited the expression of proteins in the rat sarcoma virus (RAS)/rapidly accelerated fibrosarcoma1 (RAF1) pathway in vivo. Treatment with a RAS/RAF1 pathway activator reversed the inhibitory effect of TNNT1 silencing on proliferation, migration, and invasion while promoting apoptosis of PTX resistance BC cells. CONCLUSION: Silencing of TNNT1 suppresses PTX resistance and BC progression by inhibiting the RAS/RAF1 pathway, which is a promising biomarker and therapeutic target for drug resistance in BC.


Subject(s)
Breast Neoplasms , Fibrosarcoma , MicroRNAs , Humans , Female , Paclitaxel/pharmacology , Breast Neoplasms/pathology , Troponin T/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Proto-Oncogene Proteins p21(ras)/pharmacology , Proto-Oncogene Proteins p21(ras)/therapeutic use , Drug Resistance, Neoplasm/genetics , Apoptosis/genetics , Cell Line, Tumor , Fibrosarcoma/genetics , Fibrosarcoma/drug therapy , Cell Proliferation , MicroRNAs/genetics
5.
Free Radic Biol Med ; 211: 145-157, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38043869

ABSTRACT

It is generally accepted that oxidative stress plays a key role in the development of ischemia-reperfusion injury in ischemic heart disease. However, the mechanisms how reactive oxygen species trigger cellular damage are not fully understood. Our study investigates redox state and highly reactive substances within neonatal and adult cardiomyocytes under hypoxia conditions. We have found that hypoxia induced an increase in H2O2 production in adult cardiomyocytes, while neonatal cardiomyocytes experienced a decrease in H2O2 levels. This finding correlates with our observation of the difference between the electron transport chain (ETC) properties and mitochondria amount in adult and neonatal cells. We demonstrated that in adult cardiomyocytes hypoxia caused the significant increase in the ETC loading with electrons compared to normoxia. On the contrary, in neonatal cardiomyocytes ETC loading with electrons was similar under both normoxic and hypoxic conditions that could be due to ETC non-functional state and the absence of the electrons transfer to O2 under normoxia. In addition to the variations in H2O2 production, we also noted consistent pH dynamics under hypoxic conditions. Notably, the pH levels exhibited a similar decrease in both cell types, thus, acidosis is a more universal cellular response to hypoxia. We also demonstrated that the amount of mitochondria and the levels of cardiac isoforms of troponin I, troponin T, myoglobin and GAPDH were significantly higher in adult cardiomyocytes compared to neonatal ones. Remarkably, we found out that under hypoxia, the levels of cardiac isoforms of troponin T, myoglobin, and GAPDH were elevated in adult cardiomyocytes, while their level in neonatal cells remained unchanged. Obtained data contribute to the understanding of the mechanisms of neonatal cardiomyocytes' resistance to hypoxia and the ability to maintain the metabolic homeostasis in contrast to adult ones.


Subject(s)
Hydrogen Peroxide , Myocytes, Cardiac , Rats , Animals , Myocytes, Cardiac/metabolism , Hydrogen Peroxide/metabolism , Myoglobin , Troponin T/metabolism , Cell Hypoxia , Hypoxia/metabolism , Oxidation-Reduction , Protein Isoforms/metabolism
6.
Cardiology ; 148(6): 506-516, 2023.
Article in English | MEDLINE | ID: mdl-37544298

ABSTRACT

INTRODUCTION: N-terminal pro-B-type natriuretic peptide (NT-proBNP) and cardiac troponin T (cTnT) measurements are recommended in patients with acute dyspnea. We aimed to assess the prognostic merit of cTnT compared to NT-proBNP for 30-day readmission or death in patients hospitalized with acute dyspnea. METHODS: We measured cTnT and NT-proBNP within 24 h in 314 patients hospitalized with acute dyspnea and adjudicated the cause of the index admission. Time to first event of readmission or death ≤30 days after hospital discharge was recorded, and cTnT and NT-proBNP measurements were compared head-to-head. RESULTS: Patients who died (12/314) or were readmitted (71/314) within 30 days had higher cTnT concentrations (median: 32.6, Q1-Q3: 18.4-74.2 ng/L vs. median: 19.4, Q1-Q3: 8.4-36.1 ng/L; p for comparison <0.001) and NT-proBNP concentrations (median: 1,753.6, Q1-Q3: 464.2-6,862.0 ng/L vs. median 984, Q1-Q3 201-3,600 ng/L; for comparison p = 0.027) compared to patients who survived and were not readmitted. cTnT concentrations were associated with readmission or death within 30 days after discharge both in the total cohort (adjusted hazard ratio [aHR]: 1.64, 95% confidence interval [CI]: 1.30-2.05) and in patients with heart failure (HF) (aHR: 1.58, 95% CI: 1.14-2.18). In contrast, NT-proBNP concentrations were not associated with short-term events, neither in the total cohort (aHR: 1.10, 95% CI: 0.94-1.30) nor in patients with adjudicated HF (aHR: 1.06, 95% CI: 0.80-1.40). CONCLUSION: cTnT concentrations are associated with 30-day readmission or death in patients hospitalized with acute dyspnea, as well as in patients adjudicated HF.


Subject(s)
Dyspnea , Natriuretic Peptide, Brain , Patient Readmission , Troponin T , Troponin T/blood , Troponin T/metabolism , Natriuretic Peptide, Brain/blood , Natriuretic Peptide, Brain/metabolism , Patient Readmission/statistics & numerical data , Dyspnea/blood , Dyspnea/diagnosis , Dyspnea/mortality , Predictive Value of Tests , Humans , Male , Female , Middle Aged , Aged , Aged, 80 and over , Biomarkers/blood , Biomarkers/metabolism , Kaplan-Meier Estimate
7.
Philos Trans R Soc Lond B Biol Sci ; 378(1879): 20220176, 2023 06 19.
Article in English | MEDLINE | ID: mdl-37122209

ABSTRACT

In this perspective, we discussed emerging data indicating a role for Notch signalling in inherited disorders of the heart failure with focus on hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) linked to variants of genes encoding mutant proteins of the sarcomere. We recently reported an upregulation of elements in the Notch signalling cascade in cardiomyocytes derived from human inducible pluripotent stem cells expressing a TNNT2 variant encoding cardiac troponin T (cTnT-I79N+/-), which induces hypertrophy, remodelling, abnormalities in excitation-contraction coupling and electrical instabilities (Shafaattalab S et al. 2021 Front. Cell Dev. Biol. 9, 787581. (doi:10.3389/fcell.2021.787581)). Our search of the literature revealed the novelty of this finding and stimulated us to discuss potential connections between the Notch signalling pathway and familial cardiomyopathies. Our considerations focused on the potential role of these interactions in arrhythmias, microvascular ischaemia, and fibrosis. This finding underscored a need to consider the role of Notch signalling in familial cardiomyopathies which are trigged by sarcomere mutations engaging mechano-signalling pathways for which there is evidence of a role for Notch signalling with crosstalk with Hippo signalling. Our discussion included a role for both cardiac myocytes and non-cardiac myocytes in progression of HCM and DCM. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.


Subject(s)
Cardiomyopathies , Cardiomyopathy, Dilated , Cardiomyopathy, Hypertrophic , Humans , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Hypertrophic/genetics , Troponin T/genetics , Troponin T/metabolism , Hypertrophy , Mutation
8.
Am J Physiol Heart Circ Physiol ; 324(5): H675-H685, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36930654

ABSTRACT

Obesity and hypertension, independently and combined, are associated with increased risk of heart failure and heart failure-related morbidity and mortality. Interest in circulating endothelial cell-derived microvesicles (EMVs) has intensified because of their involvement in the development and progression of endothelial dysfunction, atherosclerosis, and cardiomyopathy. The experimental aim of this study was to determine, in vitro, the effects of EMVs isolated from obese/hypertensive adults on key proteins regulating cardiomyocyte hypertrophy [cardiac troponin T (cTnT), α-actinin, nuclear factor-kB (NF-kB)] and fibrosis [transforming growth factor (TGF)-ß, collagen1-α1], as well as endothelial nitric oxide synthase (eNOS) expression and nitric oxide (NO) production. EMVs (CD144+ microvesicles) were isolated from plasma by flow cytometry in 12 normal weight/normotensive [8 males/4 females; age: 56 ± 5 yr; body mass index (BMI): 23.3 ± 2.0 kg/m2; blood pressure (BP): 117/74 ± 4/5 mmHg] and 12 obese/hypertensive (8 males/4 females; 57 ± 5 yr; 31.7 ± 1.8 kg/m2; 138/83 ± 8/7 mmHg) adults. Human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) were cultured and treated with EMVs from either normal weight/normotensive or obese/hypertensive adults for 24 h. Expression of cTnT (64.1 ± 13.9 vs. 29.5 ± 7.8 AU), α-actinin (66.0 ± 14.7 vs. 36.2 ± 10.3 AU), NF-kB (166.3 ± 13.3 vs. 149.5 ± 8.8 AU), phosphorylated-NF-kB (226.1 ± 25.2 vs. 179.1 ± 25.5 AU), and TGF-ß (62.1 ± 13.3 vs. 23.5 ± 8.8 AU) were significantly higher and eNOS activation (16.4 ± 4.3 vs. 24.8 ± 3.7 AU) and nitric oxide production (6.8 ± 1.2 vs. 9.6 ± 1.3 µmol/L) were significantly lower in iPSC-CMs treated with EMVs from obese/hypertensive compared with normal weight/normotensive adults. These data indicate that EMVs from obese/hypertensive adults induce a cardiomyocyte phenotype prone to hypertrophy, fibrosis, and reduced nitric oxide production, central factors associated with heart failure risk and development.NEW & NOTEWORTHY In the present study we determined the effect of endothelial microvesicles (EMVs) isolated from obese/hypertensive adults on mediators of cardiomyocyte hypertrophy [cardiac troponin T (cTnT), α-actinin, nuclear factor-kB (NF-kB)] and fibrosis [transforming growth factor (TGF-ß), collagen1-α1] as well as endothelial nitric oxide synthase (eNOS) expression and NO production. EMVs from obese/hypertensive induced significantly higher expression of hypertrophic (cTnT, α-actinin, NF-kB) and fibrotic (TGF-ß) proteins as well as significantly lower eNOS activation and NO production in cardiomyocytes than EMVs from normal weight/normotensive adults. EMVs are a potential mediating factor in the increased risk of cardiomyopathy and heart failure with obesity/hypertension.


Subject(s)
Cell-Derived Microparticles , Heart Failure , Hypertension , Male , Female , Humans , Adult , Middle Aged , Myocytes, Cardiac/metabolism , Nitric Oxide Synthase Type III/metabolism , Troponin T/metabolism , Nitric Oxide/metabolism , Actinin/metabolism , Actinin/pharmacology , NF-kappa B/metabolism , Hypertension/metabolism , Hypertrophy/metabolism , Hypertrophy/pathology , Cell-Derived Microparticles/metabolism , Obesity/metabolism , Heart Failure/metabolism , Transforming Growth Factor beta/metabolism , Fibrosis
9.
Stem Cell Res ; 67: 103048, 2023 03.
Article in English | MEDLINE | ID: mdl-36801602

ABSTRACT

Cardiac muscle troponin T protein binds to tropomyosin and regulates the calcium-dependent actin-myosin interaction on thin filaments in cardiomyocytes. Recent genetic studies have revealed that TNNT2 mutations are strongly linked to dilated cardiomyopathy (DCM). In this study, we generated YCMi007-A, a human induced pluripotent stem cell (hiPSC) line from a DCM patient with a p. Arg205Trp mutation in the TNNT2 gene. The YCMi007-A cells show high expression of pluripotent markers, normal karyotype, and differentiation into three germ layers. Thus, YCMi007-A-an established iPSC-could be useful for the investigation of DCM.


Subject(s)
Cardiomyopathy, Dilated , Induced Pluripotent Stem Cells , Humans , Cardiomyopathy, Dilated/genetics , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Troponin T/genetics , Troponin T/metabolism , Heterozygote , Mutation
10.
Biofactors ; 49(2): 351-364, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36518005

ABSTRACT

The cardiac troponins (cTns), cardiac troponin C (cTnC), cTnT, and cTnI are key elements of myocardial apparatus, fixed as protein complex on the thin filament of sarcomere and are involved in the regulation of excitation-contraction coupling of cardiomyocytes in the presence of Ca2+ . Circulating cTnT and cTnI (cTns) increase following cardiac tissue necrosis, and they are consolidated biomarkers of acute myocardial infarction (AMI). However, the use of high sensitivity (hs)-immunoassay tests for cTnT and cTnI has made it possible to identify a multitude of other clinical conditions associated with increased circulating levels of cTns. cTns can be measured also in the peripheral circulation of healthy subjects or athletes, suggesting that different mechanisms are involved in the release of cTns in the blood independently of cardiac cell necrosis. In this review, the molecular/cellular mechanisms involved in cTns release in blood and the exploitation of cTnI and cTnT as biomarkers of cardiac adverse events, in addition to cardiac necrosis, are discussed.


Subject(s)
Myocardial Infarction , Humans , Troponin T/metabolism , Troponin I/metabolism , Biomarkers , Necrosis
11.
Food Chem ; 404(Pt B): 134647, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36283311

ABSTRACT

In this study, the effects of different oxidation intensities on the degradation of myofibrillar protein by endogenous enzymes in iron-catalyzed oxidizing (IOS) and metmyoglobin oxidizing system (MOS) were compared. The results showed that carbonyl content and endogenous enzyme activities (caspase-3, caspase-6 and calpain-1) increased significantly and the total sulfhydryl content decreased significantly with H2O2 concentration in both oxidation systems. Meanwhile, the rate of carbonyl formation and the inhibition of endogenous enzymes activities of IOS were significantly lower than MOS for the same oxidation intensity. In addition, IOS and MOS mainly produced myosin light chains degradation products of 20-25 kDa and 20-17 kDa. At the same oxidation intensity, MOS of myofibrillar protein significantly enhanced the degradation of troponin-T and desmin by caspase-3/-6 compared with IOS, while inhibiting the degradation of troponin-T by calpain-1. In conclusion, MOS inhibited endogenous enzyme degradation in vitro more than IOS during post-slaughter maturation of yak meat.


Subject(s)
Calpain , Myofibrils , Animals , Cattle , Proteolysis , Myofibrils/metabolism , Calpain/metabolism , Caspase 3/metabolism , Methemoglobin/metabolism , Troponin T/metabolism , Iron/metabolism , Hydrogen Peroxide/metabolism , Muscle, Skeletal/metabolism , Meat/analysis , Oxidation-Reduction , Metmyoglobin/metabolism , Catalysis
12.
Insect Mol Biol ; 32(1): 46-55, 2023 02.
Article in English | MEDLINE | ID: mdl-36214335

ABSTRACT

Insects are the most widely distributed and successful animals on the planet. A large number of insects are capable of flight with functional wings. Wing expansion is an important process for insects to achieve functional wings after eclosion and healthy genital morphology is crucial for adult reproduction. Myofilaments are functional units that constitute sarcomeres and trigger muscle contraction. Here, we identified four myofilament proteins, including Myosin, Paramyosin, Tropomyosin and Troponin T, from the wing pads of nymphs in the American cockroach, Periplaneta americana. RNAi-mediated knockdown of Myosin, Paramyosin, Tropomyosin and Troponin T in the early stage of final instar nymphs caused a severely curly wing phenotype in the imaginal moult, especially in the Paramyosin and Troponin T knockdown groups, indicating that these myofilament proteins are involved in controlling wing expansion behaviours during the nymph-adult transition. In addition, the knockdown resulted in abnormal external genitalia, caused ovulation failure, and affected male accessory gland development. Interestingly, the expression of myofilament genes was induced by methoprene, a juvenile hormone (JH) analogue, and decreased by the depletion of the JH receptor gene Met. Altogether, we have determined that myofilament genes play an important role in promoting wing expansion and maintaining adult genitalia morphology, and their expression is induced by JH signalling. Our data reveal a novel mechanism by which wing expansion is regulated by myofilaments and the functions of myofilaments are involved in maintaining genitalia morphology.


Subject(s)
Periplaneta , Female , Male , Animals , Periplaneta/genetics , Periplaneta/metabolism , Myofibrils , Tropomyosin/genetics , Tropomyosin/metabolism , Troponin T/metabolism , Metamorphosis, Biological , Insecta , Juvenile Hormones/metabolism , Nymph
13.
Int J Mol Sci ; 23(24)2022 Dec 11.
Article in English | MEDLINE | ID: mdl-36555368

ABSTRACT

Tropomyosin (Tpm) mutations cause inherited cardiac diseases such as hypertrophic and dilated cardiomyopathies. We applied various approaches to investigate the role of cardiac troponin (Tn) and especially the troponin T (TnT) in the pathogenic effects of Tpm cardiomyopathy-associated mutations M8R, K15N, A277V, M281T, and I284V located in the overlap junction of neighboring Tpm dimers. Using co-sedimentation assay and viscosity measurements, we showed that TnT1 (fragment of TnT) stabilizes the overlap junction of Tpm WT and all Tpm mutants studied except Tpm M8R. However, isothermal titration calorimetry (ITC) indicated that TnT1 binds Tpm WT and all Tpm mutants similarly. By using ITC, we measured the direct KD of the Tpm overlap region, N-end, and C-end binding to TnT1. The ITC data revealed that the Tpm C-end binds to TnT1 independently from the N-end, while N-end does not bind. Therefore, we suppose that Tpm M8R binds to TnT1 without forming the overlap junction. We also demonstrated the possible role of Tn isoform composition in the cardiomyopathy development caused by M8R mutation. TnT1 dose-dependently reduced the velocity of F-actin-Tpm filaments containing Tpm WT, Tpm A277V, and Tpm M281T mutants in an in vitro motility assay. All mutations impaired the calcium regulation of the actin-myosin interaction. The M281T and I284V mutations increased the calcium sensitivity, while the K15N and A277V mutations reduced it. The Tpm M8R, M281T, and I284V mutations under-inhibited the velocity at low calcium concentrations. Our results demonstrate that Tpm mutations likely implement their pathogenic effects through Tpm interaction with Tn, cardiac myosin, or other protein partners.


Subject(s)
Cardiomyopathies , Tropomyosin , Troponin , Humans , Actins/metabolism , Calcium/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Mutation , Tropomyosin/genetics , Troponin/genetics , Troponin T/metabolism
14.
Sci Rep ; 12(1): 22501, 2022 12 28.
Article in English | MEDLINE | ID: mdl-36577774

ABSTRACT

Cardiomyopathies have unresolved genotype-phenotype relationships and lack disease-specific treatments. Here we provide a framework to identify genotype-specific pathomechanisms and therapeutic targets to accelerate the development of precision medicine. We use human cardiac electromechanical in-silico modelling and simulation which we validate with experimental hiPSC-CM data and modelling in combination with clinical biomarkers. We select hypertrophic cardiomyopathy as a challenge for this approach and study genetic variations that mutate proteins of the thick (MYH7R403Q/+) and thin filaments (TNNT2R92Q/+, TNNI3R21C/+) of the cardiac sarcomere. Using in-silico techniques we show that the destabilisation of myosin super relaxation observed in hiPSC-CMs drives disease in virtual cells and ventricles carrying the MYH7R403Q/+ variant, and that secondary effects on thin filament activation are necessary to precipitate slowed relaxation of the cell and diastolic insufficiency in the chamber. In-silico modelling shows that Mavacamten corrects the MYH7R403Q/+ phenotype in agreement with hiPSC-CM experiments. Our in-silico model predicts that the thin filament variants TNNT2R92Q/+ and TNNI3R21C/+ display altered calcium regulation as central pathomechanism, for which Mavacamten provides incomplete salvage, which we have corroborated in TNNT2R92Q/+ and TNNI3R21C/+ hiPSC-CMs. We define the ideal characteristics of a novel thin filament-targeting compound and show its efficacy in-silico. We demonstrate that hybrid human-based hiPSC-CM and in-silico studies accelerate pathomechanism discovery and classification testing, improving clinical interpretation of genetic variants, and directing rational therapeutic targeting and design.


Subject(s)
Cardiomyopathy, Hypertrophic , Precision Medicine , Humans , Mutation , Myosin Heavy Chains/genetics , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/therapy , Cardiomyopathy, Hypertrophic/metabolism , Troponin T/metabolism , Troponin I/genetics
15.
Sci Rep ; 12(1): 17554, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36266413

ABSTRACT

Myocardial injury influenced by cisplatin (Cis) is a compelling reason to hunt out a treatment modality to overcome such a threat in cisplatin-treated patients. Breast Milk mesenchymal stem cells (Br-MSCs) are a non-invasive, highly reproducible source of stem cells. Herein, we investigate Br-MSCs' role in cardiotoxicity induced by cisplatin. Rats were divided into; control, Cis-treated (received 12 mg/kg single intraperitoneal injection), BrMSCs-treated (received single intraperitoneal injection of 0.5 ml sterilized phosphate-buffered saline containing 2 × 107 cells of Br-MSCs); metformin-treated (received 250 mg/kg/day orally) and BrMSCs + metformin + Cis treated groups. At the experiment end, serum creatine kinase (CK-MB) and cardiac troponin T (cTnT) activates were estimated, cardiac malondialdehyde (MDA), superoxide dismutase (SOD), interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α) levels were measured, cardiac expression of Bax and Bcl-2 and AMP-activated protein kinase (AMPK), as well as heart histopathology, were evaluated. Study results showed that Cis explored acute cardiotoxicity evidenced by deteriorated cardiac indices, induction of oxidative stress, and inflammation with myocardium histological alterations. Treatment with Br-MSCs restored heart function and structure deteriorated by Cis injection. The antioxidant/anti-inflammatory/anti-apoptotic results of Br-MSCs were supported by AMPK activation denoting their protective role against cisplatin-induced cardiac injury. These results were superior when metformin was added to the treatment protocol.


Subject(s)
Cardiotoxicity , Cisplatin , Mesenchymal Stem Cells , Metformin , Humans , Male , Rats , AMP-Activated Protein Kinases/metabolism , Anti-Inflammatory Agents/pharmacology , Antioxidants/metabolism , Apoptosis , bcl-2-Associated X Protein/metabolism , Cardiotoxicity/therapy , Cisplatin/toxicity , Creatine Kinase, MB Form/metabolism , Malondialdehyde/metabolism , Mesenchymal Stem Cells/cytology , Metformin/pharmacology , Milk, Human/cytology , Oxidative Stress , Superoxide Dismutase/metabolism , Troponin T/metabolism , Tumor Necrosis Factor-alpha/metabolism
16.
J Food Sci ; 87(10): 4440-4452, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36102040

ABSTRACT

Control on the moisture distribution, protein structure changes, and protein degradation of Antarctic krill meat during freeze-thaw (F-T) cycles by presoaking with antifreeze protein (AFP) was investigated. The results from the thawing loss rate and cooking loss rate indicated that 0.1% was the optimal AFP concentration. Magnetic resonance imaging and low-field nuclear magnetic resonance results showed that AFP inhibited the changes in moisture distribution and maintained the moisture in Antarctic krill meat. The contents of nonprotein nitrogen and trichloroacetic acid-soluble peptides indicated that AFP reduced protein degradation. Further, SDS-PAGE showed that AFP reduced the degradation of actin, troponin T, and myosin light chain. The results of fluorescence spectra, circular dichroism, and chemical bond contents indicated that AFP reduced the damage of the protein tertiary and secondary structures of Antarctic krill meat by holding it in a weak polar environment. This study supplied basic theory for the quality control of Antarctic krill meat. PRACTICAL APPLICATION: Protein degradation, moisture distribution, and protein structure changes occurred to Antarctic krill meat during freeze-thaw cycles due to ice crystal growth and recrystallization, which leads to the decrease in quality. Antifreeze protein has been proven to avoid ice crystals' growth and inhibit ice recrystallization. During freeze-thaw cycles, the moisture distribution of Antarctic krill meat treated with antifreeze protein was more uniform, the degree of protein degradation was lower, and the protein structure was protected. This study demonstrated the potential of antifreeze protein as a water and protein protectant of Antarctic krill meat during freeze-thaw cycles.


Subject(s)
Euphausiacea , Animals , Ice , Troponin T/metabolism , Actins/metabolism , Myosin Light Chains/metabolism , Trichloroacetic Acid , alpha-Fetoproteins/metabolism , Antifreeze Proteins/metabolism , Meat/analysis , Nitrogen/metabolism
17.
Shock ; 58(2): 147-157, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35953459

ABSTRACT

ABSTRACT: Hypoxia/reoxygenation (H/R) induces pyroptosis in the setting of acute myocardial infarction (AMI). Previous studies have shown that the expression of the miR-15 family is stimulated in myocardial ischemia-reperfusion injury or H/R-induced cardiomyocyte injury, and miR-15 is a promoter of cardiac ischemia-reperfusion or H/R injury. However, whether miR-15b-5p regulates H/R injury and cardiomyocyte pyroptosis and its mechanism still need to be further clarified. Bioinformatics analysis elicited that SIRT3 was the downstream regulatory target gene of miR-15b-5p. SIRT3 has been shown to participate in the regulation of pyroptosis by negatively regulating the NLRP3 inflammasome pathway. Therefore, we hypothesized that miR-15b-5p targets SIRT3 and activated the NLRP3 inflammasome pathway to promote H/R-induced cardiomyocyte pyroptosis. We first show that H/R increases miR-15b-5p in rat cardiomyocytes H9C2. Next, we tested the effects of inhibition of miR-15b-5p or overexpression of SIRT3. We found that miR-15b-5p downregulation or SIRT3 overexpression could reverse the H/R-induced pyroptosis. Furthermore, silencing SIRT3 antagonized the protective effect of miR-15b-5p downregulation on H9C2 cells. NLRP3 inhibitor MCC950 annulled the previously mentioned antagonistic effect of silencing SIRT3 on the protection of miR-15b-5p downregulation against pyroptosis. We then used a rat AMI model to analyze myocardial infarction area by triphenyl tetrazolium chloride staining and assess serum cardiac troponin T level by ELISA and found that miR-15b-5p silencing reduced AMI injury in rats. Collectively, these results suggest that miR-15b-5p increase H/R-induced pyroptosis in cardiomyocytes by targeting SIRT3 and activating the NLRP3 inflammasome.


Subject(s)
MicroRNAs , Sirtuin 3 , Animals , Apoptosis , Chlorides/metabolism , Chlorides/pharmacology , Hypoxia/metabolism , Inflammasomes/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Myocytes, Cardiac/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis , Rats , Sirtuin 3/genetics , Sirtuin 3/metabolism , Sirtuin 3/pharmacology , Sirtuins , Troponin T/metabolism
18.
Circulation ; 146(12): 934-954, 2022 09 20.
Article in English | MEDLINE | ID: mdl-35983756

ABSTRACT

BACKGROUND: Cytokines such as tumor necrosis factor-α (TNFα) have been implicated in cardiac dysfunction and toxicity associated with doxorubicin (DOX). Although TNFα can elicit different cellular responses, including survival or death, the mechanisms underlying these divergent outcomes in the heart remain cryptic. The E3 ubiquitin ligase TRAF2 (TNF receptor associated factor 2) provides a critical signaling platform for K63-linked polyubiquitination of RIPK1 (receptor interacting protein 1), crucial for nuclear factor-κB (NF-κB) activation by TNFα and survival. Here, we investigate alterations in TNFα-TRAF2-NF-κB signaling in the pathogenesis of DOX cardiotoxicity. METHODS: Using a combination of in vivo (4 weekly injections of DOX 5 mg·kg-1·wk-1) in C57/BL6J mice and in vitro approaches (rat, mouse, and human inducible pluripotent stem cell-derived cardiac myocytes), we monitored TNFα levels, lactate dehydrogenase, cardiac ultrastructure and function, mitochondrial bioenergetics, and cardiac cell viability. RESULTS: In contrast to vehicle-treated mice, ultrastructural defects, including cytoplasmic swelling, mitochondrial perturbations, and elevated TNFα levels, were observed in the hearts of mice treated with DOX. While investigating the involvement of TNFα in DOX cardiotoxicity, we discovered that NF-κB was readily activated by TNFα. However, TNFα-mediated NF-κB activation was impaired in cardiac myocytes treated with DOX. This coincided with loss of K63- linked polyubiquitination of RIPK1 from the proteasomal degradation of TRAF2. Furthermore, TRAF2 protein abundance was markedly reduced in hearts of patients with cancer treated with DOX. We further established that the reciprocal actions of the ubiquitinating and deubiquitinating enzymes cellular inhibitors of apoptosis 1 and USP19 (ubiquitin-specific peptidase 19), respectively, regulated the proteasomal degradation of TRAF2 in DOX-treated cardiac myocytes. An E3-ligase mutant of cellular inhibitors of apoptosis 1 (H588A) or gain of function of USP19 prevented proteasomal degradation of TRAF2 and DOX-induced cell death. Furthermore, wild-type TRAF2, but not a RING finger mutant defective for K63-linked polyubiquitination of RIPK1, restored NF-κB signaling and suppressed DOX-induced cardiac cell death. Last, cardiomyocyte-restricted expression of TRAF2 (cardiac troponin T-adeno-associated virus 9-TRAF2) in vivo protected against mitochondrial defects and cardiac dysfunction induced by DOX. CONCLUSIONS: Our findings reveal a novel signaling axis that functionally connects the cardiotoxic effects of DOX to proteasomal degradation of TRAF2. Disruption of the critical TRAF2 survival pathway by DOX sensitizes cardiac myocytes to TNFα-mediated necrotic cell death and DOX cardiotoxicity.


Subject(s)
Cardiomyopathies , NF-kappa B , TNF Receptor-Associated Factor 2 , Animals , Apoptosis , Cardiomyopathies/metabolism , Cardiotoxicity , Deubiquitinating Enzymes/metabolism , Doxorubicin/toxicity , Endopeptidases , Humans , Lactate Dehydrogenases/metabolism , Mice , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Rats , TNF Receptor-Associated Factor 2/genetics , Troponin T/metabolism , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Proteases/metabolism , Ubiquitin-Specific Proteases/pharmacology
19.
Biomed Pharmacother ; 154: 113583, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35994819

ABSTRACT

The prevalence of cardiovascular complications in diabetes has become one of the major cause of diabetes related morbidity/mortality. The onset and progression of diabetic cardiomyopathy (DCM) has been majorly linked to lipid alterations, oxidative stress, inflammation and apoptosis. This present study investigated the cardioprotective role of Lycium chinense leaf extract (LCME) in fructose/streptozotocin induced diabetic rats. Diabetic animals were orally gavaged with LCME (100 and 400 mg/kg) for five weeks. The results indicated that diabetic rats showed increased blood glucose concentration, serum cardiac function markers (troponin T, creatine kinase-MB, aspartate aminotransferase and lactate dehydrogenase) and lipid profile (triglycerides and cholesterol). In addition, the cardiac tissues of diabetic rats showed increased levels of nuclear factor-κB (NF-κB), tumor necrosis factor alpha (TNF-α), interleukin 1 beta (IL 1ß), interleukin 6 (IL-6), caspase-3 and malondialdehyde as well as significantly reduced activities of catalase, superoxide dismutase, reduced glutathione and glutathione peroxidase. LCME significantly ameliorated hyperglycemia and markedly decreased serum concentrations of troponin T, creatine kinase-MB, aspartate aminotransferase and lactate dehydrogenase, triglycerides and cholesterol. Furthermore, LCME notably suppressed cardiac oxido-inflammatory mediators and boosted cardiac antioxidant defense. Histopathologically, LCME restored cardiac structural alterations and also suppressed the immunohistochemical expression of collagen IV, smooth muscle alpha-actin (α-SMA) and p53, while Bcl2 expression was significantly increased. In conclusion, our result indicated that LCME protected against diabetic cardiomyopathy suppressing oxidative stress, inflammation, apoptosis and fibrosis.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies , Lycium , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Apoptosis , Aspartate Aminotransferases/metabolism , Biomarkers/metabolism , Creatine Kinase/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetic Cardiomyopathies/complications , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/prevention & control , Inflammation/pathology , Lactate Dehydrogenases/metabolism , Lipids , Lycium/chemistry , Oxidative Stress , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Rats , Triglycerides , Troponin T/metabolism
20.
J Biochem Mol Toxicol ; 36(9): e23143, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35815753

ABSTRACT

Bergapten (BeG) is explored for its anti-inflammatory and antioxidant properties. Myocardial infarction (MI) is reported to be one of the leading cardiovascular diseases characterized by mitochondrial dysfunction and apoptosis. The main purpose of this study is to assess the cardiopreventive effects of BeG (50 mg/kg) in isoproterenol (ISO)-induced MI in Wistar rats. The increased infarct size after ISO induction was reduced simultaneously on treatment with BeG. Similarly, augmented levels of cardiac biomarkers, namely cardiac troponin T, creatine kinase (CK), cardiac troponin I, and CK-MB were also suppressed by BeG. The increased rate of lipid hydroperoxides and thiobarbituric acid reactive substances owing to the oxidative stress caused by free radical generation in ISO-induced rats were also inhibited by BeG. Antioxidants reduce oxidative stress by scavenging free radicals. ISO induction reduces these antioxidant enzymes glutathione peroxidase, catalase, superoxide dismutase, and glutathione, and levels causing oxidative cardiac damage to the heart tissue. BeG supplementation improved these enzymes synthesis preventing potential damage to the myocardium. Inflammation caused by ISO pretreatment increased the secretion of proinflammatory cytokines in ISO-induced rats. Pretreatment with BeG suppressed these inflammatory cytokines to a normal level in ISO + BeG-treated rats. The histopathological examination of the morphological characteristics showed that the intensity of cardiac damage caused by ISO induction was less in BeG pretreated rats with less inflammatory cells and no necrosis. BeG also showed promising results in the molecular alteration of AMP-activated protein kinase/endothelial nitric oxide synthase/protein kinase B signaling molecules. These observations emphasize the cardioprotective effects of BeG and its potential use as a drug in the near future.


Subject(s)
AMP-Activated Protein Kinases , Myocardial Infarction , 5-Methoxypsoralen/adverse effects , AMP-Activated Protein Kinases/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Apoptosis , Biomarkers/metabolism , Catalase/metabolism , Creatine Kinase, MB Form , Cytokines/metabolism , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Isoproterenol/toxicity , Lipid Peroxides/metabolism , Myocardial Infarction/chemically induced , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Myocardium/metabolism , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Signal Transduction , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Troponin I/adverse effects , Troponin I/metabolism , Troponin T/metabolism , Troponin T/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...