157 results on '"Varga ZV"'
Search Results
2. Transcriptomic analysis and comparative characterization of rat H9C2, human AC16 and murine HL-1 cardiac cell lines
- Author
-
Gulyas-Onodi, Z, primary, Visnovitz, T, additional, Koncz, A, additional, Varadi, B, additional, Agg, B, additional, Kiss, B, additional, Makkos, A, additional, Nagy, NR, additional, Toth, VE, additional, Leszek, P, additional, Gorbe, A, additional, Giricz, Z, additional, Buzas, EI, additional, Ferdinandy, P, additional, and Varga, ZV, additional
- Published
- 2022
- Full Text
- View/download PDF
3. Rictor is a central target of the molecular network of cardiac ProtectomiRs
- Author
-
Makkos, A, primary, Agg, B, additional, Varga, ZV, additional, Giricz, Z, additional, Gyongyosi, M, additional, Lukovic, D, additional, Schulz, R, additional, Bartekova, M, additional, Gorbe, A, additional, and Ferdinandy, P, additional
- Published
- 2022
- Full Text
- View/download PDF
4. Immune checkpoint inhibition with PD-1 inhibitor induces cardiac dysfunction without overt myocarditis in C57BL/6J mice
- Author
-
Gergely, T, primary, Kucsera, D, additional, Toth, VE, additional, Petrovich, B, additional, Agg, B, additional, Onodi, ZS, additional, Ruppert, M, additional, Radovits, T, additional, Merkely, B, additional, Ferdinandy, P, additional, and Varga, ZV, additional
- Published
- 2022
- Full Text
- View/download PDF
5. Investigation of cardiotoxicity by dipeptidyl-peptidase-4 inhibitors in a human cardiomyocyte cell line as well as in samples from chronic heart failure patients
- Author
-
Voros, I, primary, Onodi, ZS, additional, Toth, VE, additional, Gergely, T, additional, Saghy, E, additional, Gorbe, A, additional, Kemeny, A, additional, Leszek, P, additional, Helyes, ZS, additional, Ferdinandy, P, additional, and Varga, ZV, additional
- Published
- 2022
- Full Text
- View/download PDF
6. New targets and indications for probenecid
- Author
-
Onódi Z, Koch S, Rubinstein J, Ferdinandy P, Varga ZV.
- Published
- 2022
- Full Text
- View/download PDF
7. Molecular characterization of immune checkpoint inhibitor-induced cardiotoxicity reveals IL-17A as a driver of cardiac dysfunction after anti-PD-1 treatment
- Author
-
Gergely TG, Kucsera D, Tóth VE, Kovács T, Sayour NV, Drobni ZD, Ruppert M, Petrovich B, Ágg B, Onódi Z, Fekete N, Pállinger É, Buzás EI, Yousif LI, Meijers WC, Radovits T, Merkely B, Ferdinandy P, Varga ZV.
- Published
- 2022
- Full Text
- View/download PDF
8. P425Cytoprotection by the NO-donor SNAP and BNP against ischemia/reperfusion injury in rat engineered heart tissue
- Author
-
Gorbe, A, Eder, A, Varga, ZV, Paloczi, J, Hansen, A, Ferdinandy, P, and Eschenhagen, T
- Published
- 2014
- Full Text
- View/download PDF
9. P435Extracellular vesicles mediate cardioprotection exerted by remote ischemic preconditioning in rats
- Author
-
Baranyai, T, Giricz, Z, Varga, ZV, Sipos, P, Paloczi, K, Kittel, A, Buzas, E, and Ferdinandy, P
- Published
- 2014
- Full Text
- View/download PDF
10. P366The time-course of cardiac and pulmonary matrix metalloproteinase-2 and -9 activities after chronic cigarette smoke exposure in mice
- Author
-
Kiss, K, Bencsik, P, Varga, ZV, Szitter, I, Ferdinandy, P, and Helyes, ZS
- Published
- 2014
- Full Text
- View/download PDF
11. P352High cholesterol diet deteriorates cardiac autophagy
- Author
-
Giricz, Z, Varga, ZV, Csonka, C, Szucs, G, Adameova, A, Gottlieb, RA, Mentzer, RM, and Ferdinandy, P
- Published
- 2014
- Full Text
- View/download PDF
12. P153MicroRNAs involved in cardioprotection by ischemic pre- and postconditioning: ProtectomiRs
- Author
-
Varga, ZV, Zvara, A, Farago, N, Pipicz, M, Gaspar, R, Gorbe, A, Puskas, GL, Csont, T, Thum, T, and Ferdinandy, P
- Published
- 2014
- Full Text
- View/download PDF
13. P148Cardiac myocytes are protected by small leucine rich proteoglycans against simulated ischemia/reperfusion injury
- Author
-
Gaspar, R, Gorbe, A, Paloczi, J, Varga, ZV, Gomori, K, Ferdinandy, P, and Csont, T
- Published
- 2014
- Full Text
- View/download PDF
14. Cardiovascular Glycobiology11Acute hyperglycemia abolishes cardioprotection by remote ischemic perconditioning12Deregulation of thioredoxin system contributes to monocyte dysfunction in diabetes mellitus: Implications for impaired arteriogenesis in type2 diabetic patients13High glucose increases gamma-glutamyltransferase-induced tissue factor expression in human peripheral blood mononuclear cells
- Author
-
Baranyai, T, primary, Godfrey, R, primary, Scalise, V, primary, Nagy, CT, additional, Koncsos, G, additional, Onodi, Z, additional, Karolyi-Szabo, M, additional, Makkos, A, additional, Varga, ZV, additional, Ferdinandy, P, additional, Giricz, Z, additional, Schulten, HM, additional, Shanmuganathan, SK, additional, Loeffler, I, additional, Mueller, N, additional, Wolf, G, additional, Mueller, UA, additional, Boehmer, FD, additional, Waltenberger, J, additional, Balia, C, additional, Cianchetti, S, additional, Carnicelli, V, additional, Faita, F, additional, Neri, T, additional, Zucchi, R, additional, Corti, A, additional, Celi, A, additional, and Pedrinelli, R, additional
- Published
- 2016
- Full Text
- View/download PDF
15. Searching for cardioprotective microRNA families by bioinformatics analysis of cross-species transcriptomic datasets.
- Author
-
Agg, B, Benczik, B, Kemendi, BV, Makkos, A, Petervari, M, Varga, ZV, and Ferdinandy, P
- Subjects
MICRORNA ,TRANSCRIPTOMES ,GENE ontology ,DEVELOPMENTAL biology ,MOLECULAR biology ,ISCHEMIC preconditioning - Abstract
Funding Acknowledgements Type of funding sources: Public grant(s) – National budget only. Main funding source(s): This study was supported by the National Research, Development and Innovation Office of Hungary (NKFIA; NVKP-16-1-2016-0017 National Heart Program) and the Thematic Excellence Programme (2020-4.1.1.-TKP2020) of the Ministry for Innovation and Technology in Hungary, within the framework of the Therapeutic Development and Molecular Biology thematic programmes of the Semmelweis University. Introduction There is a growing body of evidence that changes in cardiac microRNA expression pattern plays an important role in cardioprotective cellular signaling. Purpose Our aim was to identify evolutionarily conserved cardioprotective microRNA (protectomiR) families with a high probability of translation into human clinical practice. Methods Global cardiac microRNA expression profiles were measured in both a rat and a porcine model of myocardial infarction with or without ischemic preconditioning and postconditioning by microarray and high-throughput polymerase chain reaction, respectively. A software developed by our team was used to identify cross-species microRNA families (i.e. microRNAs with identic seed sequence) with cardioprotective expression patterns. By selecting experimentally validated human target genes from the miRTarBase database we built a microRNA family-target interaction network by the miRNAtarget.com software. Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed on the selected target genes. Results Due to potential patenting, microRNAs and microRNA families were anonymized. We identified 7 microRNA families, showing cardioprotective expression pattern. Based on the analysis of the microRNA family-target network, expression change of the target hubs were predicted to be consistent with previously described changes in pathways with high relevance in cardioprotection (e.g. positive regulation of TGF-beta, HIF-1, PI3K-Akt signaling pathways). Conclusion By an unbiased transcriptomics and bioinformatics based approach we successfully identified microRNA families with robust cross-species protectomiR expression pattern and targets involved in well-established cardioprotective pathways. These cardioprotective microRNA families are of high translational value for human clinical application. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
16. P352 High cholesterol diet deteriorates cardiac autophagy.
- Author
-
Giricz, Z, Varga, ZV, Csonka, C, Szucs, G, Adameova, A, Gottlieb, RA, Mentzer, RM, and Ferdinandy, P
- Subjects
- *
HIGH cholesterol diet , *AUTOPHAGY , *ISCHEMIA , *HYPERCHOLESTEREMIA , *CELLULAR signal transduction , *APOPTOSIS - Abstract
Introduction: We have previously shown that autophagy is necessary for the ischemic preconditioning (IPC) of the heart and that IPC is compromised in the setting of hypercholesterolemia (HC). The mTOR pathway, an upstream modulator of autophagy, has been shown to be activated in HC. The purpose of this study was to test the hypothesis that HC results in down regulation of autophagy due in part to activation of mTOR signalling and to investigate whether HC modulates programmed cell death mechanism such as apoptosis and necroptosis in the heart.Methods: Male Wistar rats were fed either normal chow or with 2% cholesterol and 1% cholic acid-enriched diet for 10 weeks. HC rats exhibited a 41% increase in plasma total cholesterol level over that of rats fed standard chow (2.89mmol/L vs. 4.09mmol/L) at the end of diet period. Animals were sacrificed, hearts were excised and briefly washed out, then left ventricles were snap-frozen for determination of autophagy, mTOR pathway, apoptosis, and necroptosis-related proteins by Western blot.Results: HC was associated with a significant reduction in LC3-II and in Beclin-1 expression compared to control values. Consistent with mTOR activation, S6 phosphorylation, an mTOR target, was increased in HC animals (p=0.021). Bax/Bcl2 ratio and cleaved caspase-3 signal increases in HC animlas, while no difference in the expression of RIP3 or MLKL was detected between treatments.Conclusion: These data indicate that hypercholesterolemia suppresses basal cardiac autophagy and that the decrease in autophagy may be a result of an upregulated mTOR pathway; however the exact signaling mechanism is yet to be elucidated. Reduced autophagy was accompanied by signs of increased apoptosis, while cardiac necroptosis was not modulated by HC. Decreased basal autophagy and elevated apoptosis may be responsible for the loss of cardioprotection reported in hypercholesterolemic states. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
17. P153 MicroRNAs involved in cardioprotection by ischemic pre- and postconditioning: ProtectomiRs.
- Author
-
Varga, ZV, Zvara, A, Farago, N, Pipicz, M, Gaspar, R, Gorbe, A, Puskas, GL, Csont, T, Thum, T, and Ferdinandy, P
- Subjects
- *
MICRORNA , *CARDIOTONIC agents , *ISCHEMIA , *GENE expression , *REPERFUSION injury - Abstract
Purpose: We aimed to characterize early changes in microRNA expression in acute cardioprotection by ischemic pre- and postconditioning in rat hearts.Methods: Hearts isolated from male Wistar rats were subjected to i) time-matched non-ischemic perfusion, ii) ischemia/reperfusion (30 min coronary occlusion and 120 min reperfusion), iii) preconditioning (3x5 min coronary occlusion) followed by ischemia/reperfusion, or iv) ischemia/reperfusion with postconditioning (6x10s global ischemia/reperfusion at the onset of reperfusion, respectively. Infarct size was significantly reduced by both interventions.Results: Out of 350 different microRNAs assessed by microarray analysis, 147-160 showed detectable expression levels. As compared to microRNA alterations induced by ischemia/reperfusion vs. time-matched non-ischemic controls, 5 microRNAs were significantly affected by both pre- and postconditioning (microRNA-125b*, 139-3p, 320, 532-3p, 188), 4 microRNAs by preconditioning (microRNA-487b, 139-5p, 192, 212), and 9 by postconditioning (microRNA-1, let-7i, let-7e, let7b, 181a, 208, 328, 335, 503), respectively. Expression of randomly selected microRNAs was validated by QRT-PCR. By a systematic comparison of the direction of microRNA expression changes in all groups, we identified microRNAs, specific mimics or antagomiRs of which may have pre- and postconditioning-like cardioprotective effect (protectomiRs). Transfection of selected protectomiRs (mimics of microRNA-139-5p, -125b*, and let-7b) into cardiac myocytes subjected to simulated ischemia/reperfusion showed significant cytoprotective effect.Conclusions: This is the first demonstration that ischemia/reperfusion-induced microRNA expression profile is significantly influenced by both pre- and postconditioning, which shows the involvement of microRNAs in cardioprotective signaling. Moreover, by analysis of microRNA expression patterns in cardioprotection by pre- and postconditioning, specific protectomiRs can be revealed as potential therapeutic tools treating ischemia/reperfusion injury. [ABSTRACT FROM PUBLISHER]
- Published
- 2014
- Full Text
- View/download PDF
18. P435 Extracellular vesicles mediate cardioprotection exerted by remote ischemic preconditioning in rats.
- Author
-
Baranyai, T, Giricz, Z, Varga, ZV, Sipos, P, Paloczi, K, Kittel, A, Buzas, E, and Ferdinandy, P
- Subjects
VESICLES (Cytology) ,REMOTE sensing ,ISCHEMIA ,LABORATORY rats ,HEART physiology ,EXOSOMES - Abstract
Background: Remote ischemic preconditioning (RIPC) of the heart (i.e. brief ischemia/reperfusion cycles, subjected to a remote organ or a remote area, reduce the consecutive damage of a sustained cardiac ischemia. Although, its mechanism has been investigated thoroughly, exact interorgan transfer mechanisms of cardioprotection by RIPC are required to be further investigated. Exosomes and microvesicles/microparticles are vesicles of 30-100 nm and 100-1000 nm in diameter, respectively (collectively termed extracellular vesicles [EVs]). As EVs are able to shuttle proteins, mRNAs and microRNAs, they are ideal candidates for inter-organ communication. However, whether EVs are involved in RIPC, is unknown.Purpose: Here we investigated whether (1) IPC induces release of EVs from the heart, and (2) EVs are necessary for cardioprotection by RIPC.Methods: Hearts of male Wistar rats were isolated and perfused as described by Langendorff. Hearts were exposed to 3 × 5 min global ischemia/reperfusion cycles (IPC) or 30 min aerobic perfusion, while coronary perfusates were collected. Coronary perfusates of these donor hearts were given to another set of recipient isolated hearts either unharmed or depleted of EVs by differential ultracentrifugation. Infarct size was determined by triphenyl tetrazolium chloride at the end of 30 min global ischemia and 120 min reperfusion period. The presence or absence of EVs in perfusates was confirmed by dynamic light scattering, the EV marker hsp60 Western blot, and electron microscopy.Results: IPC markedly increased EV release from the heart as assessed by HSP60 immunoblot. Administration of coronary perfusate from IPC donor hearts attenuated infarct size in non-preconditioned recipient hearts (12.9 ± 1.6% vs. 25.0 ± 2.7%, respectively), similarly to cardioprotection afforded by IPC (7.3 ± 2.7% vs. 22.1 ± 2.9%, respectively) on the donor hearts. Furthermore, perfusates of IPC hearts depleted of EVs failed to exert cardioprotection in recipient hearts (22.0 ± 2.3%).Conclusions: This is the first demonstration that EVs released from the heart after IPC are necessary for cardioprotection by RIPC, evidencing the importance of vesicular transfer mechanisms in remote cardioprotection. [ABSTRACT FROM PUBLISHER]
- Published
- 2014
- Full Text
- View/download PDF
19. P148 Cardiac myocytes are protected by small leucine rich proteoglycans against simulated ischemia/reperfusion injury.
- Author
-
Gaspar, R, Gorbe, A, Paloczi, J, Varga, ZV, Gomori, K, Ferdinandy, P, and Csont, T
- Subjects
MUSCLE cells ,HEART cells ,ISCHEMIA ,CELL death ,CARDIOTONIC agents ,REPERFUSION injury ,CHONDROITIN - Abstract
It was previously shown by our group that the overexpression of biglycan in a transgenic mouse model enhances the induction of cardioprotective genes. Ischemia induced cell death was decreased by bigylcan treatment in neonatal cardiomyocytes. In this study we aimed to test whether the biglycan core protein or the glycosaminoglycan is responsible for the cardioprotective effect of biglycan. We also investigated whether decorin, (which has a similar structure to biglycan), has any cardio-protective effects on cardiomyocytes during simulated ischemia/reperfusion (SI/R) injury.Primary cardiomyocyte cultures were isolated from neonatal rat hearts. The 2 day-old cultures were pretreated with 0, 1, 3, 10, 30, and 100 nM decorin, biglycan, biglycan core protein or chondroitin sulfate A for 20 hours followed by an SI/R protocol. The treatment was maintained throughout SI , 240 min, hypoxic solution, hypoxic chamber gassed with 95% N2 ès 5% CO2) and reperfusion (120 min, normoxic conditions, medium). At the end of the protocol, a viability assay was performed using calcein staining.Compared to the normoxic untreated control, approximately 40% of the cardiomyocytes died during the simulated ischemia/reperfusion injury. Doses of 10 and 30 nM biglycan increased cell viability significantly, compared to SI/R control (29±4 and 18±5%). The biglycan core protein showed similar cytoprotective effects at 3 and 10 nM doses (21±4 and 24±5%), however, the chondroitin sulfate had no influence on the viability of cardiomyocytes. In the case of decorin, the 3 and 10 nM doses significantly increased the survival of the cells (28±3 and 20±4%).Based on our experiments, we concluded that both decorin and biglycan exert a cyto-protective effect on cardiomyocytes against SI/R injury and this protective effect of biglycan is likely due to the biglycan core protein.Grants: NKTM-OTKA-PD-106001, South-East European Foundation Lykeon Scholarship, GA holds Bolyai Janos Fellowship [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
20. P366 The time-course of cardiac and pulmonary matrix metalloproteinase-2 and -9 activities after chronic cigarette smoke exposure in mice.
- Author
-
Kiss, K, Bencsik, P, Varga, ZV, Szitter, I, Ferdinandy, P, and Helyes, ZS
- Subjects
CIGARETTE smoke ,LABORATORY mice ,MATRIX metalloproteinases ,GELATINASES ,INFLAMMATION ,CARDIOVASCULAR diseases - Abstract
Background: Gelatinase-type matrix metalloproteinases (MMP-2 and -9) have crucial role in inflammatory diseases, e.g. chronic obstructive pulmonary disease as well as in cardiovascular pathology. It is known that chronic smoke exposure increases MMP-2 and -9 activities in the lung. However, the smoke-induced alterations in MMP-2 and -9 activities in the heart and the time-course of their pulmonary and myocardial activity during chronic smoke exposure have not been examined.Purpose: Therefore, the aim of this study was to characterize the time-course of cardiac and pulmonary MMP-2 and -9 activities during chronic tobacco smoke exposure.Methods: Eight week-old male C57Bl/6 wild type mice (25-30 g; n=5-6/group) were exposed to whole body tobacco smoke with a manual system twice a day for 30 min throughout periods of 1, 2 and 3 months. Age-matched mice without smoke administration were used as controls. Then hearts and lungs were isolated at the end of each month, rinsed in PBS and snap-frozen in liquid nitrogen. MMPs activities were determined by gelatin zymography.Results: The myocardial activity of 72- and 75 kDa-MMP-2 showed an increasing tendency after 3-month smoke exposure compared to the time-matched control group, however, the difference did not reach statistical significance. Although MMP-9 yielded weak signals from heart samples, MMP-9 activity decreased significantly at the end of the second month compared to the corresponding controls (7,140±3,809 and 2,266±750 AU, p=0.008), and showed a mild increase after 3 months. In the lung, we have found a non-significant decrease in the activity of both MMP-2 isoforms in the 3-month smoking group when compared to the time-matched controls. MMP-9 activity was significantly lower after 3-month smoke exposure compared to the control mice (35,476±6,628 and 28,424±2,578 AU, p=0.03 respectively). Tobacco smoke administration for 1 or 2 months did not influence MMP-2 or -9 activities in either the heart or the lung compared to the respective controls.Conclusion: MMP-9 activity decreased after 3-month tobacco smoke exposure in the lung, while MMP-2 activity showed only a tendency of decrease. Meanwhile, in the heart, a moderate increase was observed in the activity of both MMPs. Further experiments are needed with longer smoke exposure to evaluate the time-dependent alterations and possible roles of MMP-2 and -9 in tobacco smoke-induced cardiac and pulmonary changes. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
21. P425 Cytoprotection by the NO-donor SNAP and BNP against ischemia/reperfusion injury in rat engineered heart tissue.
- Author
-
Gorbe, A, Eder, A, Varga, ZV, Paloczi, J, Hansen, A, Ferdinandy, P, and Eschenhagen, T
- Subjects
CYTOPROTECTION ,NITROGEN oxides ,TISSUE engineering ,REPERFUSION injury ,CARDIOTONIC agents ,LABORATORY rats - Abstract
Purpose: In vitro drug screening and disease modeling is a rapidly growing research field and may help to replace animal experiments. The aim of this study was to test the response of engineered heart tissue (EHT) to ischemia/reperfusion (I/R) and the effect of known cardioprotective molecules.Method: Fibrin-based mini-EHT were generated in a 24-well format from neonatal rat heart cells and cultured at 37°C, 7% CO2 and 40% O2 for 15-22 days (DMEM, 10% horse serum). 24 h after the last medium change, EHTs were subjected to 180 min ischemia (93% N2 and 7 % CO2 gas flow) followed by 120 min reperfusion (40% O2) or remained at 40% O2 as time-matched controls. The following treatments were applied during I/R (n=6 each): fresh EHT medium (positive control), the NO-donor S-Nitroso-N-acetyl-D,L-penicillamine (SNAP, 10-7, 10-6, 10-5 M) or the particulate guanylate cyclase activator brain type natriuretic peptide (BNP, 10-9, 10-8, 10-7 M). Beating rate and force of contraction were monitored during the entire experiment; pH, Troponin I and LDH release and glucose consumption were measured in EHT medium after I/R.Result: During the 180 min ischemia, EHTs stopped to beat or beat at significantly lower rate. Most EHTs started to beat during reoxygenation and survived until the end of a 2-day follow up period. The cardioprotective interventions did not significantly affect contractile activity during the ischemic phase. In contrast, fresh medium as well as SNAP and BNP increased rate and/or rate x force product during reoxygenation. Troponin I (ranged between 0,2-0,8 ng/ml) and LDH release (0,07-0,08 U/mL) as well as glucose consumption (3-3,5 mmol/L; fresh medium 4,8 mmol/L) and medium pH (7,5) was not significantly affected by SNAP or BNP.Conclusion: The 24-well EHT test system is applicable to test cardioprotective compounds in vitro and monitor several functional and biochemical endpoints, which otherwise could be only measured in vivo or ex vivo heart preparations.Grants: German Research Foundation (DFG Es/88-12), the European Commission (FP7 Angioscaff, FP7 Biodesign), NKFP 07 1-ES2HEART-HU (OM-00202/2007), National Development Agency - New Hungary Development Plan (TÁMOP-4.2.2-08/1/2008-0013, TÁMOP-4.2.1/B-09/1); OTKA PD 106001. [ABSTRACT FROM PUBLISHER]
- Published
- 2014
- Full Text
- View/download PDF
22. Pharmacokinetics and cardioprotective efficacy of intravenous miR-125b* microRNA mimic in a mouse model of acute myocardial infarction.
- Author
-
Szabados T, Makkos A, Ágg B, Benczik B, Brenner GG, Szabó M, Váradi B, Vörös I, Gömöri K, Varga ZV, Görbe A, Bencsik P, and Ferdinandy P
- Abstract
Background and Purpose: MicroRNA (miRNA) therapy is a promising approach to induce cardioprotection. We have previously identified cardiac microRNA-125b* (microRNA-125b-2-3p; miR-125b*) as a potential cardioprotective miRNA, termed ProtectomiR. We aimed to characterize the pharmacokinetics and pharmacodynamics, and the effect of miR-125b* mimic on infarct size using an in vivo mouse model., Experimental Approach: To characterize the pharmacokinetics properties of miR-125b* mimic, a single injection of 10-μg miR-125b* mimic or its scramble miRNA control, or vehicle i.v. was given to C57BL/6 mice. MiR-125b* expression was measured from plasma, heart, kidney and liver samples. Effect of miR-125b* on area at risk and infarct size was assessed after 45-min coronary occlusion, followed by 24-h reperfusion; 10-μg miR-125b* mimic or 10-μg non-targeting miRNA mimic control or vehicle were administered via the right jugular vein at 10th mins of coronary occlusion. To assess molecular mechanism involved in cardioprotection, expression of mRNA targets of miR-125b* were measured from ventricular myocardium at 1, 2, 4, 8 or 24 h post-treatment using quantitative real time polymerase chain reaction., Key Results: MiR-125b* expression was markedly increased in plasma and myocardium 1 h, and in the liver 2h after treatment. Infarct size was significantly reduced after miR-125b* mimic treatment when compared to the vehicle. The expression of Ccna2, Eef2k and Cacnb2 target mRNAs was significantly reduced 8 h after injection of miR-125b* mimic., Conclusion and Implications: This is the first demonstration of pharmacokinetic and molecular pharmacodynamic properties as well as the cardioprotective effect of miR-125b* mimic in vivo., (© 2024 The Author(s). British Journal of Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.)
- Published
- 2024
- Full Text
- View/download PDF
23. Reduced circulating CD63 + extracellular vesicle levels associate with atherosclerosis in hypercholesterolaemic mice and humans.
- Author
-
Kestecher BM, Németh K, Ghosal S, Sayour NV, Gergely TG, Bodnár BR, Försönits AI, Sódar BW, Oesterreicher J, Holnthoner W, Varga ZV, Giricz Z, Ferdinandy P, Buzás EI, and Osteikoetxea X
- Subjects
- Aged, Animals, Female, Humans, Male, Middle Aged, Aortic Diseases pathology, Aortic Diseases metabolism, Aortic Diseases blood, Aortic Diseases etiology, Aortic Diseases physiopathology, Case-Control Studies, Cholesterol blood, Mice, Inbred C57BL, Mice, Knockout, Plaque, Atherosclerotic, Atherosclerosis pathology, Atherosclerosis metabolism, Atherosclerosis blood, Atherosclerosis etiology, Atherosclerosis physiopathology, Biomarkers blood, Diet, High-Fat, Disease Models, Animal, Extracellular Vesicles metabolism, Hypercholesterolemia blood, Hypercholesterolemia complications, Proprotein Convertase 9 metabolism, Proprotein Convertase 9 genetics, Proprotein Convertase 9 blood, Receptors, LDL deficiency, Receptors, LDL genetics, Tetraspanin 30 metabolism
- Abstract
Aims: The association and co-isolation of low-density lipoproteins (LDL) and extracellular vesicles (EVs) have been shown in blood plasma. Here we explore this relationship to better understand the role of EVs in atherogenesis., Methods and Results: Wild type (WT), PCSK9
-/- , and LDLR-/- C57BL/6 mice were used in this study. Eleven week-old male mice were fed high-fat diet (HFD) for 12 weeks or kept on normal diet until old age (22-months). Cardiac function was assessed by ultrasound, cholesterol was quantified with a colorimetric kit and circulating EVs were measured using flow cytometry. Plaques were analysed post-mortem using Oil-Red-O staining of the aortic arch. EVs were measured from platelet free blood plasma samples of normal and hypercholesterolaemic clinical patients. Based on annexin V and CD63 staining, we found a significant increase in EV levels in LDLR-/- and PCSK9-/- mice after HFD, but CD81 showed no significant change in either group. There was no significant change in plaque formation after HFD. PCSK9-/- mice show a favourable cardiac function after HFD. Blood cholesterol levels progressively increased during HFD, with LDLR-/- mice showing high levels while PCSK9-/- were significantly lowered compared to WT animals. In mice at old age, similar cholesterol levels were observed as in young mice. In old age, LDLR-/- mice showed significantly increased plaques. At old age, ejection fraction was decreased in all groups of mice, as were CD63+ EVs. Similarly to mice, in patients with hypercholesterolaemia, CD63+ EVs were significantly depleted., Conclusions: This research demonstrates an inverse relationship between circulating EVs and cholesterol, making EVs a potential marker for cardiovascular disease (CVD). HFD causes reduced cardiac function, but atherosclerotic development is slowly progressing in hypercholesterolaemic models and only observed with old animals. These results also bring further evidence for the benefit of using of PCSK9 inhibitors as therapeutic agents in CVD., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
24. NASH triggers cardiometabolic HFpEF in aging mice.
- Author
-
Kucsera D, Ruppert M, Sayour NV, Tóth VE, Kovács T, Hegedűs ZI, Onódi Z, Fábián A, Kovács A, Radovits T, Merkely B, Pacher P, Ferdinandy P, and Varga ZV
- Subjects
- Animals, Male, Mice, Mice, Inbred C57BL, Aging physiology, Non-alcoholic Fatty Liver Disease physiopathology, Non-alcoholic Fatty Liver Disease metabolism, Non-alcoholic Fatty Liver Disease pathology, Heart Failure physiopathology, Heart Failure etiology, Disease Models, Animal, Echocardiography, Stroke Volume physiology
- Abstract
Both heart failure with preserved ejection fraction (HFpEF) and non-alcoholic fatty liver disease (NAFLD) develop due to metabolic dysregulation, has similar risk factors (e.g., insulin resistance, systemic inflammation) and are unresolved clinical challenges. Therefore, the potential link between the two disease is important to study. We aimed to evaluate whether NASH is an independent factor of cardiac dysfunction and to investigate the age dependent effects of NASH on cardiac function. C57Bl/6 J middle aged (10 months old) and aged mice (24 months old) were fed either control or choline deficient (CDAA) diet for 8 weeks. Before termination, echocardiography was performed. Upon termination, organ samples were isolated for histological and molecular analysis. CDAA diet led to the development of NASH in both age groups, without inducing weight gain, allowing to study the direct effect of NASH on cardiac function. Mice with NASH developed hepatomegaly, fibrosis, and inflammation. Aged animals had increased heart weight. Conventional echocardiography revealed normal systolic function in all cohorts, while increased left ventricular volumes in aged mice. Two-dimensional speckle tracking echocardiography showed subtle systolic and diastolic deterioration in aged mice with NASH. Histologic analyses of cardiac samples showed increased cross-sectional area, pronounced fibrosis and Col1a1 gene expression, and elevated intracardiac CD68
+ macrophage count with increased Il1b expression. Conventional echocardiography failed to reveal subtle change in myocardial function; however, 2D speckle tracking echocardiography was able to identify diastolic deterioration. NASH had greater impact on aged animals resulting in cardiac hypertrophy, fibrosis, and inflammation., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
25. Cardioprotective microRNAs (protectomiRs) in a pig model of acute myocardial infarction and cardioprotection by ischaemic conditioning: MiR-450a.
- Author
-
Nagy RN, Makkos A, Baranyai T, Giricz Z, Szabó M, Kravcsenko-Kiss B, Bereczki Z, Ágg B, Puskás LG, Faragó N, Schulz R, Gyöngyösi M, Lukovic D, Varga ZV, Görbe A, and Ferdinandy P
- Abstract
Background and Purpose: Cardioprotective miRNAs (protectomiRs) are promising therapeutic tools. Here, we aimed to identify protectomiRs in a translational porcine model of acute myocardial infarction (AMI) and to validate their cardiocytoprotective effect., Experimental Approach: ProtectomiR candidates were selected after systematic analysis of miRNA expression changes in cardiac tissue samples from a closed-chest AMI model in pigs subjected to sham operation, AMI and ischaemic preconditioning, postconditioning or remote preconditioning, respectively. Cross-species orthologue protectomiR candidates were validated in simulated ischaemia-reperfusion injury (sI/R) model of isolated rat ocardiomyocytes and in human AC16 cells as well. For miR-450a, we performed target prediction and analysed the potential mechanisms of action by GO enrichment and KEGG pathway analysis., Key Results: Out of the 220 detected miRNAs, four were up-regulated and 10 were down-regulated due to all three conditionings versus AMI. MiR-450a and miR-451 mimics at 25 nM were protective in rat cardiomyocytes, and miR-450a showed protection in human cardiomyocytes as well. MiR-450a has 3987 predicted mRNA targets in pigs, 4279 in rats and 8328 in humans. Of these, 607 genes are expressed in all three species. A total of 421 common enriched GO terms were identified in all three species, whereas KEGG pathway analysis revealed 13 common pathways., Conclusion and Implications: This is the first demonstration that miR-450a is associated with cardioprotection by ischaemic conditioning in a clinically relevant porcine model and shows cardiocytoprotective effect in human cardiomyocytes, making it a promising drug candidate. The mechanism of action of miR-450a involves multiple cardioprotective pathways., (© 2024 The Author(s). British Journal of Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.)
- Published
- 2024
- Full Text
- View/download PDF
26. Comparison of mouse models of heart failure with reduced ejection fraction.
- Author
-
Sayour NV, Gergely TG, Váradi B, Tóth VÉ, Ágg B, Kovács T, Kucsera D, Kovácsházi C, Brenner GB, Giricz Z, Ferdinandy P, and Varga ZV
- Abstract
Aims: Heart failure with reduced ejection fraction (HFrEF) is a leading cause of death worldwide; thus, therapeutic improvements are needed. In vivo preclinical models are essential to identify molecular drug targets for future therapies. Transverse aortic constriction (TAC) is a well-established model of HFrEF; however, highly experienced personnel are needed for the surgery, and several weeks of follow-up are necessary to develop HFrEF. To this end, we aimed (i) to develop an easy-to-perform mouse model of HFrEF by treating Balb/c mice with angiotensin-II (Ang-II) for 2 weeks by minipump and (ii) to compare its cardiac phenotype and transcriptome to the well-established TAC model of HFrEF in C57BL/6J mice., Methods: Mortality and gross pathological data, cardiac structural and functional characteristics assessed by echocardiography and immunohistochemistry and differential gene expression obtained by RNA-sequencing and gene-ontology analyses were used to characterize and compare the two models. To achieve statistical comparability between the two models, changes in treatment groups related to the corresponding control were compared (ΔTAC vs. ΔAng-II)., Results: Compared with the well-established TAC model, chronic Ang-II treatment of Balb/c mice shares similarities in cardiac systolic functional decline (left ventricular ejection fraction: -57.25 ± 7.17% vs. -43.68 ± 5.31% in ΔTAC vs. ΔAng-II; P = 0.1794) but shows a lesser degree of left ventricular dilation (left ventricular end-systolic volume: 190.81 ± 44.13 vs. 57.37 ± 10.18 mL in ΔTAC vs. ΔAng-II; P = 0.0252) and hypertrophy (cell surface area: 58.44 ± 6.1 vs. 10.24 ± 2.87 μm
2 in ΔTAC vs. ΔAng-II; P < 0.001); nevertheless, transcriptomic changes in the two HFrEF models show strong correlation (Spearman's r = 0.727; P < 0.001). In return, Ang-II treatment in Balb/c mice needs significantly less procedural time [38 min, interquartile range (IQR): 31-46 min in TAC vs. 6 min, IQR: 6-7 min in Ang-II; P < 0.001] and surgical expertise, is less of an object for peri-procedural mortality (15.8% in TAC vs. 0% in Ang-II; P = 0.105) and needs significantly shorter follow-up for developing HFrEF., Conclusions: Here, we demonstrate for the first time that chronic Ang-II treatment of Balb/c mice is also a relevant, reliable but significantly easier-to-perform preclinical model to identify novel pathomechanisms and targets in future HFrEF research., (© 2024 The Author(s). ESC Heart Failure published by John Wiley & Sons Ltd on behalf of European Society of Cardiology.)- Published
- 2024
- Full Text
- View/download PDF
27. Molecular fingerprints of cardiovascular toxicities of immune checkpoint inhibitors.
- Author
-
Gergely TG, Drobni ZD, Sayour NV, Ferdinandy P, and Varga ZV
- Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy by unleashing the power of the immune system against malignant cells. However, their use is associated with a spectrum of adverse effects, including cardiovascular complications, which can pose significant clinical challenges. Several mechanisms contribute to cardiovascular toxicity associated with ICIs. First, the dysregulation of immune checkpoints, such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein-1 (PD-1) and its ligand (PD-L1), and molecular mimicry with cardiac autoantigens, leads to immune-related adverse events, including myocarditis and vasculitis. These events result from the aberrant activation of T cells against self-antigens within the myocardium or vascular endothelium. Second, the disruption of immune homeostasis by ICIs can lead to autoimmune-mediated inflammation of cardiac tissues, manifesting as cardiac dysfunction and heart failure, arrhythmias, or pericarditis. Furthermore, the upregulation of inflammatory cytokines, particularly tumor necrosis factor-alpha, interferon-γ, interleukin-1β, interleukin-6, and interleukin-17 contributes to cardiac and endothelial dysfunction, plaque destabilization, and thrombosis, exacerbating cardiovascular risk on the long term. Understanding the intricate mechanisms of cardiovascular side effects induced by ICIs is crucial for optimizing patient care and to ensure the safe and effective integration of immunotherapy into a broader range of cancer treatment protocols. The clinical implications of these mechanisms underscore the importance of vigilant monitoring and early detection of cardiovascular toxicity in patients receiving ICIs. Future use of these key pathological mediators as biomarkers may aid in prompt diagnosis of cardiotoxicity and will allow timely interventions., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
28. Immune checkpoints in cardiac physiology and pathology: therapeutic targets for heart failure.
- Author
-
Gergely TG, Drobni ZD, Kallikourdis M, Zhu H, Meijers WC, Neilan TG, Rassaf T, Ferdinandy P, and Varga ZV
- Subjects
- Humans, Animals, Signal Transduction, Cardiotoxicity, Heart Failure drug therapy, Heart Failure physiopathology, Heart Failure immunology, Immune Checkpoint Inhibitors adverse effects, Immune Checkpoint Inhibitors therapeutic use
- Abstract
Immune checkpoint molecules are physiological regulators of the adaptive immune response. Immune checkpoint inhibitors (ICIs), such as monoclonal antibodies targeting programmed cell death protein 1 or cytotoxic T lymphocyte-associated protein 4, have revolutionized cancer treatment and their clinical use is increasing. However, ICIs can cause various immune-related adverse events, including acute and chronic cardiotoxicity. Of these cardiovascular complications, ICI-induced acute fulminant myocarditis is the most studied, although emerging clinical and preclinical data are uncovering the importance of other ICI-related chronic cardiovascular complications, such as accelerated atherosclerosis and non-myocarditis-related heart failure. These complications could be more difficult to diagnose, given that they might only be present alongside other comorbidities. The occurrence of these complications suggests a potential role of immune checkpoint molecules in maintaining cardiovascular homeostasis, and disruption of physiological immune checkpoint signalling might thus lead to cardiac pathologies, including heart failure. Although inflammation is a long-known contributor to the development of heart failure, the therapeutic targeting of pro-inflammatory pathways has not been successful thus far. The increasingly recognized role of immune checkpoint molecules in the failing heart highlights their potential use as immunotherapeutic targets for heart failure. In this Review, we summarize the available data on ICI-induced cardiac dysfunction and heart failure, and discuss how immune checkpoint signalling is altered in the failing heart. Furthermore, we describe how pharmacological targeting of immune checkpoints could be used to treat heart failure., (© 2024. Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
29. Treatment options for immune-related adverse events associated with immune checkpoint inhibitors.
- Author
-
Chen YH, Kovács T, Ferdinandy P, and Varga ZV
- Abstract
The immunotherapy revolution with the use of immune checkpoint inhibitors (ICIs) started with the clinical use of the first ICI, ipilimumab, in 2011. Since then, the field of ICI therapy has rapidly expanded - with the FDA approval of 10 different ICI drugs so far and their incorporation into the therapeutic regimens of a range of malignancies. While ICIs have shown high anti-cancer efficacy, they also have characteristic side effects, termed immune-related adverse events (irAEs). These side effects hinder the therapeutic potential of ICIs and, therefore, finding ways to prevent and treat them is of paramount importance. The current protocols to manage irAEs follow an empirical route of steroid administration and, in more severe cases, ICI withdrawal. However, this approach is not optimal in many cases, as there are often steroid-refractory irAEs, and there is a potential for corticosteroid use to promote tumour progression. This review surveys the current alternative approaches to the treatments for irAEs, with the goal of summarizing and highlighting the best attempts to treat irAEs, without compromising anti-tumour immunity and allowing for rechallenge with ICIs after resolution of the irAEs., (© 2024 The Author(s). British Journal of Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.)
- Published
- 2024
- Full Text
- View/download PDF
30. Author Correction: Single-cell transcriptomics reveal extracellular vesicles secretion with a cardiomyocyte proteostasis signature during pathological remodeling.
- Author
-
Schoger E, Bleckwedel F, Germena G, Rocha C, Tucholla P, Sobitov I, Möbius W, Sitte M, Lenz C, Samak M, Hinkel R, Varga ZV, Giricz Z, Salinas G, Gross JC, and Zelarayán LC
- Published
- 2024
- Full Text
- View/download PDF
31. Heart failure pharmacotherapy and cancer: pathways and pre-clinical/clinical evidence.
- Author
-
Sayour NV, Paál ÁM, Ameri P, Meijers WC, Minotti G, Andreadou I, Lombardo A, Camilli M, Drexel H, Grove EL, Dan GA, Ivanescu A, Semb AG, Savarese G, Dobrev D, Crea F, Kaski JC, de Boer RA, Ferdinandy P, and Varga ZV
- Subjects
- Humans, Heart Failure drug therapy, Neoplasms epidemiology
- Abstract
Heart failure (HF) patients have a significantly higher risk of new-onset cancer and cancer-associated mortality, compared to subjects free of HF. While both the prevention and treatment of new-onset HF in patients with cancer have been investigated extensively, less is known about the prevention and treatment of new-onset cancer in patients with HF, and whether and how guideline-directed medical therapy (GDMT) for HF should be modified when cancer is diagnosed in HF patients. The purpose of this review is to elaborate and discuss the effects of pillar HF pharmacotherapies, as well as digoxin and diuretics on cancer, and to identify areas for further research and novel therapeutic strategies. To this end, in this review, (i) proposed effects and mechanisms of action of guideline-directed HF drugs on cancer derived from pre-clinical data will be described, (ii) the evidence from both observational studies and randomized controlled trials on the effects of guideline-directed medical therapy on cancer incidence and cancer-related outcomes, as synthetized by meta-analyses will be reviewed, and (iii) considerations for future pre-clinical and clinical investigations will be provided., (© The Author(s) 2024. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2024
- Full Text
- View/download PDF
32. A dual role of lysophosphatidic acid type 2 receptor (LPAR2) in nonsteroidal anti-inflammatory drug-induced mouse enteropathy.
- Author
-
Hutka B, Várallyay A, László SB, Tóth AS, Scheich B, Paku S, Vörös I, Pós Z, Varga ZV, Norman DD, Balogh A, Benyó Z, Tigyi G, Gyires K, and Zádori ZS
- Subjects
- Mice, Animals, Receptors, Lysophosphatidic Acid genetics, Receptors, Lysophosphatidic Acid metabolism, Mice, Inbred NOD, Mice, SCID, Anti-Inflammatory Agents, Non-Steroidal, Indomethacin adverse effects, Intestinal Diseases chemically induced, Diabetes Mellitus, Type 2, Lysophospholipids
- Abstract
Lysophosphatidic acid (LPA) is a bioactive phospholipid mediator that has been found to ameliorate nonsteroidal anti-inflammatory drug (NSAID)-induced gastric injury by acting on lysophosphatidic acid type 2 receptor (LPAR2). In this study, we investigated whether LPAR2 signaling was implicated in the development of NSAID-induced small intestinal injury (enteropathy), another major complication of NSAID use. Wild-type (WT) and Lpar2 deficient (Lpar2
-/- ) mice were treated with a single, large dose (20 or 30 mg/kg, i.g.) of indomethacin (IND). The mice were euthanized at 6 or 24 h after IND treatment. We showed that IND-induced mucosal enteropathy and neutrophil recruitment occurred much earlier (at 6 h after IND treatment) in Lpar2-/- mice compared to WT mice, but the tissue levels of inflammatory mediators (IL-1β, TNF-α, inducible COX-2, CAMP) remained at much lower levels. Administration of a selective LPAR2 agonist DBIBB (1, 10 mg/kg, i.g., twice at 24 h and 30 min before IND treatment) dose-dependently reduced mucosal injury and neutrophil activation in enteropathy, but it also enhanced IND-induced elevation of several proinflammatory chemokines and cytokines. By assessing caspase-3 activation, we found significantly increased intestinal apoptosis in IND-treated Lpar2-/- mice, but it was attenuated after DBIBB administration, especially in non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mice. Finally, we showed that IND treatment reduced the plasma activity and expression of autotaxin (ATX), the main LPA-producing enzyme, and also reduced the intestinal expression of Lpar2 mRNA, which preceded the development of mucosal damage. We conclude that LPAR2 has a dual role in NSAID enteropathy, as it contributes to the maintenance of mucosal integrity after NSAID exposure, but also orchestrates the inflammatory responses associated with ulceration. Our study suggests that IND-induced inhibition of the ATX-LPAR2 axis is an early event in the pathogenesis of enteropathy., (© 2023. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
33. YB-1 Is a Novel Target for the Inhibition of α-Adrenergic-Induced Hypertrophy.
- Author
-
Heger J, Partsch S, Harjung C, Varga ZV, Baranyai T, Weiß J, Kremer L, Locquet F, Leszek P, Ágg B, Benczik B, Ferdinandy P, Schulz R, and Euler G
- Subjects
- Adult, Humans, Animals, Rats, Phenylephrine, Myocytes, Cardiac, RNA, Small Interfering genetics, Adenoviridae, Cardiomegaly genetics, Transcription Factors, Adrenergic Agents, Heart Failure genetics
- Abstract
Cardiac hypertrophy resulting from sympathetic nervous system activation triggers the development of heart failure. The transcription factor Y-box binding protein 1 (YB-1) can interact with transcription factors involved in cardiac hypertrophy and may thereby interfere with the hypertrophy growth process. Therefore, the question arises as to whether YB-1 influences cardiomyocyte hypertrophy and might thereby influence the development of heart failure. YB-1 expression is downregulated in human heart biopsies of patients with ischemic cardiomyopathy ( n = 8), leading to heart failure. To study the impact of reduced YB-1 in cardiac cells, we performed small interfering RNA (siRNA) experiments in H9C2 cells as well as in adult cardiomyocytes (CMs) of rats. The specificity of YB-1 siRNA was analyzed by a miRNA-like off-target prediction assay identifying potential genes. Testing three high-scoring genes by transfecting cardiac cells with YB-1 siRNA did not result in downregulation of these genes in contrast to YB-1 , whose downregulation increased hypertrophic growth. Hypertrophic growth was mediated by PI3K under PE stimulation, as well by downregulation with YB-1 siRNA. On the other hand, overexpression of YB-1 in CMs, caused by infection with an adenovirus encoding YB-1 (AdYB-1), prevented hypertrophic growth under α-adrenergic stimulation with phenylephrine (PE), but not under stimulation with growth differentiation factor 15 (GDF15; n = 10-16). An adenovirus encoding the green fluorescent protein (AdGFP) served as the control. YB-1 overexpression enhanced the mRNA expression of the Gq inhibitor regulator of G-protein signaling 2 ( RGS2 ) under PE stimulation ( n = 6), potentially explaining its inhibitory effect on PE-induced hypertrophic growth. This study shows that YB-1 protects cardiomyocytes against PE-induced hypertrophic growth. Like in human end-stage heart failure, YB-1 downregulation may cause the heart to lose its protection against hypertrophic stimuli and progress to heart failure. Therefore, the transcription factor YB-1 is a pivotal signaling molecule, providing perspectives for therapeutic approaches.
- Published
- 2023
- Full Text
- View/download PDF
34. Depletion of muscularis macrophages ameliorates inflammation-driven dysmotility in murine colitis model.
- Author
-
Ferenczi S, Mogor F, Takacs P, Kovacs T, Toth VE, Varga ZV, Kovács K, Lohinai Z, Vass KC, Nagy N, and Dora D
- Subjects
- Mice, Animals, Inflammation, Macrophages, Colon, Dextran Sulfate toxicity, Mice, Inbred C57BL, Disease Models, Animal, Clodronic Acid pharmacology, Colitis chemically induced
- Abstract
Previously, the presence of a blood-myenteric plexus barrier and its disruption was reported in experimentally induced colitis via a macrophage-dependent process. The aim of this study is to reveal how myenteric barrier disruption and subsequent neuronal injury affects gut motility in vivo in a murine colitis model. We induced colitis with dextran sulfate sodium (DSS), with the co-administration of liposome-encapsulated clodronate (L-clodronate) to simultaneously deplete blood monocytes contributing to macrophage infiltration in the inflamed muscularis of experimental mice. DSS-treated animals receiving concurrent L-clodronate injection showed significantly decreased blood monocyte numbers and colon muscularis macrophage (MM) density compared to DSS-treated control (DSS-vehicle). DSS-clodronate-treated mice exhibited significantly slower whole gut transit time than DSS-vehicle-treated animals and comparable to that of controls. Experiments with oral gavage-fed Evans-blue dye showed similar whole gut transit times in DSS-clodronate-treated mice as in control animals. Furthermore, qPCR-analysis and immunofluorescence on colon muscularis samples revealed that factors associated with neuroinflammation and neurodegeneration, including Bax1, Hdac4, IL-18, Casp8 and Hif1a are overexpressed after DSS-treatment, but not in the case of concurrent L-clodronate administration. Our findings highlight that MM-infiltration in the muscularis layer is responsible for colitis-associated dysmotility and enteric neuronal dysfunction along with the release of mediators associated with neurodegeneration in a murine experimental model., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
35. Antineoplastic drugs inducing cardiac and vascular toxicity - An update.
- Author
-
Barachini S, Ghelardoni S, Varga ZV, Mehanna RA, Montt-Guevara MM, Ferdinandy P, and Madonna R
- Subjects
- Humans, Cardiotoxicity drug therapy, Cardiotoxicity etiology, Heart, Antineoplastic Agents adverse effects, Heart Diseases chemically induced, Heart Failure, Neoplasms drug therapy, Neoplasms complications
- Abstract
With the improvement in cancer prognosis due to advances in antitumor therapeutic protocols and new targeted and immunotherapies, we are witnessing a growing increase in survival, however, at the same timeincrease in morbidity among cancer survivors as a consequences of the increased cardiovascular adverse effects of antineoplastic drugs. Common cardiovascular complications of antineoplastic therapies may include cardiac complications such as arrhythmias, myocardial ischemia, left ventricular dysfunction culminating in heart failure as well as vascular complications including arterial hypertension, thromboembolic events, and accelerated atherosclerosis. The toxicity results from the fact that these drugs not only target cancer cells but also affect normal cells within the cardiovascular system. In this article, we review the clinical features and main mechanisms implicated in antineoplastic drug-induced cardiovascular toxicity, including oxidative stress, inflammation, immunothrombosis and growth factors-induced signaling pathways., Competing Interests: Declaration of Competing Interest The authors declare no conflict of interest., (Copyright © 2023. Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
36. Organ-specific model of simulated ischemia/reperfusion and hyperglycemia based on engineered heart tissue.
- Author
-
Pálóczi J, Paál Á, Pigler J, Kiss B, Rhoden A, Varga ZV, Ferdinandy P, Eschenhagen T, and Görbe A
- Subjects
- Rats, Animals, Ischemia, Reperfusion, Myocytes, Cardiac, Hyperglycemia
- Abstract
Here we aimed to establish an in vitro engineered heart tissue (EHT) co-morbidity mimicking model of ischemia-reperfusion injury and diabetes. EHTs were generated from primary neonatal rat cardiomyocytes. Hyperglycemic conditions or hyperosmolar controls were applied for one day to model acute hyperglycemia and for seven days to model chronic hyperglycemia. 120 min' simulated ischemia (SI) was followed by 120 min' reperfusion (R) and 1-day follow-up reperfusion (FR). Normoxic controls (N) were not subjected to SI/R. Half of the EHTs was paced, the other half was left unpaced. To assess cell injury, lactate-dehydrogenase (LDH) concentration was measured. Beating force and activity (frequency) were monitored as cardiomyocyte functional parameters. LDH-release indicated relevant cell injury after SI/N in each experimental condition, with much higher effects in the chronically hyperglycemic/hyperosmolar groups. SI stopped beating of EHTs in each condition, which returned during reperfusion, with weaker recovery in chronic conditions than in acute conditions. Acutely treated EHTs showed small LDH-release and ∼80% recovery of force during reperfusion and follow-up, while chronically treated EHTs showed a marked LDH-release, only ∼30% recovery with reperfusion and complete loss of beating activity during 24 h follow-up reperfusion. We conclude that EHTs respond differently to SI/R injury in acute and chronic hyperglycemia/hyperosmolarity, and that our EHT model is a novel in vitro combination of diabetes and ischemia-reperfusion., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests. P. Ferdinandy is the founder and CEO of Pharmahungary Group, a group of R&D companies. This research was funded by grants from the National Research, Development and Innovation Office (NKFIH) of Hungary (K139237). Project no. RRF-2.3.1-21-2022-00003 has been implemented with the support provided by the European Union. Project no. TKP2021-EGA-23 has been implemented with the support provided by the Ministry of Innovation and Technology of Hungary from the National Research, Development and Innovation Fund, financed under the TKP2021-EGA funding scheme., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
37. Droplet Digital PCR Is a Novel Screening Method Identifying Potential Cardiac G-Protein-Coupled Receptors as Candidate Pharmacological Targets in a Rat Model of Pressure-Overload-Induced Cardiac Dysfunction.
- Author
-
Sayour NV, Tóth VÉ, Nagy RN, Vörös I, Gergely TG, Onódi Z, Nagy N, Bödör C, Váradi B, Ruppert M, Radovits T, Bleckwedel F, Zelarayán LC, Pacher P, Ágg B, Görbe A, Ferdinandy P, and Varga ZV
- Subjects
- Male, Rats, Mice, Animals, Rats, Wistar, Myocytes, Cardiac, Polymerase Chain Reaction, Hypertrophy, Heart Failure genetics
- Abstract
The identification of novel drug targets is needed to improve the outcomes of heart failure (HF). G-protein-coupled receptors (GPCRs) represent the largest family of targets for already approved drugs, thus providing an opportunity for drug repurposing. Here, we aimed (i) to investigate the differential expressions of 288 cardiac GPCRs via droplet digital PCR (ddPCR) and bulk RNA sequencing (RNAseq) in a rat model of left ventricular pressure-overload; (ii) to compare RNAseq findings with those of ddPCR; and (iii) to screen and test for novel, translatable GPCR drug targets in HF. Male Wistar rats subjected to transverse aortic constriction (TAC, n = 5) showed significant systolic dysfunction vs. sham operated animals (SHAM, n = 5) via echocardiography. In TAC vs. SHAM hearts, RNAseq identified 69, and ddPCR identified 27 significantly differentially expressed GPCR mRNAs, 8 of which were identified using both methods, thus showing a correlation between the two methods. Of these, Prostaglandin-F2α-receptor ( Ptgfr ) was further investigated and localized on cardiomyocytes and fibroblasts in murine hearts via RNA-Scope. Antagonizing Ptgfr via AL-8810 reverted angiotensin-II-induced cardiomyocyte hypertrophy in vitro. In conclusion, using ddPCR as a novel screening method, we were able to identify GPCR targets in HF. We also show that the antagonism of Ptgfr could be a novel target in HF by alleviating cardiomyocyte hypertrophy.
- Published
- 2023
- Full Text
- View/download PDF
38. Editorial: Cardiovascular sequelae of chemotherapy and radiotherapy in cancer survivors: current evidence and perspectives.
- Author
-
Felici T, Skinner R, Ferdinandy P, Varga ZV, Lombardo A, and Camilli M
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
39. Inflammasome Activity in the Skeletal Muscle and Heart of Rodent Models for Duchenne Muscular Dystrophy.
- Author
-
Onódi Z, Szabó PL, Kucsera D, Pokreisz P, Dostal C, Hilber K, Oudit GY, Podesser BK, Ferdinandy P, Varga ZV, and Kiss A
- Subjects
- Mice, Rats, Animals, Inflammasomes metabolism, Mice, Inbred mdx, Rodentia metabolism, Muscle, Skeletal metabolism, Inflammation metabolism, Cytokines metabolism, Disease Models, Animal, Muscular Dystrophy, Duchenne metabolism
- Abstract
Duchenne muscular dystrophy (DMD) is characterized by wasting of muscles that leads to difficulty moving and premature death, mainly from heart failure. Glucocorticoids are applied in the management of the disease, supporting the hypothesis that inflammation may be driver as well as target. However, the inflammatory mechanisms during progression of cardiac and skeletal muscle dysfunction are still not well characterized. Our objective was to characterize the inflammasomes in myocardial and skeletal muscle in rodent models of DMD. Gastrocnemius and heart samples were collected from mdx mice and DMD
mdx rats (3 and 9-10 months). Inflammasome sensors and effectors were assessed by immunoblotting. Histology was used to assess leukocyte infiltration and fibrosis. In gastrocnemius, a tendency towards elevation of gasdermin D irrespective of the age of the animal was observed. The adaptor protein was elevated in the mdx mouse skeletal muscle and heart. Increased cleavage of the cytokines was observed in the skeletal muscle of the DMDmdx rats. Sensor or cytokine expression was not changed in the tissue samples of the mdx mice. In conclusion, inflammatory responses are distinct between the skeletal muscle and heart in relevant models of DMD. Inflammation tends to decrease over time, supporting the clinical observations that the efficacy of anti-inflammatory therapies might be more prominent in the early stage.- Published
- 2023
- Full Text
- View/download PDF
40. Telmisartan Is a Promising Agent for Managing Neuropathic Pain and Delaying Opioid Analgesic Tolerance in Rats.
- Author
-
Karádi DÁ, Galambos AR, Lakatos PP, Apenberg J, Abbood SK, Balogh M, Király K, Riba P, Essmat N, Szűcs E, Benyhe S, Varga ZV, Szökő É, Tábi T, and Al-Khrasani M
- Subjects
- Rats, Animals, Telmisartan pharmacology, Telmisartan therapeutic use, Losartan therapeutic use, Guanosine 5'-O-(3-Thiotriphosphate), Drug Tolerance, Analgesics pharmacology, Analgesics therapeutic use, Morphine pharmacology, Morphine therapeutic use, Glutamates therapeutic use, Analgesics, Opioid therapeutic use, Neuralgia drug therapy
- Abstract
Despite the large arsenal of analgesic medications, neuropathic pain (NP) management is not solved yet. Angiotensin II receptor type 1 (AT1) has been identified as a potential target in NP therapy. Here, we investigate the antiallodynic effect of AT1 blockers telmisartan and losartan, and particularly their combination with morphine on rat mononeuropathic pain following acute or chronic oral administration. The impact of telmisartan on morphine analgesic tolerance was also assessed using the rat tail-flick assay. Morphine potency and efficacy in spinal cord samples of treated neuropathic animals were assessed by [
35 S]GTPγS-binding assay. Finally, the glutamate content of the cerebrospinal fluid (CSF) was measured by capillary electrophoresis. Oral telmisartan or losartan in higher doses showed an acute antiallodynic effect. In the chronic treatment study, the combination of subanalgesic doses of telmisartan and morphine ameliorated allodynia and resulted in a leftward shift in the dose-response curve of morphine in the [35 S]GTPγS binding assay and increased CSF glutamate content. Telmisartan delayed morphine analgesic-tolerance development. Our study has identified a promising combination therapy composed of telmisartan and morphine for NP and opioid tolerance. Since telmisartan is an inhibitor of AT1 and activator of PPAR-γ, future studies are needed to analyze the effect of each component.- Published
- 2023
- Full Text
- View/download PDF
41. Drug repurposing for cardiovascular diseases: New targets and indications for probenecid.
- Author
-
Onódi Z, Koch S, Rubinstein J, Ferdinandy P, and Varga ZV
- Subjects
- Humans, Probenecid pharmacology, Probenecid therapeutic use, Drug Repositioning, Cardiovascular Diseases drug therapy, Heart Failure drug therapy, Myocardial Ischemia drug therapy, Coronary Artery Disease
- Abstract
The available pharmacological options in the management of cardiovascular diseases such as ischaemic heart disease and subsequent heart failure are effective in slowing the progression of this condition. However, the long-term prognosis is still poor, raising the demand for new therapeutic strategies. Drug repurposing is a time- and cost-effective drug development strategy that offers approved and abandoned drugs a new chance for new indications. Recently, drugs used for the management of gout-related inflammation such as canakinumab or colchicine have been considered for drug repurposing in cardiovascular indications. The old uricosuric drug, probenecid, has been identified as a novel therapeutic option in the management of specific cardiac diseases as well. Probenecid can modulate myocardial contractility and vascular tone and exerts anti-inflammatory properties. The mechanisms behind these beneficial effects might be related inhibition of inflammasomes, and to modulation purinergic-pannexin-1 signalling and TRPV2 channels, which are recently identified molecular targets of probenecid. In this review, we provide an overview on repurposing probenecid for ischaemic heart disease and subsequent heart failure by summarizing the related experimental and clinical data and propose its potential repurposing to treat cardiovascular diseases., (© 2022 The Authors. British Journal of Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.)
- Published
- 2023
- Full Text
- View/download PDF
42. Characterization of immune checkpoint inhibitor-induced cardiotoxicity reveals interleukin-17A as a driver of cardiac dysfunction after anti-PD-1 treatment.
- Author
-
Gergely TG, Kucsera D, Tóth VE, Kovács T, Sayour NV, Drobni ZD, Ruppert M, Petrovich B, Ágg B, Onódi Z, Fekete N, Pállinger É, Buzás EI, Yousif LI, Meijers WC, Radovits T, Merkely B, Ferdinandy P, and Varga ZV
- Subjects
- Mice, Animals, Immune Checkpoint Inhibitors adverse effects, Interleukin-17, Mice, Inbred C57BL, Inflammation complications, Cardiotoxicity etiology, Heart Diseases
- Abstract
Background and Purpose: Immune checkpoint inhibitors (ICI), such as anti-PD-1 monoclonal antibodies, have revolutionized cancer therapy by enhancing the cytotoxic effects of T-cells against tumours. However, enhanced T-cell activity also may cause myocarditis and cardiotoxicity. Our understanding of the mechanisms of ICI-induced cardiotoxicity is limited. Here, we aimed to investigate the effect of PD-1 inhibition on cardiac function and explore the molecular mechanisms of ICI-induced cardiotoxicity., Experimental Approach: C57BL6/J and BALB/c mice were treated with isotype control or anti-PD-1 antibody. Echocardiography was used to assess cardiac function. Cardiac transcriptomic changes were investigated by bulk RNA sequencing. Inflammatory changes were assessed by qRT-PCR and immunohistochemistry in heart, thymus, and spleen of the animals. In follow-up experiments, anti-CD4 and anti-IL-17A antibodies were used along with PD-1 blockade in C57BL/6J mice., Key Results: Anti-PD-1 treatment led to cardiac dysfunction and left ventricular dilation in C57BL/6J mice, with increased nitrosative stress. Only mild inflammation was observed in the heart. However, PD-1 inhibition resulted in enhanced thymic inflammatory signalling, where Il17a increased most prominently. In BALB/c mice, cardiac dysfunction was not evident, and thymic inflammatory activation was more balanced. Inhibition of IL-17A prevented anti-PD-1-induced cardiac dysfunction in C57BL6/J mice. Comparing myocardial transcriptomic changes in C57BL/6J and BALB/c mice, differentially regulated genes (Dmd, Ass1, Chrm2, Nfkbia, Stat3, Gsk3b, Cxcl9, Fxyd2, and Ldb3) were revealed, related to cardiac structure, signalling, and inflammation., Conclusions: PD-1 blockade induces cardiac dysfunction in mice with increased IL-17 signalling in the thymus. Pharmacological inhibition of IL-17A treatment prevents ICI-induced cardiac dysfunction., (© 2022 The Authors. British Journal of Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.)
- Published
- 2023
- Full Text
- View/download PDF
43. BRAF RNA is prognostic and widely expressed in lung adenocarcinoma.
- Author
-
Dora D, Vörös I, Varga ZV, Takacs P, Teglasi V, Moldvay J, and Lohinai Z
- Abstract
Background: BRAF is a critical member of proliferation pathways in cancer, and a mutation is present in only 2-4% of lung adenocarcinomas (LADC). There is no data available on the expression pattern of BRAF RNA that might result in enhanced signalling and drug resistance., Methods: LADC tissue samples (n=64) were fixed and processed into paraffin blocks. Tissue microarrays (TMA) were constructed, and RNAScope
® in situ hybridization (ISH) assay was performed for wild-type (WT) BRAF RNA. Apart from pathological assessment of tumor samples (grade, necrosis, vascular involvement and peritumoral infiltration), anti-programmed death ligand 1 (PD-L1) and anti-programmed death 1 (PD-1) immunohistochemistry and validation in public databases [The Cancer Genome Atlas (TCGA), Human Protein Atlas (HPA)] were carried out., Results: WT BRAF RNA is expressed in LADC, with no significant expressional difference between early-stage (I-II) and advanced-stage (III-IV) patients (P=0.317). Never smokers exhibited significantly increased BRAF expression (compared to current and ex-smokers, P<0.01) and tumor necrosis correlated significantly with BRAF expression (P=0.014). PD-L1 expression was assessed on tumor cells and immune cells, PD-1 expression was evaluated on immune cells. There was no significant difference in BRAF RNA expression between tumor cell PD-L1-high vs. low patients (P=0.124), but it was decreased in immune cell PD-L1-high patients (P=0.03). Kaplan-Meier survival analysis showed that high BRAF expression was associated with significantly decreased OS (P<0.01) and was an independent negative prognostic factor according to multivariate Cox hazard regression (P=0.024). TCGA validation cohort confirmed our findings regarding OS in early-stage patients (P=0.034)., Conclusions: We found an increased expression of BRAF RNA in all stages in LADC. High BRAF expression was associated with tumor necrosis, distinct immune checkpoint biology and outcomes. We recommend further evaluating the potential of targeting overexpressed BRAF pathways in LADC., Competing Interests: Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-22-449/coif). ZVV reports that this work was supported by the European Union’s Horizon 2020 Research and Innovation Programme under grant agreement No. 739593 and by a Momentum Research Grant from the Hungarian Academy of Sciences. ZL and ZVV acknowledge funding from the Hungarian National Research, Development and Innovation Office (OTKA #124652, OTKA #129664, and OTKA #128666, and OTKA #134751). IV was supported by the New National Excellence Program of the Ministry for Innovation and Technology from the source of the National Research, Development and Innovation Fund (No. ÚNKP-21-3-II-SE-14) and EFOP-3.6.3-VEKOP-16-2017-00009 (Project No. RRF-2.3.1-21-2022-00003, implemented with the support provided by the European Union). The authors have no other conflicts of interest to declare., (2023 Translational Lung Cancer Research. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
44. Single-cell transcriptomics reveal extracellular vesicles secretion with a cardiomyocyte proteostasis signature during pathological remodeling.
- Author
-
Schoger E, Bleckwedel F, Germena G, Rocha C, Tucholla P, Sobitov I, Möbius W, Sitte M, Lenz C, Samak M, Hinkel R, Varga ZV, Giricz Z, Salinas G, Gross JC, and Zelarayán LC
- Subjects
- Mice, Humans, Animals, Myocytes, Cardiac metabolism, Proteostasis, Proteomics, Transcriptome, Proteins metabolism, Gene Expression Profiling, Induced Pluripotent Stem Cells metabolism, Extracellular Vesicles genetics, Extracellular Vesicles metabolism
- Abstract
Aberrant Wnt activation has been reported in failing cardiomyocytes. Here we present single cell transcriptome profiling of hearts with inducible cardiomyocyte-specific Wnt activation (β-cat
Δex3 ) as well as with compensatory and failing hypertrophic remodeling. We show that functional enrichment analysis points to an involvement of extracellular vesicles (EVs) related processes in hearts of β-catΔex3 mice. A proteomic analysis of in vivo cardiac derived EVs from β-catΔex3 hearts has identified differentially enriched proteins involving 20 S proteasome constitutes, protein quality control (PQC), chaperones and associated cardiac proteins including α-Crystallin B (CRYAB) and sarcomeric components. The hypertrophic model confirms that cardiomyocytes reacted with an acute early transcriptional upregulation of exosome biogenesis processes and chaperones transcripts including CRYAB, which is ameliorated in advanced remodeling. Finally, human induced pluripotent stem cells (iPSC)-derived cardiomyocytes subjected to pharmacological Wnt activation recapitulated the increased expression of exosomal markers, CRYAB accumulation and increased PQC signaling. These findings reveal that secretion of EVs with a proteostasis signature contributes to early patho-physiological adaptation of cardiomyocytes, which may serve as a read-out of disease progression and can be used for monitoring cellular remodeling in vivo with a possible diagnostic and prognostic role in the future., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
45. IL-1β neutralization prevents diastolic dysfunction development, but lacks hepatoprotective effect in an aged mouse model of NASH.
- Author
-
Kucsera D, Tóth VE, Sayour NV, Kovács T, Gergely TG, Ruppert M, Radovits T, Fábián A, Kovács A, Merkely B, Ferdinandy P, and Varga ZV
- Subjects
- Male, Mice, Animals, Interleukin-1beta metabolism, Liver metabolism, Liver Cirrhosis pathology, Disease Models, Animal, Fibrosis, Mice, Inbred C57BL, Non-alcoholic Fatty Liver Disease pathology
- Abstract
Interleukin-1β (IL-1β) is a key mediator of non-alcoholic steatohepatitis (NASH), a chronic liver disease, and of systemic inflammation-driven aging. IL-1β contributes to cardio-metabolic decline, and may promote hepatic oncogenic transformation. Therefore, IL-1β is a potential therapeutic target in these pathologies. We aimed to investigate the hepatic and cardiac effects of an IL-1β targeting monoclonal antibody in an aged mouse model of NASH. 24 months old male C57Bl/6J mice were fed with control or choline deficient (CDAA) diet and were treated with isotype control or anti-IL-1β Mab for 8 weeks. Cardiac functions were assessed by conventional-and 2D speckle tracking echocardiography. Liver samples were analyzed by immunohistochemistry and qRT-PCR. Echocardiography revealed improved cardiac diastolic function in anti-IL-1β treated mice with NASH. Marked hepatic fibrosis developed in CDAA-fed group, but IL-1β inhibition affected fibrosis only at transcriptomic level. Hepatic inflammation was not affected by the IL-1β inhibitor. PCNA staining revealed intensive hepatocyte proliferation in CDAA-fed animals, which was not influenced by neutralization of IL-1β. IL-1β inhibition increased hepatic expression of Pd-1 and Ctla4, while Pd-l1 expression increased in NASH. In conclusion, IL-1β inhibition improved cardiac diastolic function, but did not ameliorate features of NASH; moreover, even promoted hepatic immune checkpoint expression, with concomitant NASH-related hepatocellular proliferation., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
46. Inflammasome activation in end-stage heart failure-associated atrial fibrillation.
- Author
-
Kugler S, Onódi Z, Ruppert M, Sayour AA, Oláh A, Benke K, Ferdinandy P, Merkely B, Radovits T, and Varga ZV
- Subjects
- Caspases, Fibrosis, Humans, Inflammasomes metabolism, Inflammation, Male, Atrial Fibrillation complications, Heart Failure etiology
- Abstract
Aims: Inflammatory pathways are increasingly recognized as an important factor in the pathophysiology of both heart failure (HF) and atrial fibrillation (AF). However, there is no data about inflammation-related histological and molecular alterations in HF-associated AF. The objective of our study was to investigate inflammatory pathways and fibrosis in end-stage HF-associated AF., Methods and Results: Left atrial samples of 24 male patients with end stage ischemic HF undergoing heart transplantation were analysed. Twelve patients suffered from sustained AF while the others had no documented AF. The expression of inflammasome sensors and their downstream signalling were investigated by Western blot. No differences were observed in the expression of inflammasome sensors between the two groups, while cleaved caspase-1 increased tendentiously in the AF group (P = 0.051). Cleaved caspase-1 also showed significant correlation with the expression of interleukin-1β and its cleaved form in the total population and in the AF group (P < 0.05). The presence of myocardial and epicardial macrophages were assessed by ionized calcium-binding adaptor molecule 1 (Iba1) immunostaining. Number of macrophages showed a tendency towards elevation in the left atrial myocardium and epicardium of AF compared with SR group. The amount of total and interstitial fibrosis was determined on Masson's trichrome-stained sections. Histological assessment revealed no difference between AF and SR groups in the amount of either total or interstitial fibrosis., Conclusions: This is the first study on inflammation-related differences between HF with SR or AF showing elevated inflammasome activity and enhanced macrophage infiltration in left atrial samples of patients with AF., (© 2022 The Authors. ESC Heart Failure published by John Wiley & Sons Ltd on behalf of European Society of Cardiology.)
- Published
- 2022
- Full Text
- View/download PDF
47. Saxagliptin Cardiotoxicity in Chronic Heart Failure: The Role of DPP4 in the Regulation of Neuropeptide Tone.
- Author
-
Vörös I, Onódi Z, Tóth VÉ, Gergely TG, Sághy É, Görbe A, Kemény Á, Leszek P, Helyes Z, Ferdinandy P, and Varga ZV
- Abstract
Dipeptidyl-peptidase-4 (DPP4) inhibitors are novel medicines for diabetes. The SAVOR-TIMI-53 clinical trial revealed increased heart-failure-associated hospitalization in saxagliptin-treated patients. Although this side effect could limit therapeutic use, the mechanism of this potential cardiotoxicity is unclear. We aimed to establish a cellular platform to investigate DPP4 inhibition and the role of its neuropeptide substrates substance P (SP) and neuropeptide Y (NPY), and to determine the expression of DDP4 and its neuropeptide substrates in the human heart. Western blot, radio-, enzyme-linked immuno-, and RNA scope assays were performed to investigate the expression of DPP4 and its substrates in human hearts. Calcein-based viability measurements and scratch assays were used to test the potential toxicity of DPP4 inhibitors. Cardiac expression of DPP4 and NPY decreased in heart failure patients. In human hearts, DPP4 mRNA is detectable mainly in cardiomyocytes and endothelium. Treatment with DPP4 inhibitors alone/in combination with neuropeptides did not affect viability but in scratch assays neuropeptides decreased, while saxagliptin co-administration increased fibroblast migration in isolated neonatal rat cardiomyocyte-fibroblast co-culture. Decreased DPP4 activity takes part in the pathophysiology of end-stage heart failure. DPP4 compensates against the elevated sympathetic activity and altered neuropeptide tone. Its inhibition decreases this adaptive mechanism, thereby exacerbating myocardial damage.
- Published
- 2022
- Full Text
- View/download PDF
48. Cannabinoid receptor 2 activation alleviates diabetes-induced cardiac dysfunction, inflammation, oxidative stress, and fibrosis.
- Author
-
Rajesh M, Mukhopadhyay P, Bátkai S, Arif M, Varga ZV, Mátyás C, Paloczi J, Lehocki A, Haskó G, and Pacher P
- Subjects
- Animals, Fibrosis, Inflammation pathology, Mice, Oxidative Stress, Receptors, Cannabinoid metabolism, Receptors, Cannabinoid therapeutic use, Diabetes Mellitus, Experimental complications, Diabetes Mellitus, Experimental drug therapy, Diabetic Cardiomyopathies etiology, Diabetic Cardiomyopathies metabolism, Diabetic Cardiomyopathies pathology
- Abstract
Diabetes mellitus promotes accelerated cardiovascular aging and inflammation, which in turn facilitate the development of cardiomyopathy/heart failure. High glucose-induced oxidative/nitrative stress, activation of various pro-inflammatory, and cell death pathways are critical in the initiation and progression of the changes culminating in diabetic cardiomyopathy. Cannabinoid 2 receptor (CB
2 R) activation in inflammatory cells and activated endothelium attenuates the pathological changes associated with atherosclerosis, myocardial infarction, stroke, and hepatic cardiomyopathy. In this study, we explored the role of CB2 R signaling in myocardial dysfunction, oxidative/nitrative stress, inflammation, cell death, remodeling, and fibrosis associated with diabetic cardiomyopathy in type 1 diabetic mice. Control human heart left ventricles and atrial appendages, similarly to mouse hearts, had negligible CB2 R expression determine by RNA sequencing or real-time RT-PCR. Diabetic cardiomyopathy was characterized by impaired diastolic and systolic cardiac function, enhanced myocardial CB2 R expression, oxidative/nitrative stress, and pro-inflammatory response (tumor necrosis factor-α, interleukin-1β, intracellular adhesion molecule 1, macrophage inflammatory protein-1, monocyte chemoattractant protein-1), macrophage infiltration, fibrosis, and cell death. Pharmacological activation of CB2 R with a selective agonist attenuated diabetes-induced inflammation, oxidative/nitrative stress, fibrosis and cell demise, and consequent cardiac dysfunction without affecting hyperglycemia. In contrast, genetic deletion of CB2 R aggravated myocardial pathology. Thus, selective activation of CB2 R ameliorates diabetes-induced myocardial tissue injury and preserves the functional contractile capacity of the myocardium in the diabetic milieu. This is particularly encouraging, since unlike CB1 R agonists, CB2 R agonists do not elicit psychoactive activity and cardiovascular side effects and are potential clinical candidates in the treatment of diabetic cardiovascular and other complications., (© 2022. This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply.)- Published
- 2022
- Full Text
- View/download PDF
49. Correction to: Cannabinoid receptor 2 activation alleviates diabetes-induced cardiac dysfunction, inflammation, oxidative stress, and fibrosis.
- Author
-
Rajesh M, Mukhopadhyay P, Bátkai S, Arif M, Varga ZV, Mátyás C, Paloczi J, Lehocki A, Haskó G, and Pacher P
- Published
- 2022
- Full Text
- View/download PDF
50. Systematic transcriptomic and phenotypic characterization of human and murine cardiac myocyte cell lines and primary cardiomyocytes reveals serious limitations and low resemblances to adult cardiac phenotype.
- Author
-
Onódi Z, Visnovitz T, Kiss B, Hambalkó S, Koncz A, Ágg B, Váradi B, Tóth VÉ, Nagy RN, Gergely TG, Gergő D, Makkos A, Pelyhe C, Varga N, Reé D, Apáti Á, Leszek P, Kovács T, Nagy N, Ferdinandy P, Buzás EI, Görbe A, Giricz Z, and Varga ZV
- Subjects
- Animals, Biomarkers metabolism, Cell Differentiation genetics, Cell Line, Humans, Mice, Phenotype, Transcriptome, Induced Pluripotent Stem Cells metabolism, Myocytes, Cardiac metabolism
- Abstract
Background: Cardiac cell lines and primary cells are widely used in cardiovascular research. Despite increasing number of publications using these models, comparative characterization of these cell lines has not been performed, therefore, their limitations are undetermined. We aimed to compare cardiac cell lines to primary cardiomyocytes and to mature cardiac tissues in a systematic manner., Methods and Results: Cardiac cell lines (H9C2, AC16, HL-1) were differentiated with widely used protocols. Left ventricular tissue, neonatal primary cardiomyocytes, and human induced pluripotent stem cell-derived cardiomyocytes served as reference tissue or cells. RNA expression of cardiac markers (e.g. Tnnt2, Ryr2) was markedly lower in cell lines compared to references. Differentiation induced increase in cardiac- and decrease in embryonic markers however, the overall transcriptomic profile and annotation to relevant biological processes showed consistently less pronounced cardiac phenotype in all cell lines in comparison to the corresponding references. Immunocytochemistry confirmed low expressions of structural protein sarcomeric alpha-actinin, troponin I and caveolin-3 in cell lines. Susceptibility of cell lines to sI/R injury in terms of viability as well as mitochondrial polarization differed from the primary cells irrespective of their degree of differentiation., Conclusion: Expression patterns of cardiomyocyte markers and whole transcriptomic profile, as well as response to sI/R, and to hypertrophic stimuli indicate low-to-moderate similarity of cell lines to primary cells/cardiac tissues regardless their differentiation. Low resemblance of cell lines to mature adult cardiac tissue limits their potential use. Low translational value should be taken into account while choosing a particular cell line to model cardiomyocytes., (Copyright © 2021 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.