128 results on '"Zuurbier CJ"'
Search Results
2. Reducing mitochondrial bound hexokinase II mediates transition from non-injurious into injurious ischemia/reperfusion of the intact heart
- Author
-
Nederlof, R, Gurel-Gurevin, E, Eerbeek, O, Xie, CQ, Deijs, GS, Konkel, M, Hu, J, Weber, NC, Schumacher, CA, Baartscheer, A, Mik, Bert, Hollmann, MW, Akar, FG, Zuurbier, CJ, Nederlof, R, Gurel-Gurevin, E, Eerbeek, O, Xie, CQ, Deijs, GS, Konkel, M, Hu, J, Weber, NC, Schumacher, CA, Baartscheer, A, Mik, Bert, Hollmann, MW, Akar, FG, and Zuurbier, CJ
- Published
- 2017
3. Increased in vivo mitochondrial oxygenation with right ventricular failure induced by pulmonary arterial hypertension: mitochondrial inhibition as driver of cardiac failure?
- Author
-
Balestra, Gianmarco, Mik, Bert, Eerbeek, O, Specht, Patricia, van der Laarse, WJ, Zuurbier, CJ, Balestra, Gianmarco, Mik, Bert, Eerbeek, O, Specht, Patricia, van der Laarse, WJ, and Zuurbier, CJ
- Abstract
Background: The leading cause of mortality due to pulmonary arterial hypertension (PAH) is failure of the cardiac right ventricle. It has long been hypothesized that during the development of chronic cardiac failure the heart becomes energy deprived, possibly due to shortage of oxygen at the level of cardiomyocyte mitochondria. However, direct evaluation of oxygen tension levels within the in vivo right ventricle during PAH is currently lacking. Here we directly evaluated this hypothesis by using a recently reported technique of oxygen-dependent quenching of delayed fluorescence of mitochondrial protoprophyrin IX, to determine the distribution of mitochondrial oxygen tension (mitoPO(2)) within the right ventricle (RV) subjected to progressive PAH. Methods: PAH was induced through a single injection of monocrotaline (MCT). Control (saline-injected), compensated RV hypertrophy (30 mg/kg MCT; MCT30), and RV failure (60 mg/kg MCT; MCT60) rats were compared 4 wk after treatment. The distribution of mitoPO(2) within the RV was determined in mechanically-ventilated, anaesthetized animals, applying different inspired oxygen (FiO(2)) levels and two increment dosages of dobutamine. Results: MCT60 resulted in RV failure (increased mortality, weight loss, increased lung weight), MCT30 resulted in compensated RV hypertrophy. At 30% or 40% FiO(2), necessary to obtain physiological arterial PO2 in the diseased animals, RV failure rats had significantly less mitochondria (15% of total mitochondria) in the 0-20 mmHg mitoPO(2) range than hypertrophied RV rats (48%) or control rats (54%). Only when oxygen supply was reduced to 21% FiO(2), resulting in low arterial PO2 for the MCT60 animals, or when oxygen demand increased with high dose dobutamine, the number of failing RV mitochondria with low oxygen became similar to control RV. In addition, metabolic enzyme analysis revealed similar mitochondrial mass, increased glycolytic hexokinase activity following MCT, with increased lactate d
- Published
- 2015
4. The renal microcirculation as a target for the treatment of acute kidney injury in models of critical illness
- Author
-
Ergin, Bulent, Gommers, D. A. M. P. J., Ince, Can, Zuurbier, Coert J., Translational Physiology, ACS - Atherosclerosis & ischemic syndromes, ACS - Microcirculation, Graduate School, Gommers, Diederik, Zuurbier, CJ, and Intensive Care
- Published
- 2018
5. Empagliflozin prevents TNF-α induced endothelial dysfunction under flow -the potential involvement of calcium and sodium-hydrogen exchanger.
- Author
-
Li X, Wang M, Wolfsgruber M, Klatt OC, Hollmann MW, Preckel B, Zuurbier CJ, and Weber NC
- Subjects
- Humans, Nitric Oxide metabolism, Nitric Oxide Synthase Type III metabolism, Sodium-Glucose Transporter 2 Inhibitors pharmacology, Endothelium, Vascular drug effects, Endothelium, Vascular metabolism, Endothelium, Vascular cytology, Tumor Necrosis Factor-alpha metabolism, Glucosides pharmacology, Calcium metabolism, Benzhydryl Compounds pharmacology, Sodium-Hydrogen Exchanger 1 metabolism, Sodium-Hydrogen Exchanger 1 genetics, Reactive Oxygen Species metabolism, Endothelial Cells drug effects, Endothelial Cells metabolism
- Abstract
Background: Empagliflozin (EMPA) attenuates inflammation-induced ROS generation in static endothelial cells through inhibition of sodium hydrogen exchanger 1 (NHE1) and modulation of ion homeostasis. We hypothesize that EMPA will alleviate TNF-α stimulated endothelial dysfunction under flow conditions, and that this might be mediated by NHE1 and intracellular Ca
2+ ., Methods: Human coronary artery endothelial cells were pre-treated with EMPA or vehicle before starting flow with or without TNF-α. Intracellular Ca2+ was recorded for 5 min at the start of flow. ROS generation and NO bioavailability, Piezo-1, cytokines, adhesion molecules, VE-cadherin and eNOS were detected after 6 h. BAPTA-AM was applied to chelate intracellular Ca2+ and NHE1 was knocked down with specific siRNA., Results: Under flow conditions, EMPA inhibited ROS production and [Ca2+ ] increase in cells exposed to TNF-α (P < 0.05). BAPTA-AM and NHE1 knockdown both reduced ROS generation (P < 0.05), and genetical inhibition of NHE1 led to reduction of intracellular [Ca2+ ] in HCAECs receiving TNF-α (P < 0.05). Yet, EMPA showed no effect on the increased cytokine production, adhesion molecule expression and phosphorylation of eNOS in endothelial cells exposed to TNF-α., Conclusion: EMPA mitigates increased ROS production and impaired NO bioavailability in TNF-α stimulated cells under flow. The anti-oxidative effect of EMPA is mediated by the decreased intracellular [Ca2+ ] following NHE1 inhibition., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2025
- Full Text
- View/download PDF
6. Empagliflozin prevents heart failure through inhibition of the NHE1-NO pathway, independent of SGLT2.
- Author
-
Chen S, Wang Q, Bakker D, Hu X, Zhang L, van der Made I, Tebbens AM, Kovácsházi C, Giricz Z, Brenner GB, Ferdinandy P, Schaart G, Gemmink A, Hesselink MKC, Rivaud MR, Pieper MP, Hollmann MW, Weber NC, Balligand JL, Creemers EE, Coronel R, and Zuurbier CJ
- Subjects
- Animals, Male, Mice, Mice, Inbred C57BL, Disease Models, Animal, Sodium-Hydrogen Exchangers metabolism, Sodium-Hydrogen Exchangers genetics, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Heart Failure metabolism, Heart Failure prevention & control, Heart Failure drug therapy, Heart Failure pathology, Sodium-Hydrogen Exchanger 1 metabolism, Sodium-Hydrogen Exchanger 1 genetics, Glucosides pharmacology, Benzhydryl Compounds pharmacology, Sodium-Glucose Transporter 2 Inhibitors pharmacology, Mice, Knockout, Nitric Oxide metabolism, Sodium-Glucose Transporter 2 metabolism, Sodium-Glucose Transporter 2 genetics, Signal Transduction drug effects
- Abstract
Sodium glucose cotransporter 2 inhibitors (SGLT2i) constitute the only medication class that consistently prevents or attenuates human heart failure (HF) independent of ejection fraction. We have suggested earlier that the protective mechanisms of the SGLT2i Empagliflozin (EMPA) are mediated through reductions in the sodium hydrogen exchanger 1 (NHE1)-nitric oxide (NO) pathway, independent of SGLT2. Here, we examined the role of SGLT2, NHE1 and NO in a murine TAC/DOCA model of HF. SGLT2 knockout mice only showed attenuated systolic dysfunction without having an effect on other signs of HF. EMPA protected against systolic and diastolic dysfunction, hypertrophy, fibrosis, increased Nppa/Nppb mRNA expression and lung/liver edema. In addition, EMPA prevented increases in oxidative stress, sodium calcium exchanger expression and calcium/calmodulin-dependent protein kinase II activation to an equal degree in WT and SGLT2 KO animals. In particular, while NHE1 activity was increased in isolated cardiomyocytes from untreated HF, EMPA treatment prevented this. Since SGLT2 is not required for the protective effects of EMPA, the pathway between NHE1 and NO was further explored in SGLT2 KO animals. In vivo treatment with the specific NHE1-inhibitor Cariporide mimicked the protection by EMPA, without additional protection by EMPA. On the other hand, in vivo inhibition of NOS with L-NAME deteriorated HF and prevented protection by EMPA. In conclusion, the data support that the beneficial effects of EMPA are mediated through the NHE1-NO pathway in TAC/DOCA-induced heart failure and not through SGLT2 inhibition., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
7. Ketonaemia during cardiopulmonary bypass surgery: a prospective observational study.
- Author
-
Snel LIP, Li X, Weber NC, Zuurbier CJ, Preckel B, van Raalte DH, Hermanides J, and Hulst AH
- Subjects
- Aged, Female, Humans, Male, Middle Aged, Intraoperative Complications, Ketosis etiology, Prospective Studies, Cardiopulmonary Bypass adverse effects
- Published
- 2024
- Full Text
- View/download PDF
8. Insulin and glycolysis dependency of cardioprotection by nicotinamide riboside.
- Author
-
Xiao Y, Wang Q, Zhang H, Nederlof R, Bakker D, Siadari BA, Wesselink MW, Preckel B, Weber NC, Hollmann MW, Schomakers BV, van Weeghel M, and Zuurbier CJ
- Subjects
- Animals, Mice, Citric Acid Cycle drug effects, Disease Models, Animal, Isolated Heart Preparation, Metabolomics, Mice, Inbred C57BL, Myocardium metabolism, NAD metabolism, Cardiotonic Agents pharmacology, Glycolysis drug effects, Insulin metabolism, Myocardial Reperfusion Injury metabolism, Myocardial Reperfusion Injury prevention & control, Niacinamide pharmacology, Niacinamide analogs & derivatives, Pyridinium Compounds pharmacology
- Abstract
Decreased nicotinamide adenine dinucleotide (NAD
+ ) levels contribute to various pathologies such as ageing, diabetes, heart failure and ischemia-reperfusion injury (IRI). Nicotinamide riboside (NR) has emerged as a promising therapeutic NAD+ precursor due to efficient NAD+ elevation and was recently shown to be the only agent able to reduce cardiac IRI in models employing clinically relevant anesthesia. However, through which metabolic pathway(s) NR mediates IRI protection remains unknown. Furthermore, the influence of insulin, a known modulator of cardioprotective efficacy, on the protective effects of NR has not been investigated. Here, we used the isolated mouse heart allowing cardiac metabolic control to investigate: (1) whether NR can protect the isolated heart against IRI, (2) the metabolic pathways underlying NR-mediated protection, and (3) whether insulin abrogates NR protection. NR protection against cardiac IRI and effects on metabolic pathways employing metabolomics for determination of changes in metabolic intermediates, and13 C-glucose fluxomics for determination of metabolic pathway activities (glycolysis, pentose phosphate pathway (PPP) and mitochondrial/tricarboxylic acid cycle (TCA cycle) activities), were examined in isolated C57BL/6N mouse hearts perfused with either (a) glucose + fatty acids (FA) ("mild glycolysis group"), (b) lactate + pyruvate + FA ("no glycolysis group"), or (c) glucose + FA + insulin ("high glycolysis group"). NR increased cardiac NAD+ in all three metabolic groups. In glucose + FA perfused hearts, NR reduced IR injury, increased glycolytic intermediate phosphoenolpyruvate (PEP), TCA intermediate succinate and PPP intermediates ribose-5P (R5P) / sedoheptulose-7P (S7P), and was associated with activated glycolysis, without changes in TCA cycle or PPP activities. In the "no glycolysis" hearts, NR protection was lost, whereas NR still increased S7P. In the insulin hearts, glycolysis was largely accelerated, and NR protection abrogated. NR still increased PPP intermediates, with now high13 C-labeling of S7P, but NR was unable to increase metabolic pathway activities, including glycolysis. Protection by NR against IRI is only present in hearts with low glycolysis, and is associated with activation of glycolysis. When activation of glycolysis was prevented, through either examining "no glycolysis" hearts or "high glycolysis" hearts, NR protection was abolished. The data suggest that NR's acute cardioprotective effects are mediated through glycolysis activation and are lost in the presence of insulin because of already elevated glycolysis., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
9. Empagliflozin mitigates cardiac hypertrophy through cardiac RSK/NHE-1 inhibition.
- Author
-
Chen S, Overberg K, Ghouse Z, Hollmann MW, Weber NC, Coronel R, and Zuurbier CJ
- Subjects
- Animals, Mice, Phosphorylation drug effects, Male, Sodium-Glucose Transporter 2 Inhibitors pharmacology, Cell Line, Rats, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Mice, Inbred C57BL, Signal Transduction drug effects, Sodium-Hydrogen Exchanger 1 metabolism, Sodium-Hydrogen Exchanger 1 antagonists & inhibitors, Glucosides pharmacology, Cardiomegaly drug therapy, Cardiomegaly pathology, Cardiomegaly prevention & control, Cardiomegaly metabolism, Ribosomal Protein S6 Kinases, 90-kDa metabolism, Ribosomal Protein S6 Kinases, 90-kDa antagonists & inhibitors, Benzhydryl Compounds pharmacology
- Abstract
Background: SGLT2i reduce cardiac hypertrophy, but underlying mechanisms remain unknown. Here we explore a role for serine/threonine kinases (STK) and sodium hydrogen exchanger 1(NHE1) activities in SGLT2i effects on cardiac hypertrophy., Methods: Isolated hearts from db/db mice were perfused with 1 µM EMPA, and STK phosphorylation sites were examined using unbiased multiplex analysis to detect the most affected STKs by EMPA. Subsequently, hypertrophy was induced in H9c2 cells with 50 µM phenylephrine (PE), and the role of the most affected STK (p90 ribosomal S6 kinase (RSK)) and NHE1 activity in hypertrophy and the protection by EMPA was evaluated., Results: In db/db mice hearts, EMPA most markedly reduced STK phosphorylation sites regulated by RSKL1, a member of the RSK family, and by Aurora A and B kinases. GO and KEGG analysis suggested that EMPA inhibits hypertrophy, cell cycle, cell senescence and FOXO pathways, illustrating inhibition of growth pathways. EMPA prevented PE-induced hypertrophy as evaluated by BNP and cell surface area in H9c2 cells. EMPA blocked PE-induced activation of NHE1. The specific NHE1 inhibitor Cariporide also prevented PE-induced hypertrophy without added effect of EMPA. EMPA blocked PE-induced RSK phosphorylation. The RSK inhibitor BIX02565 also suppressed PE-induced hypertrophy without added effect of EMPA. Cariporide mimicked EMPA's effects on PE-treated RSK phosphorylation. BIX02565 decreased PE-induced NHE1 activity, with no further decrease by EMPA., Conclusions: RSK inhibition by EMPA appears as a novel direct cardiac target of SGLT2i. Direct cardiac effects of EMPA exert their anti-hypertrophic effect through NHE-inhibition and subsequent RSK pathway inhibition., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. On behalf of all authors, the corresponding author CJ Zuurbier declares that none of the authors have known competing interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier Masson SAS.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
10. Hypertrophic cardiomyopathy dysfunction mimicked in human engineered heart tissue and improved by sodium-glucose cotransporter 2 inhibitors.
- Author
-
Wijnker PJM, Dinani R, van der Laan NC, Algül S, Knollmann BC, Verkerk AO, Remme CA, Zuurbier CJ, Kuster DWD, and van der Velden J
- Subjects
- Humans, Canagliflozin, Calcium, Myocytes, Cardiac pathology, Troponin T genetics, Sodium, Glucose, Induced Pluripotent Stem Cells, Cardiomyopathy, Hypertrophic drug therapy, Cardiomyopathy, Hypertrophic genetics, Cardiomyopathy, Hypertrophic pathology, Benzhydryl Compounds, Glucosides
- Abstract
Aims: Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiomyopathy, often caused by pathogenic sarcomere mutations. Early characteristics of HCM are diastolic dysfunction and hypercontractility. Treatment to prevent mutation-induced cardiac dysfunction is lacking. Sodium-glucose cotransporter 2 inhibitors (SGLT2i) are a group of antidiabetic drugs that recently showed beneficial cardiovascular outcomes in patients with acquired forms of heart failure. We here studied if SGLT2i represent a potential therapy to correct cardiomyocyte dysfunction induced by an HCM sarcomere mutation., Methods and Results: Contractility was measured of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) harbouring an HCM mutation cultured in 2D and in 3D engineered heart tissue (EHT). Mutations in the gene encoding β-myosin heavy chain (MYH7-R403Q) or cardiac troponin T (TNNT2-R92Q) were investigated. In 2D, intracellular [Ca2+], action potential and ion currents were determined. HCM mutations in hiPSC-CMs impaired relaxation or increased force, mimicking early features observed in human HCM. SGLT2i enhance the relaxation of hiPSC-CMs, to a larger extent in HCM compared to control hiPSC-CMs. Moreover, SGLT2i-effects on relaxation in R403Q EHT increased with culture duration, i.e. hiPSC-CMs maturation. Canagliflozin's effects on relaxation were more pronounced than empagliflozin and dapagliflozin. SGLT2i acutely altered Ca2+ handling in HCM hiPSC-CMs. Analyses of SGLT2i-mediated mechanisms that may underlie enhanced relaxation in mutant hiPSC-CMs excluded SGLT2, Na+/H+ exchanger, peak and late Nav1.5 currents, and L-type Ca2+ current, but indicate an important role for the Na+/Ca2+ exchanger. Indeed, electrophysiological measurements in mutant hiPSC-CM indicate that SGLT2i altered Na+/Ca2+ exchange current., Conclusion: SGLT2i (canagliflozin > dapagliflozin > empagliflozin) acutely enhance relaxation in human EHT, especially in HCM and upon prolonged culture. SGLT2i may represent a potential therapy to correct early cardiac dysfunction in HCM., Competing Interests: Conflict of interest: none declared., (© The Author(s) 2024. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2024
- Full Text
- View/download PDF
11. Empagliflozin prevents oxidative stress in human coronary artery endothelial cells via the NHE/PKC/NOX axis.
- Author
-
Li X, Wang M, Kalina JO, Preckel B, Hollmann MW, Albrecht M, Zuurbier CJ, and Weber NC
- Subjects
- Humans, Reactive Oxygen Species metabolism, Protein Kinase C metabolism, Ouabain metabolism, Oxidative Stress, Sodium-Hydrogen Exchangers metabolism, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Endothelial Cells metabolism, Coronary Vessels metabolism, Guanidines, Indoles, Glucosides, Maleimides, Sulfones, Benzhydryl Compounds
- Abstract
Background: Empagliflozin (EMPA) ameliorates reactive oxygen species (ROS) generation in human endothelial cells (ECs) exposed to 10 % stretch, but the underlying mechanisms are still unclear. Pathological stretch is supposed to stimulate protein kinase C (PKC) by increasing intracellular calcium (Ca
2+ ), therefore activating nicotinamide adenine dinucleotide phosphate oxidase (NOX) and promoting ROS production in human ECs. We hypothesized that EMPA inhibits stretch-induced NOX activation and ROS generation through preventing PKC activation., Methods: Human coronary artery endothelial cells (HCAECs) were pre-incubated for 2 h before exposure to cyclic stretch (5 % or 10 %) with either vehicle, EMPA or the PKC inhibitor LY-333531 or PKC siRNA. PKC activity, NOX activity and ROS production were detected after 24 h. Furthermore, the Ca2+ chelator BAPTA-AM, NCX inhibitor ORM-10962 or NCX siRNA, sodium/potassium pump inhibitor ouabain and sodium hydrogen exchanger (NHE) inhibitor cariporide were applied to explore the involvement of the NHE/Na+ /NCX/Ca2+ in the ROS inhibitory capacity of EMPA., Results: Compared to 5 % stretch, 10 % significantly increased PKC activity, which was reduced by EMPA and PKC inhibitor LY-333531. EMPA and LY-333531 showed a similar inhibitory capacity on NOX activity and ROS generation induced by 10 % stretch, which was not augmented by combined treatment with both drugs. PKC-β knockdown inhibits the NOX activation induced by Ca2+ and 10 % stretch. BAPTA, pharmacologic or genetic NCX inhibition and cariporide reduced Ca2+ in static HCAECs and prevented the activation of PKC and NOX in 10%-stretched cells. Ouabain increased ROS generation in cells exposed to 5 % stretch., Conclusion: EMPA reduced NOX activity via attenuation of the NHE/Na+ /NCX/Ca2+ /PKC axis, leading to less ROS generation in HCAECs exposed to 10 % stretch., Competing Interests: Declaration of competing interest Authors declare that there are no conflicts of interest., (Copyright © 2023 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
12. NLRX1 Prevents M2 Macrophage Polarization and Excessive Renal Fibrosis in Chronic Obstructive Nephropathy.
- Author
-
Liu Y, Kors L, Butter LM, Stokman G, Claessen N, Zuurbier CJ, Girardin SE, Leemans JC, Florquin S, and Tammaro A
- Subjects
- Animals, Mice, Macrophages, Genes, Regulator, Fibrosis, Transforming Growth Factor beta, Mitochondrial Proteins, Renal Insufficiency, Chronic
- Abstract
Background: Chronic kidney disease often leads to kidney dysfunction due to renal fibrosis, regardless of the initial cause of kidney damage. Macrophages are crucial players in the progression of renal fibrosis as they stimulate inflammation, activate fibroblasts, and contribute to extracellular matrix deposition, influenced by their metabolic state. Nucleotide-binding domain and LRR-containing protein X (NLRX1) is an innate immune receptor independent of inflammasomes and is found in mitochondria, and it plays a role in immune responses and cell metabolism. The specific impact of NLRX1 on macrophages and its involvement in renal fibrosis is not fully understood., Methods: To explore the specific role of NLRX1 in macrophages, bone-marrow-derived macrophages (BMDMs) extracted from wild-type (WT) and NLRX1 knockout (KO) mice were stimulated with pro-inflammatory and pro-fibrotic factors to induce M1 and M2 polarization in vitro. The expression levels of macrophage polarization markers ( Nos2 , Mgl1 , Arg1 , and Mrc1 ), as well as the secretion of transforming growth factor β (TGFβ), were measured using RT-PCR and ELISA. Seahorse-based bioenergetics analysis was used to assess mitochondrial respiration in naïve and polarized BMDMs obtained from WT and NLRX1 KO mice. In vivo, WT and NLRX1 KO mice were subjected to unilateral ureter obstruction (UUO) surgery to induce renal fibrosis. Kidney injury, macrophage phenotypic profile, and fibrosis markers were assessed using RT-PCR. Histological staining (PASD and Sirius red) was used to quantify kidney injury and fibrosis., Results: Compared to the WT group, an increased gene expression of M2 markers-including Mgl1 and Mrc1 -and enhanced TGFβ secretion were found in naïve BMDMs extracted from NLRX1 KO mice, indicating functional polarization towards the pro-fibrotic M2 subtype. NLRX1 KO naïve macrophages also showed a significantly enhanced oxygen consumption rate compared to WT cells and increased basal respiration and maximal respiration capacities that equal the level of M2-polarized macrophages. In vivo, we found that NLRX1 KO mice presented enhanced M2 polarization markers together with enhanced tubular injury and fibrosis demonstrated by augmented TGFβ levels, fibronectin, and collagen accumulation., Conclusions: Our findings highlight the unique role of NLRX1 in regulating the metabolism and function of macrophages, ultimately protecting against excessive renal injury and fibrosis in UUO.
- Published
- 2023
- Full Text
- View/download PDF
13. Protease XIV abolishes NHE inhibition by empagliflozin in cardiac cells.
- Author
-
Chen S, Schumacher CA, Van Amersfoorth SCM, Fiolet JWT, Baartscheer A, Veldkamp MW, Coronel R, and Zuurbier CJ
- Abstract
Background: SGLT2i directly inhibit the cardiac sodium-hydrogen exchanger-1 (NHE1) in isolated ventricular cardiomyocytes (CMs). However, other studies with SGLT2i have yielded conflicting results. This may be explained by methodological factors including cell isolation techniques, cell types and ambient pH. In this study, we tested whether the use of protease XIV (PXIV) may abrogate inhibition of SGLT2i on cardiac NHE1 activity in isolated rabbit CMs or rat cardiomyoblast cells (H9c2), in a pH dependent manner. Methods: Rabbit ventricular CMs were enzymatically isolated from Langendorff-perfused hearts during a 30-min perfusion period followed by a 25-min after-dissociation period, using a collagenase mixture without or with a low dose PXIV (0.009 mg/mL) present for different periods. Empagliflozin (EMPA) inhibition on NHE activity was then assessed at pH of 7.0, 7.2 and 7.4. In addition, effects of 10 min PXIV treatment were also evaluated in H9c2 cells for EMPA and cariporide NHE inhibition. Results: EMPA reduced NHE activity in rabbit CMs that were not exposed to PXIV treatment or undergoing a 35-min PXIV treatment, independent of pH levels. However, when exposure time to PXIV was extended to 55 min, NHE inhibition by Empa was completely abolished at all three pH levels. In H9c2 cells, NHE inhibition by EMPA was evident in non-treated cells but lost after 10-min incubation with PXIV. NHE inhibition by cariporide was unaffected by PXIV. Conclusion: The use of protease XIV in cardiac cell isolation procedures obliterates the inhibitory effects of SGLT2i on NHE1 activity in isolated cardiac cells, independent of pH., 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., (Copyright © 2023 Chen, Schumacher, Van Amersfoorth, Fiolet, Baartscheer, Veldkamp, Coronel and Zuurbier.)
- Published
- 2023
- Full Text
- View/download PDF
14. Cardioprotective efficacy of limb remote ischaemic preconditioning in rats: discrepancy between a meta-analysis and a three-centre in vivo study.
- Author
-
Sayour NV, Brenner GB, Makkos A, Kiss B, Kovácsházi C, Gergely TG, Aukrust SG, Tian H, Zenkl V, Gömöri K, Szabados T, Bencsik P, Heinen A, Schulz R, Baxter GF, Zuurbier CJ, Vokó Z, Ferdinandy P, and Giricz Z
- Subjects
- Rats, Male, Animals, Rats, Wistar, Reproducibility of Results, Ischemic Preconditioning methods, Myocardial Reperfusion Injury prevention & control
- Abstract
Aims: Remote ischaemic preconditioning (RIPC) is a robust cardioprotective intervention in preclinical studies. To establish a working and efficacious RIPC protocol in our laboratories, we performed randomized, blinded in vivo studies in three study centres in rats, with various RIPC protocols. To verify that our experimental settings are in good alignment with in vivo rat studies showing cardioprotection by limb RIPC, we performed a systematic review and meta-analysis. In addition, we investigated the importance of different study parameters., Methods and Results: Male Wistar rats were subjected to 20-45 min cardiac ischaemia followed by 120 min reperfusion with or without preceding RIPC by 3 or 4 × 5-5 min occlusion/reperfusion of one or two femoral vessels by clamping, tourniquet, or pressure cuff. RIPC did not reduce infarct size (IS), microvascular obstruction, or arrhythmias at any study centres. Systematic review and meta-analysis focusing on in vivo rat models of myocardial ischaemia/reperfusion injury with limb RIPC showed that RIPC reduces IS by 21.28% on average. In addition, the systematic review showed methodological heterogeneity and insufficient reporting of study parameters in a high proportion of studies., Conclusion: We report for the first time the lack of cardioprotection by RIPC in rats, assessed in individually randomized, blinded in vivo studies, involving three study centres, using different RIPC protocols. These results are in discrepancy with the meta-analysis of similar in vivo rat studies; however, no specific methodological reason could be identified by the systematic review, probably due to the overall insufficient reporting of several study parameters that did not improve over the past two decades. These results urge for publication of more well-designed and well-reported studies, irrespective of the outcome, which are required for preclinical reproducibility, and the development of clinically translatable cardioprotective interventions., Competing Interests: Conflict of interest: P.F. is the founder and CEO, Z.G. is involved in the management, and G.F.B is a member of the Advisory Board of Pharmahungary Group., (© The Author(s) 2023. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2023
- Full Text
- View/download PDF
15. Pharmacological Cardioprotection against Ischemia Reperfusion Injury-The Search for a Clinical Effective Therapy.
- Author
-
Wang Q, Zuurbier CJ, Huhn R, Torregroza C, Hollmann MW, Preckel B, van den Brom CE, and Weber NC
- Subjects
- Animals, Humans, Mitochondrial Permeability Transition Pore, Myocytes, Cardiac, Ischemia, Mitochondrial Membrane Transport Proteins, Myocardial Reperfusion Injury drug therapy, Myocardial Reperfusion Injury prevention & control
- Abstract
Pharmacological conditioning aims to protect the heart from myocardial ischemia-reperfusion injury (IRI). Despite extensive research in this area, today, a significant gap remains between experimental findings and clinical practice. This review provides an update on recent developments in pharmacological conditioning in the experimental setting and summarizes the clinical evidence of these cardioprotective strategies in the perioperative setting. We start describing the crucial cellular processes during ischemia and reperfusion that drive acute IRI through changes in critical compounds (∆G
ATP , Na+ , Ca2+ , pH, glycogen, succinate, glucose-6-phosphate, mitoHKII, acylcarnitines, BH4 , and NAD+ ). These compounds all precipitate common end-effector mechanisms of IRI, such as reactive oxygen species (ROS) generation, Ca2+ overload, and mitochondrial permeability transition pore opening (mPTP). We further discuss novel promising interventions targeting these processes, with emphasis on cardiomyocytes and the endothelium. The limited translatability from basic research to clinical practice is likely due to the lack of comorbidities, comedications, and peri-operative treatments in preclinical animal models, employing only monotherapy/monointervention, and the use of no-flow (always in preclinical models) versus low-flow ischemia (often in humans). Future research should focus on improved matching between preclinical models and clinical reality, and on aligning multitarget therapy with optimized dosing and timing towards the human condition., Competing Interests: The authors declare no conflict of interest.- Published
- 2023
- Full Text
- View/download PDF
16. Canagliflozin inhibits inflammasome activation in diabetic endothelial cells - Revealing a novel calcium-dependent anti-inflammatory effect of canagliflozin on human diabetic endothelial cells.
- Author
-
Li X, Kerindongo RP, Preckel B, Kalina JO, Hollmann MW, Zuurbier CJ, and Weber NC
- Subjects
- Humans, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Canagliflozin pharmacology, Calcium, Endothelial Cells metabolism, Caspase 1 metabolism, Reactive Oxygen Species metabolism, Tumor Necrosis Factor-alpha, Signal Transduction, Interleukin-1beta metabolism, Inflammasomes metabolism, Diabetes Mellitus
- Abstract
Background: Canagliflozin (CANA) shows anti-inflammatory and anti-oxidative effects on endothelial cells (ECs). In diabetes mellitus (DM), excessive reactive oxygen species (ROS) generation, increased intracellular calcium (Ca
2+ ) and enhanced extracellular signal regulated kinase (ERK) 1/2 phosphorylation are crucial precursors for inflammasome activation. We hypothesized that: (1) CANA prevents the TNF-α triggered ROS generation in ECs from diabetic donors and in turn suppresses the inflammasome activation; and (2) the anti-inflammatory effect of CANA is mediated via intracellular Ca2+ and ERK1/2., Methods: Human coronary artery endothelial cells from donors with DM (D-HCAECs) were pre-incubated with either CANA or vehicle for 2 h before exposure to 50 ng/ml TNF-α for 2-48 h. NAC was applied to scavenge ROS, BAPTA-AM to chelate intracellular Ca2+ , and PD 98059 to inhibit the activation of ERK1/2. Live cell imaging was performed at 6 h to measure ROS and intracellular Ca2+ . At 48 h, ELISA and infra-red western blot were applied to detect IL-1β, NLRP3, pro-caspase-1 and ASC., Results: 10 µM CANA significantly reduced TNF-α related ROS generation, IL-1β production and NLRP3 expression (P all <0.05), but NAC did not alter the inflammasome activation (P > 0.05). CANA and BAPTA both prevented intracellular Ca2+ increase in cells exposed to TNF-α (P both <0.05). Moreover, BAPTA and PD 98059 significantly reduced the TNF-α triggered IL-1β production as well as NLRP3 and pro-caspase-1 expression (P all <0.05)., Conclusion: CANA suppresses inflammasome activation by inhibition of (1) intracellular Ca2+ and (2) ERK1/2 phosphorylation, but not by ROS reduction., Competing Interests: Conflict of interest statement The authors declare that there are no conflict of interest., (Copyright © 2023 The Authors. Published by Elsevier Masson SAS.. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
17. Sodium Glucose Cotransporter-2 Inhibitor Empagliflozin Reduces Infarct Size Independently of Sodium Glucose Cotransporter-2.
- Author
-
Chen S, Wang Q, Christodoulou A, Mylonas N, Bakker D, Nederlof R, Hollmann MW, Weber NC, Coronel R, Wakker V, Christoffels VM, Andreadou I, and Zuurbier CJ
- Subjects
- Humans, Hypoglycemic Agents pharmacology, Hypoglycemic Agents therapeutic use, Benzhydryl Compounds pharmacology, Benzhydryl Compounds therapeutic use, Glucose, Sodium-Glucose Transporter 2 Inhibitors therapeutic use, Sodium-Glucose Transporter 2 Inhibitors pharmacology, Diabetes Mellitus, Type 2 drug therapy
- Published
- 2023
- Full Text
- View/download PDF
18. Interaction of Cardiovascular Nonmodifiable Risk Factors, Comorbidities and Comedications With Ischemia/Reperfusion Injury and Cardioprotection by Pharmacological Treatments and Ischemic Conditioning.
- Author
-
Ferdinandy P, Andreadou I, Baxter GF, Bøtker HE, Davidson SM, Dobrev D, Gersh BJ, Heusch G, Lecour S, Ruiz-Meana M, Zuurbier CJ, Hausenloy DJ, and Schulz R
- Subjects
- Animals, Humans, Risk Factors, Heart Disease Risk Factors, Ischemia, Myocardial Reperfusion Injury drug therapy, Myocardial Reperfusion Injury prevention & control, Myocardial Reperfusion Injury metabolism, Ischemic Preconditioning, Myocardial, Ischemic Postconditioning, Myocardial Ischemia drug therapy, Myocardial Ischemia prevention & control
- Abstract
Preconditioning, postconditioning, and remote conditioning of the myocardium enhance the ability of the heart to withstand a prolonged ischemia/reperfusion insult and the potential to provide novel therapeutic paradigms for cardioprotection. While many signaling pathways leading to endogenous cardioprotection have been elucidated in experimental studies over the past 30 years, no cardioprotective drug is on the market yet for that indication. One likely major reason for this failure to translate cardioprotection into patient benefit is the lack of rigorous and systematic preclinical evaluation of promising cardioprotective therapies prior to their clinical evaluation, since ischemic heart disease in humans is a complex disorder caused by or associated with cardiovascular risk factors and comorbidities. These risk factors and comorbidities induce fundamental alterations in cellular signaling cascades that affect the development of ischemia/reperfusion injury and responses to cardioprotective interventions. Moreover, some of the medications used to treat these comorbidities may impact on cardioprotection by again modifying cellular signaling pathways. The aim of this article is to review the recent evidence that cardiovascular risk factors as well as comorbidities and their medications may modify the response to cardioprotective interventions. We emphasize the critical need for taking into account the presence of cardiovascular risk factors as well as comorbidities and their concomitant medications when designing preclinical studies for the identification and validation of cardioprotective drug targets and clinical studies. This will hopefully maximize the success rate of developing rational approaches to effective cardioprotective therapies for the majority of patients with multiple comorbidities. SIGNIFICANCE STATEMENT: Ischemic heart disease is a major cause of mortality; however, there are still no cardioprotective drugs on the market. Most studies on cardioprotection have been undertaken in animal models of ischemia/reperfusion in the absence of comorbidities; however, ischemic heart disease develops with other systemic disorders (e.g., hypertension, hyperlipidemia, diabetes, atherosclerosis). Here we focus on the preclinical and clinical evidence showing how these comorbidities and their routine medications affect ischemia/reperfusion injury and interfere with cardioprotective strategies., (Copyright © 2023 by The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
19. Use of sodium-glucose cotransporter-2 inhibitors and the risk for sudden cardiac arrest and for all-cause death in patients with type 2 diabetes mellitus.
- Author
-
Eroglu TE, Coronel R, Zuurbier CJ, Blom M, de Boer A, and Souverein PC
- Subjects
- Humans, Cohort Studies, Hypoglycemic Agents adverse effects, Death, Sudden, Cardiac epidemiology, Death, Sudden, Cardiac etiology, Glucose, Sodium, Diabetes Mellitus, Type 2 complications, Diabetes Mellitus, Type 2 diagnosis, Diabetes Mellitus, Type 2 drug therapy, Sodium-Glucose Transporter 2 Inhibitors adverse effects
- Abstract
Aims: Sodium-glucose cotransporter-2 inhibitors (SGLT-2is) are antidiabetic agents that can have direct cardiac effects by impacting on cardiac ion transport mechanisms that control cardiac electrophysiology. We studied the association between SGLT-2i use and all-cause mortality and the risk of sudden cardiac arrest (SCA) in patients with type 2 diabetes., Methods: Using data from the UK Clinical Practice Research Datalink, a cohort study among patients initiating a new antidiabetic drug class on or after January 2013 through September 2020 was conducted. A Cox regression with time-dependent covariates was performed to estimate the hazard ratios (HRs) of SCA and all-cause mortality comparing SGLT-2is with other second- to third-line antidiabetic drugs. Stratified analyses were performed according to sex, diabetes duration (<5 or ≥5 years), and the presence of cardiovascular disease., Results: A total of 152 591 patients were included. Use of SGLT-2i was associated with a reduced HR of SCA when compared with other second- to third-line antidiabetic drugs after adjustment for common SCA risk factors, although this association marginally failed to reach statistical significance [HR: 0.62, 95% confidence interval (95% CI): 0.38-1.01]. The HR of all-cause mortality associated with SGLT-2i use when compared with other second- to third-line antidiabetics was 0.43 (95% CI: 0.39-0.48) and did not vary by sex, diabetes duration, or the presence of cardiovascular disease. SGLT-2i use remained associated with lower all-cause mortality in patients without concomitant insulin use (HR: 0.56, 95% CI: 0.50-0.63)., Conclusion: SGLT-2i use was associated with reduced all-cause mortality in patients with type 2 diabetes. The association between use of SGLT-2i and reduced risk of SCA was not statistically significant., (© The Author(s) 2022. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2022
- Full Text
- View/download PDF
20. Amelioration of endothelial dysfunction by sodium glucose co-transporter 2 inhibitors: pieces of the puzzle explaining their cardiovascular protection.
- Author
-
Li X, Preckel B, Hermanides J, Hollmann MW, Zuurbier CJ, and Weber NC
- Subjects
- Endothelial Cells, Glucose, Humans, Hypoglycemic Agents pharmacology, Sodium, Diabetes Mellitus, Type 2 drug therapy, Sodium-Glucose Transporter 2 Inhibitors pharmacology, Symporters, Vascular Diseases
- Abstract
Sodium glucose co-transporter 2 inhibitors (SGLT-2is) improve cardiovascular outcomes in both diabetic and non-diabetic patients. Preclinical studies suggest that SGLT-2is directly affect endothelial function in a glucose-independent manner. The effects of SGLT-2is include decreased oxidative stress and inflammatory reactions in endothelial cells. Furthermore, SGLT2is restore endothelium-related vasodilation and regulate angiogenesis. The favourable cardiovascular effects of SGLT-2is could be mediated via a number of pathways: (1) inhibition of the overactive sodium-hydrogen exchanger; (2) decreased expression of nicotinamide adenine dinucleotide phosphate oxidases; (3) alleviation of mitochondrial injury; (4) suppression of inflammation-related signalling pathways (e.g., by affecting NF-κB); (5) modulation of glycolysis; and (6) recovery of impaired NO bioavailability. This review focuses on the most recent progress and existing gaps in preclinical investigations concerning the direct effects of SGLT-2is on endothelial dysfunction and the mechanisms underlying such effects., (© 2022 The Authors. British Journal of Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.)
- Published
- 2022
- Full Text
- View/download PDF
21. Cardiac mechanisms of the beneficial effects of SGLT2 inhibitors in heart failure: Evidence for potential off-target effects.
- Author
-
Dyck JRB, Sossalla S, Hamdani N, Coronel R, Weber NC, Light PE, and Zuurbier CJ
- Subjects
- Humans, Myocardium metabolism, Sodium-Glucose Transporter 2 metabolism, Sodium-Glucose Transporter 2 therapeutic use, Diabetes Mellitus, Type 2, Heart Failure, Sodium-Glucose Transporter 2 Inhibitors adverse effects
- Abstract
Sodium glucose cotransporter 2 inhibitors (SGLT2i) constitute a promising drug treatment for heart failure patients with either preserved or reduced ejection fraction. Whereas SGLT2i were originally developed to target SGLT2 in the kidney to facilitate glucosuria in diabetic patients, it is becoming increasingly clear that these drugs also have important effects outside of the kidney. In this review we summarize the literature on cardiac effects of SGLT2i, focussing on pro-inflammatory and oxidative stress processes, ion transport mechanisms controlling sodium and calcium homeostasis and metabolic/mitochondrial pathways. These mechanisms are particularly important as disturbances in these pathways result in endothelial dysfunction, diastolic dysfunction, cardiac stiffness, and cardiac arrhythmias that together contribute to heart failure. We review the findings that support the concept that SGLT2i directly and beneficially interfere with inflammation, oxidative stress, ionic homeostasis, and metabolism within the cardiac cell. However, given the very low levels of SGLT2 in cardiac cells, the evidence suggests that SGLT2-independent effects of this class of drugs likely occurs via off-target effects in the myocardium. Thus, while there is still much to be understood about the various factors which determine how SGLT2i affect cardiac cells, much of the research clearly demonstrates that direct cardiac effects of these SGLT2i exist, albeit mediated via SGLT2-independent pathways, and these pathways may play a role in explaining the beneficial effects of SGLT2 inhibitors in heart failure., (Copyright © 2022 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
22. Cardioprotection by selective SGLT-2 inhibitors in a non-diabetic mouse model of myocardial ischemia/reperfusion injury: a class or a drug effect?
- Author
-
Nikolaou PE, Mylonas N, Makridakis M, Makrecka-Kuka M, Iliou A, Zerikiotis S, Efentakis P, Kampoukos S, Kostomitsopoulos N, Vilskersts R, Ikonomidis I, Lambadiari V, Zuurbier CJ, Latosinska A, Vlahou A, Dimitriadis G, Iliodromitis EK, and Andreadou I
- Subjects
- Animals, Disease Models, Animal, Fibroblast Growth Factor 2, Glucose, Mice, Mice, Inbred C57BL, Phosphatidylinositol 3-Kinases, Wortmannin, Diabetes Mellitus, Type 2 complications, Myocardial Reperfusion Injury drug therapy, Sodium-Glucose Transporter 2 Inhibitors pharmacology
- Abstract
Major clinical trials with sodium glucose co-transporter-2 inhibitors (SGLT-2i) exhibit protective effects against heart failure events, whereas inconsistencies regarding the cardiovascular death outcomes are observed. Therefore, we aimed to compare the selective SGLT-2i empagliflozin (EMPA), dapagliflozin (DAPA) and ertugliflozin (ERTU) in terms of infarct size (IS) reduction and to reveal the cardioprotective mechanism in healthy non-diabetic mice. C57BL/6 mice randomly received vehicle, EMPA (10 mg/kg/day) and DAPA or ERTU orally at the stoichiometrically equivalent dose (SED) for 7 days. 24 h-glucose urinary excretion was determined to verify SGLT-2 inhibition. IS of the region at risk was measured after 30 min ischemia (I), and 120 min reperfusion (R). In a second series, the ischemic myocardium was collected (10th min of R) for shotgun proteomics and evaluation of the cardioprotective signaling. In a third series, we evaluated the oxidative phosphorylation capacity (OXPHOS) and the mitochondrial fatty acid oxidation capacity by measuring the respiratory rates. Finally, Stattic, the STAT-3 inhibitor and wortmannin were administered in both EMPA and DAPA groups to establish causal relationships in the mechanism of protection. EMPA, DAPA and ERTU at the SED led to similar SGLT-2 inhibition as inferred by the significant increase in glucose excretion. EMPA and DAPA but not ERTU reduced IS. EMPA preserved mitochondrial functionality in complex I&II linked oxidative phosphorylation. EMPA and DAPA treatment led to NF-kB, RISK, STAT-3 activation and the downstream apoptosis reduction coinciding with IS reduction. Stattic and wortmannin attenuated the cardioprotection afforded by EMPA and DAPA. Among several upstream mediators, fibroblast growth factor-2 (FGF-2) and caveolin-3 were increased by EMPA and DAPA treatment. ERTU reduced IS only when given at the double dose of the SED (20 mg/kg/day). Short-term EMPA and DAPA, but not ERTU administration at the SED reduce IS in healthy non-diabetic mice. Cardioprotection is not correlated to SGLT-2 inhibition, is STAT-3 and PI3K dependent and associated with increased FGF-2 and Cav-3 expression., (© 2022. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany.)
- Published
- 2022
- Full Text
- View/download PDF
23. Direct cardiac effects of SGLT2 inhibitors.
- Author
-
Chen S, Coronel R, Hollmann MW, Weber NC, and Zuurbier CJ
- Subjects
- Benzhydryl Compounds pharmacology, Glucosides pharmacology, Humans, Inflammation, Sodium metabolism, Diabetes Mellitus, Type 2 diagnosis, Diabetes Mellitus, Type 2 drug therapy, Heart Failure drug therapy, Sodium-Glucose Transporter 2 Inhibitors adverse effects
- Abstract
Sodium-glucose-cotransporter 2 inhibitors (SGLT2is) demonstrate large cardiovascular benefit in both diabetic and non-diabetic, acute and chronic heart failure patients. These inhibitors have on-target (SGLT2 inhibition in the kidney) and off-target effects that likely both contribute to the reported cardiovascular benefit. Here we review the literature on direct effects of SGLT2is on various cardiac cells and derive at an unifying working hypothesis. SGLT2is acutely and directly (1) inhibit cardiac sodium transporters and alter ion homeostasis, (2) reduce inflammation and oxidative stress, (3) influence metabolism, and (4) improve cardiac function. We postulate that cardiac benefit modulated by SGLT2i's can be commonly attributed to their inhibition of sodium-loaders in the plasma membrane (NHE-1, Nav1.5, SGLT) affecting intracellular sodium-homeostasis (the sodium-interactome), thereby providing a unifying view on the various effects reported in separate studies. The SGLT2is effects are most apparent when cells or hearts are subjected to pathological conditions (reactive oxygen species, inflammation, acidosis, hypoxia, high saturated fatty acids, hypertension, hyperglycemia, and heart failure sympathetic stimulation) that are known to prime these plasmalemmal sodium-loaders. In conclusion, the cardiac sodium-interactome provides a unifying testable working hypothesis and a possible, at least partly, explanation to the clinical benefits of SGLT2is observed in the diseased patient., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
24. Empagliflozin reduces oxidative stress through inhibition of the novel inflammation/NHE/[Na + ] c /ROS-pathway in human endothelial cells.
- Author
-
Uthman L, Li X, Baartscheer A, Schumacher CA, Baumgart P, Hermanides J, Preckel B, Hollmann MW, Coronel R, Zuurbier CJ, and Weber NC
- Subjects
- Humans, Inflammation Mediators metabolism, Ouabain pharmacology, Tumor Necrosis Factor-alpha metabolism, Benzhydryl Compounds pharmacology, Endothelial Cells drug effects, Glucosides pharmacology, Reactive Oxygen Species metabolism, Sodium metabolism, Sodium-Glucose Transporter 2 Inhibitors pharmacology, Sodium-Hydrogen Exchangers drug effects
- Abstract
Inflammation causing oxidative stress in endothelial cells contributes to heart failure development. Sodium/glucose cotransporter 2 inhibitors (SGLT2i's) were shown to reduce heart failure hospitalization and oxidative stress. However, how inflammation causes oxidative stress in endothelial cells, and how SGLT2i's can reduce this is unknown. Here we hypothesized that 1) TNF-α activates the Na
+ /H+ exchanger (NHE) and raises cytoplasmatic Na+ ([Na+ ]c ), 2) increased [Na+ ]c causes reactive oxygen species (ROS) production, and 3) empagliflozin (EMPA) reduces inflammation-induced ROS through NHE inhibition and lowering of [Na+ ]c in human endothelial cells. Human umbilical vein endothelial cells (HUVECs) and human coronary artery endothelial cells (HCAECs) were incubated with vehicle (V), 10 ng/ml TNF-α, 1 µM EMPA or the NHE inhibitor Cariporide (CARI, 10 µM) and NHE activity, intracellular [Na+ ]c and ROS were analyzed. TNF-α enhanced NHE activity in HCAECs and HUVECs by 92% (p < 0.01) and 51% (p < 0.05), respectively, and increased [Na+ ]c from 8.2 ± 1.6 to 11.2 ± 0.1 mM (p < 0.05) in HCAECs. Increasing [Na+ ]c by ouabain elevated ROS generation in both HCAECs and HUVECs. EMPA inhibited NHE activity in HCAECs and in HUVECs. EMPA concomitantly lowered [Na+ ]c in both cell types. In both cell types, TNF α-induced ROS was lowered by EMPA or CARI, with no further ROS lowering by EMPA in the presence of CARI, indicating EMPA attenuated ROS through NHE inhibition. In conclusion, inflammation induces oxidative stress in human endothelial cells through NHE activation causing elevations in [Na+ ]c , a process that is inhibited by EMPA through NHE inhibition., (Copyright © 2021 The Authors. Published by Elsevier Masson SAS.. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
25. Sodium-glucose co-transporter 2 inhibitor empagliflozin inhibits the cardiac Na+/H+ exchanger 1: persistent inhibition under various experimental conditions.
- Author
-
Zuurbier CJ, Baartscheer A, Schumacher CA, Fiolet JWT, and Coronel R
- Subjects
- Animals, Buffers, Hydrogen-Ion Concentration, Male, Myocytes, Cardiac metabolism, Rabbits, Sodium-Hydrogen Exchanger 1 metabolism, Action Potentials drug effects, Benzhydryl Compounds pharmacology, Glucosides pharmacology, Myocytes, Cardiac drug effects, Sodium-Glucose Transporter 2 Inhibitors pharmacology, Sodium-Hydrogen Exchanger 1 antagonists & inhibitors
- Published
- 2021
- Full Text
- View/download PDF
26. Novel Anti-inflammatory Effects of Canagliflozin Involving Hexokinase II in Lipopolysaccharide-Stimulated Human Coronary Artery Endothelial Cells.
- Author
-
Uthman L, Kuschma M, Römer G, Boomsma M, Kessler J, Hermanides J, Hollmann MW, Preckel B, Zuurbier CJ, and Weber NC
- Subjects
- AMP-Activated Protein Kinases, Benzhydryl Compounds pharmacology, Dose-Response Relationship, Drug, Glucosides pharmacology, Humans, Hypoglycemic Agents pharmacology, Lipopolysaccharides pharmacology, MAP Kinase Signaling System drug effects, NF-kappa B drug effects, Canagliflozin pharmacology, Coronary Vessels drug effects, Endothelial Cells drug effects, Hexokinase drug effects, Inflammation Mediators antagonists & inhibitors, Sodium-Glucose Transporter 2 Inhibitors pharmacology
- Abstract
Purpose: Vascular inflammation and disturbed metabolism are observed in heart failure and type 2 diabetes mellitus. Glycolytic enzyme hexokinase II (HKII) is upregulated by inflammation. We hypothesized that SGLT2 inhibitors Canagliflozin (Cana), Empagliflozin (Empa) or Dapagliflozin (Dapa) reduces inflammation via HKII in endothelial cells, and that HKII-dependent inflammation is determined by ERK1/2, NF-κB. and/or AMPK activity in lipopolysaccharide (LPS)-stimulated human coronary artery endothelial cells (HCAECs)., Methods: HCAECs were pre-incubated with 3 μM or 10 μM Cana, 1 μM, 3 μM or 10 μM Empa or 0.5 μM, 3 μM or 10 μM Dapa (16 h) and subjected to 3 h LPS (1 μg/mL). HKII was silenced via siRNA transfection. Interleukin-6 (IL-6) release was measured by ELISA. Protein levels of HK I and II, ERK1/2, AMPK and NF-κB were detected using infra-red western blot., Results: LPS increased IL-6 release and ERK1/2 phosphorylation; Cana prevented these pro-inflammatory responses (IL-6: pg/ml, control 46 ± 2, LPS 280 ± 154 p < 0.01 vs. control, LPS + Cana 96 ± 40, p < 0.05 vs. LPS). Cana reduced HKII expression (HKII/GAPDH, control 0.91 ± 0.16, Cana 0.71 ± 0.13 p < 0.05 vs. control, LPS 1.02 ± 0.25, LPS + Cana 0.82 ± 0.24 p < 0.05 vs. LPS). Empa and Dapa were without effect on IL-6 release and HKII expression in the model used. Knockdown of HKII by 37% resulted caused partial loss of Cana-mediated IL-6 reduction (pg/ml, control 35 ± 5, LPS 188 ± 115 p < 0.05 vs. control, LPS + Cana 124 ± 75) and ERK1/2 activation by LPS. In LPS-stimulated HCAECs, Cana, but not Empa or Dapa, activated AMPK. AMPK activator A769662 reduced IL-6 release., Conclusion: Cana conveys anti-inflammatory actions in LPS-treated HCAECs through 1) reductions in HKII and ERK1/2 phosphorylation and 2) AMPK activation. These data suggest a novel anti-inflammatory mechanism of Cana through HKII., (© 2020. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
27. IMproving Preclinical Assessment of Cardioprotective Therapies (IMPACT) criteria: guidelines of the EU-CARDIOPROTECTION COST Action.
- Author
-
Lecour S, Andreadou I, Bøtker HE, Davidson SM, Heusch G, Ruiz-Meana M, Schulz R, Zuurbier CJ, Ferdinandy P, and Hausenloy DJ
- Subjects
- Animals, Humans, Heart Failure prevention & control, Myocardial Infarction, Reperfusion Injury
- Abstract
Acute myocardial infarction (AMI) and the heart failure (HF) which may follow are among the leading causes of death and disability worldwide. As such, new therapeutic interventions are still needed to protect the heart against acute ischemia/reperfusion injury to reduce myocardial infarct size and prevent the onset of HF in patients presenting with AMI. However, the clinical translation of cardioprotective interventions that have proven to be beneficial in preclinical animal studies, has been challenging. One likely major reason for this failure to translate cardioprotection into patient benefit is the lack of rigorous and systematic in vivo preclinical assessment of the efficacy of promising cardioprotective interventions prior to their clinical evaluation. To address this, we propose an in vivo set of step-by-step criteria for IMproving Preclinical Assessment of Cardioprotective Therapies ('IMPACT'), for investigators to consider adopting before embarking on clinical studies, the aim of which is to improve the likelihood of translating novel cardioprotective interventions into the clinical setting for patient benefit., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
28. Cardioprotecive Properties of Known Agents in Rat Ischemia-Reperfusion Model Under Clinically Relevant Conditions: Only the NAD Precursor Nicotinamide Riboside Reduces Infarct Size in Presence of Fentanyl, Midazolam and Cangrelor, but Not Propofol.
- Author
-
Xiao Y, Phelp P, Wang Q, Bakker D, Nederlof R, Hollmann MW, and Zuurbier CJ
- Abstract
Background: Cardioprotective strategies against ischemia-reperfusion injury (IRI) that remain effective in the clinical arena need to be developed. Therefore, maintained efficacy of cardioprotective strategies in the presence of drugs routinely used clinically (e.g., opiates, benzodiazepines, P2Y
12 antagonist, propofol) need to be identified in preclinical models. Methods: Here, we examined the efficacy of promising cardioprotective compounds [fingolimod (Fingo), empagliflozin (Empa), melatonin (Mela) and nicotinamide riboside (NR)] administered i.v. as bolus before start ischemia. Infarct size as percentage of the area of risk (IS%) was determined following 25 min of left ascending coronary (LAD) ischemia and 2 h of reperfusion in a fentanyl-midazolam anesthetized IRI rat model. Plasma lactate dehydrogenase (LDH) activity at 30 min reperfusion was determined as secondary outcome parameter. Following pilot dose-response experiments of each compound (3 dosages, n = 4-6 animals per dosage), potential cardioprotective drugs at the optimal observed dosage were subsequently tested alone or in combination ( n = 6-8 animals per group). The effective treatment was subsequently tested in the presence of a P2Y12 antagonist (cangrelor; n = 6/7) or propofol aesthesia ( n = 6 both groups). Results: Pilot studies suggested potential cardioprotective effects for 50 mg/kg NR ( p = 0.005) and 500 μg/kg melatonin ( p = 0.12), but not for Empa or Fingo. Protection was subsequently tested in a new series of experiments for solvents, NR, Mela and NR+Mela. Results demonstrated that only singular NR was able to reduce IS% (30 ± 14 vs. 60 ± 16%, P = 0.009 vs. control). Mela (63 ± 18%) and NR+Mela (47 ± 15%) were unable to significantly decrease IS%. NR still reduced IS in the presence of cangrelor (51 ± 18 vs. 71 ± 4%, P = 0.016 vs. control), but lost protection in the presence of propofol anesthesia (62 ± 16 vs. 60 ± 14%, P = 0.839 vs. control). LDH activity measurements supported all IS% results. Conclusion: This observational study suggests that NR is a promising cardioprotective agent to target cardiac ischemia-reperfusion injury in clinical conditions employing opioid agonists, benzodiazepines and platelet P2Y12 inhibitors, but not propofol., 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., (Copyright © 2021 Xiao, Phelp, Wang, Bakker, Nederlof, Hollmann and Zuurbier.)- Published
- 2021
- Full Text
- View/download PDF
29. Quantification of Myocardial Creatine and Triglyceride Content in the Human Heart: Precision and Accuracy of in vivo Proton Magnetic Resonance Spectroscopy.
- Author
-
Bakermans AJ, Boekholdt SM, de Vries DK, Reckman YJ, Farag ES, de Heer P, Uthman L, Denis SW, Zuurbier CJ, Houtkooper RH, Koolbergen DR, Kluin J, Planken RN, Lamb HJ, Webb AG, Strijkers GJ, Beard DA, Jeneson JAL, and Nederveen AJ
- Subjects
- Heart diagnostic imaging, Humans, Proton Magnetic Resonance Spectroscopy, Triglycerides, Creatine, Myocardium
- Abstract
Background: Proton magnetic resonance spectroscopy (
1 H-MRS) of the human heart is deemed to be a quantitative method to investigate myocardial metabolite content, but thorough validations of in vivo measurements against invasive techniques are lacking., Purpose: To determine measurement precision and accuracy for quantifications of myocardial total creatine and triglyceride content with localized1 H-MRS., Study Type: Test-retest repeatability and measurement validation study., Subjects: Sixteen volunteers and 22 patients scheduled for open-heart aortic valve replacement or septal myectomy., Field Strength/sequence: Prospectively ECG-triggered respiratory-gated free-breathing single-voxel point-resolved spectroscopy (PRESS) sequence at 3 T., Assessment: Myocardial total creatine and triglyceride content were quantified relative to the total water content by fitting the1 H-MR spectra. Precision was assessed with measurement repeatability. Accuracy was assessed by validating in vivo1 H-MRS measurements against biochemical assays in myocardial tissue from the same subjects., Statistical Tests: Intrasession and intersession repeatability was assessed using Bland-Altman analyses. Agreement between1 H-MRS measurements and biochemical assay was tested with regression analyses., Results: The intersession repeatability coefficient for myocardial total creatine content was 41.8% with a mean value of 0.083% ± 0.020% of the total water signal, and 36.7% for myocardial triglyceride content with a mean value of 0.35% ± 0.13% of the total water signal. Ex vivo myocardial total creatine concentrations in tissue samples correlated with the in vivo myocardial total creatine content measured with1 H-MRS: n = 22, r = 0.44; P < 0.05. Likewise, ex vivo myocardial triglyceride concentrations correlated with the in vivo myocardial triglyceride content: n = 20, r = 0.50; P < 0.05., Data Conclusion: We validated the use of localized1 H-MRS of the human heart at 3 T for quantitative assessments of in vivo myocardial tissue metabolite content by estimating the measurement precision and accuracy., Level of Evidence: 2 TECHNICAL EFFICACY STAGE: 2., (© 2021 The Authors. Journal of Magnetic Resonance Imaging published by Wiley Periodicals LLC. on behalf of International Society for Magnetic Resonance in Medicine.)- Published
- 2021
- Full Text
- View/download PDF
30. The Redox Modulating Sonlicromanol Active Metabolite KH176m and the Antioxidant MPG Protect Against Short-Duration Cardiac Ischemia-Reperfusion Injury.
- Author
-
Xiao Y, Yim K, Zhang H, Bakker D, Nederlof R, Smeitink JAM, Renkema H, Hollmann MW, Weber NC, and Zuurbier CJ
- Subjects
- Aldehydes metabolism, Animals, Antioxidants pharmacology, Disease Models, Animal, Mice, Oxidative Stress drug effects, Time-to-Treatment, Tiopronin pharmacology, Treatment Outcome, Chromans pharmacology, Myocardial Reperfusion Injury drug therapy, Myocardial Reperfusion Injury prevention & control, Oxidation-Reduction drug effects
- Abstract
Purpose: Sonlicromanol is a phase IIB clinical stage compound developed for treatment of mitochondrial diseases. Its active component, KH176m, functions as an antioxidant, directly scavenging reactive oxygen species (ROS), and redox activator, boosting the peroxiredoxin-thioredoxin system. Here, we examined KH176m's potential to protect against acute cardiac ischemia-reperfusion injury (IRI), compare it with the classic antioxidant N-(2-mercaptopropionyl)-glycine (MPG), and determine whether protection depends on duration (severity) of ischemia., Methods: Isolated C56Bl/6N mouse hearts were Langendorff-perfused and subjected to short (20 min) or long (30 min) ischemia, followed by reperfusion. During perfusion, hearts were treated with saline, 10 μM KH176m, or 1 mM MPG. Cardiac function, cell death (necrosis), and mitochondrial damage (cytochrome c (CytC) release) were evaluated. In additional series, the effect of KH176m treatment on the irreversible oxidative stress marker 4-hydroxy-2-nonenal (4-HNE), formed during ischemia only, was determined at 30-min reperfusion., Results: During baseline conditions, both drugs reduced cardiac performance, with opposing effects on vascular resistance (increased with KH176m, decreased with MPG). For short ischemia, KH176m robustly reduced all cell death parameters: LDH release (0.2 ± 0.2 vs 0.8 ± 0.5 U/min/GWW), infarct size (15 ± 8 vs 31 ± 20%), and CytC release (168.0 ± 151.9 vs 790.8 ± 453.6 ng/min/GWW). Protection by KH176m was associated with decreased cardiac 4-HNE. MPG only reduced CytC release. Following long ischemia, IRI was doubled, and KH176m and MPG now only reduced LDH release. The reduced protection against long ischemia was associated with the inability to reduce cardiac 4-HNE., Conclusion: Protection against cardiac IRI by the antioxidant KH176m is critically dependent on duration of ischemia. The data suggest that with longer ischemia, the capacity of KH176m to reduce cardiac oxidative stress is rate-limiting, irreversible ischemic oxidative damage maximally accumulates, and antioxidant protection is strongly diminished.
- Published
- 2021
- Full Text
- View/download PDF
31. Sodium Glucose Co-Transporter 2 Inhibitors Ameliorate Endothelium Barrier Dysfunction Induced by Cyclic Stretch through Inhibition of Reactive Oxygen Species.
- Author
-
Li X, Römer G, Kerindongo RP, Hermanides J, Albrecht M, Hollmann MW, Zuurbier CJ, Preckel B, and Weber NC
- Subjects
- Benzhydryl Compounds pharmacology, Canagliflozin pharmacology, Cell Membrane Permeability drug effects, Endothelial Cells metabolism, Glucosides pharmacology, Guanidines pharmacology, Humans, Inflammation genetics, Inflammation pathology, NADPH Oxidases antagonists & inhibitors, NADPH Oxidases genetics, Oxidative Stress genetics, Pyrazoles pharmacology, Pyridones pharmacology, Reactive Oxygen Species metabolism, Sodium-Glucose Transport Proteins genetics, Sodium-Glucose Transporter 2 Inhibitors pharmacology, Sodium-Hydrogen Exchanger 1 genetics, Stress, Mechanical, Sulfones pharmacology, Endothelial Cells drug effects, Inflammation drug therapy, Oxidative Stress drug effects, Sodium-Glucose Transport Proteins antagonists & inhibitors, Sodium-Hydrogen Exchanger 1 antagonists & inhibitors
- Abstract
SGLT-2i's exert direct anti-inflammatory and anti-oxidative effects on resting endothelial cells. However, endothelial cells are constantly exposed to mechanical forces such as cyclic stretch. Enhanced stretch increases the production of reactive oxygen species (ROS) and thereby impairs endothelial barrier function. We hypothesized that the SGLT-2i's empagliflozin (EMPA), dapagliflozin (DAPA) and canagliflozin (CANA) exert an anti-oxidative effect and alleviate cyclic stretch-induced endothelial permeability in human coronary artery endothelial cells (HCAECs). HCAECs were pre-incubated with one of the SGLT-2i's (1 µM EMPA, 1 µM DAPA and 3 µM CANA) for 2 h, followed by 10% stretch for 24 h. HCAECs exposed to 5% stretch were considered as control. Involvement of ROS was measured using N-acetyl-l-cysteine (NAC). The sodium-hydrogen exchanger 1 (NHE1) and NADPH oxidases (NOXs) were inhibited by cariporide, or GKT136901, respectively. Cell permeability and ROS were investigated by fluorescence intensity imaging. Cell permeability and ROS production were increased by 10% stretch; EMPA, DAPA and CANA decreased this effect significantly. Cariporide and GKT136901 inhibited stretch-induced ROS production but neither of them further reduced ROS production when combined with EMPA. SGLT-2i's improve the barrier dysfunction of HCAECs under enhanced stretch and this effect might be mediated through scavenging of ROS. Anti-oxidative effect of SGLT-2i's might be partially mediated by inhibition of NHE1 and NOXs.
- Published
- 2021
- Full Text
- View/download PDF
32. Influence of cardiometabolic comorbidities on myocardial function, infarction, and cardioprotection: Role of cardiac redox signaling.
- Author
-
Andreadou I, Daiber A, Baxter GF, Brizzi MF, Di Lisa F, Kaludercic N, Lazou A, Varga ZV, Zuurbier CJ, Schulz R, and Ferdinandy P
- Subjects
- Humans, Infarction metabolism, Oxidation-Reduction, Oxidative Stress, Cardiovascular Diseases epidemiology, Cardiovascular Diseases metabolism, Cardiovascular Diseases prevention & control, Myocardium metabolism
- Abstract
The morbidity and mortality from cardiovascular diseases (CVD) remain high. Metabolic diseases such as obesity, hyperlipidemia, diabetes mellitus (DM), non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) as well as hypertension are the most common comorbidities in patients with CVD. These comorbidities result in increased myocardial oxidative stress, mainly from increased activity of nicotinamide adenine dinucleotide phosphate oxidases, uncoupled endothelial nitric oxide synthase, mitochondria as well as downregulation of antioxidant defense systems. Oxidative and nitrosative stress play an important role in ischemia/reperfusion injury and may account for increased susceptibility of the myocardium to infarction and myocardial dysfunction in the presence of the comorbidities. Thus, while early reperfusion represents the most favorable therapeutic strategy to prevent ischemia/reperfusion injury, redox therapeutic strategies may provide additive benefits, especially in patients with heart failure. While oxidative and nitrosative stress are harmful, controlled release of reactive oxygen species is however important for cardioprotective signaling. In this review we summarize the current data on the effect of hypertension and major cardiometabolic comorbidities such as obesity, hyperlipidemia, DM, NAFLD/NASH on cardiac redox homeostasis as well as on ischemia/reperfusion injury and cardioprotection. We also review and discuss the therapeutic interventions that may restore the redox imbalance in the diseased myocardium in the presence of these comorbidities., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
33. Chronic Empagliflozin Treatment Reduces Myocardial Infarct Size in Nondiabetic Mice Through STAT-3-Mediated Protection on Microvascular Endothelial Cells and Reduction of Oxidative Stress.
- Author
-
Nikolaou PE, Efentakis P, Abu Qourah F, Femminò S, Makridakis M, Kanaki Z, Varela A, Tsoumani M, Davos CH, Dimitriou CA, Tasouli A, Dimitriadis G, Kostomitsopoulos N, Zuurbier CJ, Vlahou A, Klinakis A, Brizzi MF, Iliodromitis EK, and Andreadou I
- Subjects
- Administration, Oral, Animals, Benzhydryl Compounds administration & dosage, Cardiotonic Agents administration & dosage, Cell Hypoxia drug effects, Glucosides administration & dosage, Humans, Male, Mice, Mice, Inbred C57BL, Myocardial Infarction metabolism, Oxidation-Reduction, Oxidative Stress drug effects, Benzhydryl Compounds pharmacology, Cardiotonic Agents pharmacology, Endothelial Cells drug effects, Glucosides pharmacology, Microvessels drug effects, Myocardial Infarction drug therapy, STAT3 Transcription Factor metabolism
- Abstract
Aims: Empagliflozin (EMPA) demonstrates cardioprotective effects on diabetic myocardium but its infarct-sparing effects in normoglycemia remain unspecified. We investigated the acute and chronic effect of EMPA on infarct size after ischemia-reperfusion (I/R) injury and the mechanisms of cardioprotection in nondiabetic mice. Results: Chronic oral administration of EMPA (6 weeks) reduced myocardial infarct size after 30 min/2 h I/R (26.5% ± 3.9% vs 45.8% ± 3.3% in the control group, p < 0.01). Body weight, blood pressure, glucose levels, and cardiac function remained unchanged between groups. Acute administration of EMPA 24 or 4 h before I/R did not affect infarct size. Chronic EMPA treatment led to a significant reduction of oxidative stress biomarkers. STAT-3 (signal transducer and activator of transcription 3) was activated by Y(705) phosphorylation at the 10th minute of R, but it remained unchanged at 2 h of R and in the acute administration protocols. Proteomic analysis was employed to investigate signaling intermediates and revealed that chronic EMPA treatment regulates several pathways at reperfusion, including oxidative stress and integrin-related proteins that were further evaluated. Superoxide dismutase and vascular endothelial growth factor were increased throughout reperfusion. EMPA pretreatment (24 h) increased the viability of human microvascular endothelial cells in normoxia and on 3 h hypoxia/1 h reoxygenation and reduced reactive oxygen species production. In EMPA-treated murine hearts, CD31-/VEGFR2-positive endothelial cells and the pSTAT-3(Y705) signal derived from endothelial cells were boosted at early reperfusion. Innovation: Chronic EMPA administration reduces infarct size in healthy mice via the STAT-3 pathway and increases the survival of endothelial cells. Conclusion: Chronic but not acute administration of EMPA reduces infarct size through STAT-3 activation independently of diabetes mellitus.
- Published
- 2021
- Full Text
- View/download PDF
34. Energy substrate metabolism and mitochondrial oxidative stress in cardiac ischemia/reperfusion injury.
- Author
-
Dambrova M, Zuurbier CJ, Borutaite V, Liepinsh E, and Makrecka-Kuka M
- Subjects
- Energy Metabolism, Humans, Ischemia metabolism, Mitochondria, Heart metabolism, Oxidative Stress, Reactive Oxygen Species metabolism, Reperfusion Injury metabolism
- Abstract
The heart is the most metabolically flexible organ with respect to the use of substrates available in different states of energy metabolism. Cardiac mitochondria sense substrate availability and ensure the efficiency of oxidative phosphorylation and heart function. Mitochondria also play a critical role in cardiac ischemia/reperfusion injury, during which they are directly involved in ROS-producing pathophysiological mechanisms. This review explores the mechanisms of ROS production within the energy metabolism pathways and focuses on the impact of different substrates. We describe the main metabolites accumulating during ischemia in the glucose, fatty acid, and Krebs cycle pathways. Hyperglycemia, often present in the acute stress condition of ischemia/reperfusion, increases cytosolic ROS concentrations through the activation of NADPH oxidase 2 and increases mitochondrial ROS through the metabolic overloading and decreased binding of hexokinase II to mitochondria. Fatty acid-linked ROS production is related to the increased fatty acid flux and corresponding accumulation of long-chain acylcarnitines. Succinate that accumulates during anoxia/ischemia is suggested to be the main source of ROS, and the role of itaconate as an inhibitor of succinate dehydrogenase is emerging. We discuss the strategies to modulate and counteract the accumulation of substrates that yield ROS and the therapeutic implications of this concept., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
35. NLRX1 Deletion Increases Ischemia-Reperfusion Damage and Activates Glucose Metabolism in Mouse Heart.
- Author
-
Zhang H, Xiao Y, Nederlof R, Bakker D, Zhang P, Girardin SE, Hollmann MW, Weber NC, Houten SM, van Weeghel M, Kibbey RG, and Zuurbier CJ
- Subjects
- Animals, Biomarkers, Cytokines metabolism, Disease Models, Animal, Mice, Mice, Knockout, Myocardial Reperfusion Injury pathology, Oxidation-Reduction, Oxidative Stress, Proto-Oncogene Proteins c-akt metabolism, Carbohydrate Metabolism, Gene Deletion, Glucose metabolism, Mitochondrial Proteins genetics, Myocardial Reperfusion Injury etiology, Myocardial Reperfusion Injury metabolism
- Abstract
Background: NOD-like receptors (NLR) are intracellular sensors of the innate immune system, with the NLRP3 being a pro-inflammatory member that modulates cardiac ischemia-reperfusion injury (IRI) and metabolism. No information is available on a possible role of anti-inflammatory NLRs on IRI and metabolism in the intact heart. Here we hypothesize that the constitutively expressed, anti-inflammatory mitochondrial NLRX1, affects IRI and metabolism of the isolated mouse heart., Methods: Isolated C57Bl/6J and NLRX1 knock-out (KO) mouse hearts were perfused with a physiological mixture of the essential substrates (lactate, glucose, pyruvate, fatty acid, glutamine) and insulin. For the IRI studies, hearts were subjected to either mild (20 min) or severe (35 min) ischemia and IRI was determined at 60 min reperfusion. Inflammatory mediators (IL-6, TNFα) and survival pathways (mito-HKII, p-Akt, p-AMPK, p-STAT3) were analyzed at 5 min of reperfusion. For the metabolism studies, hearts were perfused for 35 min with either 5.5 mM
13 C-glucose or 0.4 mM13 C-palmitate under normoxic conditions, followed by LC-MS analysis and integrated, stepwise, mass-isotopomeric flux analysis (MIMOSA)., Results: NLRX1 KO significantly increased IRI (infarct size from 63% to 73%, end-diastolic pressure from 59 mmHg to 75 mmHg, and rate-pressure-product recovery from 15% to 6%), following severe, but not mild, ischemia. The increased IRI in NLRX1 KO hearts was associated with depressed Akt signaling at early reperfusion; other survival pathways or inflammatory parameters were not affected. Metabolically, NLRX1 KO hearts displayed increased lactate production and glucose oxidation relative to fatty acid oxidation, associated with increased pyruvate dehydrogenase flux and 10% higher cardiac oxygen consumption., Conclusion: Deletion of the mitochondrially-located NOD-like sensor NLRX1 exacerbates severe cardiac IR injury, possibly through impaired Akt signaling, and increases cardiac glucose metabolism., 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., (Copyright © 2020 Zhang, Xiao, Nederlof, Bakker, Zhang, Girardin, Hollmann, Weber, Houten, van Weeghel, Kibbey and Zuurbier.)- Published
- 2020
- Full Text
- View/download PDF
36. Empagliflozin Decreases Lactate Generation in an NHE-1 Dependent Fashion and Increases α-Ketoglutarate Synthesis From Palmitate in Type II Diabetic Mouse Hearts.
- Author
-
Zhang H, Uthman L, Bakker D, Sari S, Chen S, Hollmann MW, Coronel R, Weber NC, Houten SM, van Weeghel M, and Zuurbier CJ
- Abstract
Aims/hypothesis: Changes in cardiac metabolism and ion homeostasis precede and drive cardiac remodeling and heart failure development. We previously demonstrated that sodium/glucose cotransporter 2 inhibitors (SGLT2i's) have direct cardiac effects on ion homeostasis, possibly through inhibition of the cardiac sodium/hydrogen exchanger (NHE-1). Here, we hypothesize that Empagliflozin (EMPA) also possesses direct and acute cardiac effects on glucose and fatty acid metabolism of isolated type II diabetes mellitus ( db/db ) mouse hearts. In addition, we explore whether direct effects on glucose metabolism are nullified in the presence of an NHE-1 inhibitor. Methods: Langendorff-perfused type II diabetic db/db mouse hearts were examined in three different series: 1 :
13 C glucose perfusions ( n = 32); 2 :13 C palmitate perfusions ( n = 13); and 3 :13 C glucose + 10 μM Cariporide (specific NHE-1 inhibitor) perfusions ( n = 17). Within each series, EMPA treated hearts (1 μM EMPA) were compared with vehicle-perfused hearts (0.02% DMSO). Afterwards, hearts were snap frozen and lysed for stable isotope analysis and metabolomics using LC-MS techniques. Hearts from series 1 were also analyzed for phosphorylation status of AKT, STAT3, AMPK, ERK, and eNOS ( n = 8 per group). Results: Cardiac mechanical performance, oxygen consumption and protein phosphorylation were not altered by 35 min EMPA treatment. EMPA was without an overall acute and direct effect on glucose or fatty acid metabolism. However, EMPA did specifically decrease cardiac lactate labeling in the13 C glucose perfusions (13 C labeling of lactate: 58 ± 2% vs. 50 ± 3%, for vehicle and EMPA, respectively; P = 0.02), without changes in other glucose metabolic pathways. In contrast, EMPA increased cardiac labeling in α-ketoglutarate derived from13 C palmitate perfusions (13 C labeling of α-KG: 79 ± 1% vs. 86 ± 1% for vehicle and EMPA, respectively; P = 0.01). Inhibition of the NHE by Cariporide abolished EMPA effects on lactate labeling from13 C glucose. Conclusions: The present study shows for the first time that the SGLT2 inhibitor Empagliflozin has acute specific metabolic effects in isolated diabetic hearts, i.e., decreased lactate generation from labeled glucose and increased α-ketoglutarate synthesis from labeled palmitate. The decreased lactate generation by EMPA seems to be mediated through NHE-1 inhibition., 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., (Copyright © 2020 Zhang, Uthman, Bakker, Sari, Chen, Hollmann, Coronel, Weber, Houten, van Weeghel and Zuurbier.)- Published
- 2020
- Full Text
- View/download PDF
37. Effect of hyperglycaemia and diabetes on acute myocardial ischaemia-reperfusion injury and cardioprotection by ischaemic conditioning protocols.
- Author
-
Penna C, Andreadou I, Aragno M, Beauloye C, Bertrand L, Lazou A, Falcão-Pires I, Bell R, Zuurbier CJ, Pagliaro P, and Hausenloy DJ
- Subjects
- Animals, Humans, Diabetes Mellitus, Hyperglycemia complications, Hyperglycemia drug therapy, Ischemic Preconditioning, Myocardial, Myocardial Infarction, Myocardial Reperfusion Injury prevention & control
- Abstract
Diabetic patients are at increased risk of developing coronary artery disease and experience worse clinical outcomes following acute myocardial infarction. Novel therapeutic strategies are required to protect the myocardium against the effects of acute ischaemia-reperfusion injury (IRI). These include one or more brief cycles of non-lethal ischaemia and reperfusion prior to the ischaemic event (ischaemic preconditioning [IPC]) or at the onset of reperfusion (ischaemic postconditioning [IPost]) either to the heart or to extracardiac organs (remote ischaemic conditioning [RIC]). Studies suggest that the diabetic heart is resistant to cardioprotective strategies, although clinical evidence is lacking. We overview the available animal models of diabetes, investigating acute myocardial IRI and cardioprotection, experiments investigating the effects of hyperglycaemia on susceptibility to acute myocardial IRI, the response of the diabetic heart to cardioprotective strategies e.g. IPC, IPost and RIC. Finally we highlight the effects of anti-hyperglycaemic agents on susceptibility to acute myocardial IRI and cardioprotection. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.23/issuetoc., (© 2020 The British Pharmacological Society.)
- Published
- 2020
- Full Text
- View/download PDF
38. Targeting metabolic pathways to treat cardiovascular diseases.
- Author
-
Glatz JFC, Zuurbier CJ, and Larsen TS
- Subjects
- Cardiovascular Diseases metabolism, Congresses as Topic, Energy Metabolism drug effects, Heart drug effects, Humans, Myocardial Contraction drug effects, Myocardium metabolism, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Societies, Medical, Sodium-Glucose Transporter 2 Inhibitors pharmacology, Sodium-Glucose Transporter 2 Inhibitors therapeutic use, Cardiovascular Diseases drug therapy, Metabolic Networks and Pathways drug effects
- Published
- 2020
- Full Text
- View/download PDF
39. Does acute treatment of dapagliflozin reduce cardiac infarct size through direct cardiac effects or reductions in blood glucose levels?
- Author
-
Zuurbier CJ
- Subjects
- Animals, Benzhydryl Compounds adverse effects, Glucosides adverse effects, Rats, Blood Glucose, Reperfusion Injury
- Published
- 2020
- Full Text
- View/download PDF
40. SGLT2 inhibitors reduce infarct size in reperfused ischemic heart and improve cardiac function during ischemic episodes in preclinical models.
- Author
-
Andreadou I, Bell RM, Bøtker HE, and Zuurbier CJ
- Subjects
- Cardiovascular Diseases drug therapy, Cardiovascular Diseases genetics, Cardiovascular Diseases pathology, Diabetes Mellitus, Type 2 complications, Diabetes Mellitus, Type 2 drug therapy, Diabetes Mellitus, Type 2 genetics, Diabetes Mellitus, Type 2 pathology, Heart Failure etiology, Heart Failure genetics, Humans, Hypoglycemic Agents pharmacology, Hypoglycemic Agents therapeutic use, Infarction etiology, Infarction genetics, Risk Factors, Sodium-Glucose Transporter 2 Inhibitors therapeutic use, Heart Failure drug therapy, Infarction drug therapy, Sodium-Glucose Transporter 2 genetics, Sodium-Glucose Transporter 2 Inhibitors pharmacology
- Abstract
The sodium-glucose cotransporter 2 (SGLT2) inhibitors are a new class of effective drugs managing patients, who suffer from type 2 diabetes (T2D): Landmark clinical trials including EMPA-REG, CANVAS and Declare-TIMI have demonstrated that SGLT2 inhibitors reduce cardiovascular mortality and re-hospitalization for heart failure (HF) in patients with T2D. It is well established that there is a strong independent relationship among infarct size measured within 1 month after reperfusion and all-cause death and hospitalization for HF: The fact that cardiovascular mortality was significantly reduced with the SGLT2 inhibitors, fuels the assumption that this class of therapies may attenuate myocardial infarct size. Experimental evidence demonstrates that SGLT2 inhibitors exert cardioprotective effects in animal models of acute myocardial infarction through improved function during the ischemic episode, reduction of infarct size and a subsequent attenuation of heart failure development. The aim of the present review is to outline the current state of preclinical research in terms of myocardial ischemia/reperfusion injury (I/R) and infarct size for clinically available SGLT2 inhibitors and summarize some of the proposed mechanisms of action (lowering intracellular Na
+ and Ca2+ , NHE inhibition, STAT3 and AMPK activation, CamKII inhibition, reduced inflammation and oxidative stress) that may contribute to the unexpected beneficial cardiovascular effects of this class of compounds., Competing Interests: Declaration of competing interest The authors have no declarations of interests to disclose., (Copyright © 2020 Elsevier B.V. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
41. Cardiac metabolism as a driver and therapeutic target of myocardial infarction.
- Author
-
Zuurbier CJ, Bertrand L, Beauloye CR, Andreadou I, Ruiz-Meana M, Jespersen NR, Kula-Alwar D, Prag HA, Eric Botker H, Dambrova M, Montessuit C, Kaambre T, Liepinsh E, Brookes PS, and Krieg T
- Subjects
- Animals, Energy Metabolism, Humans, Mitochondria, Heart metabolism, Myocardial Reperfusion Injury metabolism, Myocardium pathology, Molecular Targeted Therapy, Myocardial Infarction metabolism, Myocardium metabolism
- Abstract
Reducing infarct size during a cardiac ischaemic-reperfusion episode is still of paramount importance, because the extension of myocardial necrosis is an important risk factor for developing heart failure. Cardiac ischaemia-reperfusion injury (IRI) is in principle a metabolic pathology as it is caused by abruptly halted metabolism during the ischaemic episode and exacerbated by sudden restart of specific metabolic pathways at reperfusion. It should therefore not come as a surprise that therapy directed at metabolic pathways can modulate IRI. Here, we summarize the current knowledge of important metabolic pathways as therapeutic targets to combat cardiac IRI. Activating metabolic pathways such as glycolysis (eg AMPK activators), glucose oxidation (activating pyruvate dehydrogenase complex), ketone oxidation (increasing ketone plasma levels), hexosamine biosynthesis pathway (O-GlcNAcylation; administration of glucosamine/glutamine) and deacetylation (activating sirtuins 1 or 3; administration of NAD
+ -boosting compounds) all seem to hold promise to reduce acute IRI. In contrast, some metabolic pathways may offer protection through diminished activity. These pathways comprise the malate-aspartate shuttle (in need of novel specific reversible inhibitors), mitochondrial oxygen consumption, fatty acid oxidation (CD36 inhibitors, malonyl-CoA decarboxylase inhibitors) and mitochondrial succinate metabolism (malonate). Additionally, protecting the cristae structure of the mitochondria during IR, by maintaining the association of hexokinase II or creatine kinase with mitochondria, or inhibiting destabilization of FO F1 -ATPase dimers, prevents mitochondrial damage and thereby reduces cardiac IRI. Currently, the most promising and druggable metabolic therapy against cardiac IRI seems to be the singular or combined targeting of glycolysis, O-GlcNAcylation and metabolism of ketones, fatty acids and succinate., (© 2020 The Authors. Journal of Cellular and Molecular Medicine published by Foundation for Cellular and Molecular Medicine and John Wiley & Sons Ltd.)- Published
- 2020
- Full Text
- View/download PDF
42. Volume incompliance and transfusion are essential for transfusion-associated circulatory overload: a novel animal model.
- Author
-
Klanderman RB, Bosboom JJ, Maas AAW, Roelofs JJTH, de Korte D, van Bruggen R, van Buul JD, Zuurbier CJ, Veelo DP, Hollmann MW, Vroom MB, Juffermans NP, Geerts BF, and Vlaar APJ
- Subjects
- Anemia therapy, Animals, Disease Models, Animal, Heart Rate physiology, Hypertension physiopathology, Male, Myocardial Infarction therapy, Rats, Rats, Inbred Lew, Risk Factors, Transfusion Reaction physiopathology, Blood Transfusion methods, Transfusion Reaction etiology
- Abstract
Background: Transfusion-associated circulatory overload (TACO) is the predominant complication of transfusion resulting in death. The pathophysiology is poorly understood, but inability to manage volume is associated with TACO, and observational data suggest it is different from simple cardiac overload due to fluids. We developed a two-hit TACO animal model to assess the role of volume incompliance ("first-hit") and studied whether volume overload ("second-hit") by red blood cell (RBC) transfusion is different compared to fluids (Ringer's lactate [RL])., Materials and Methods: Male adult Lewis rats were stratified into a control group (no intervention) or a first hit: either myocardial infarction (MI) or acute kidney injury (AKI). Animals were randomized to a second hit of either RBC transfusion or an equal volume of RL. A clinically relevant difference was defined as an increase in left ventricular end-diastolic pressure (ΔLVEDP) of +4.0 mm Hg between the RBC and RL groups., Results: In control animals (without first hit) LVEDP was not different between infusion groups (Δ + 1.6 mm Hg). LVEDP increased significantly more after RBCs compared to RL in animals with MI (Δ7.4 mm Hg) and AKI (Δ + 5.4 mm Hg), respectively. Volume-incompliant rats matched clinical TACO criteria in 92% of transfused versus 25% of RL-infused animals, with a greater increase in heart rate and significantly higher blood pressure., Conclusion: To our knowledge, this is the first animal model for TACO, showing that a combination of volume incompliance and transfusion is essential for development of circulatory overload. This model allows for further testing of mechanistic factors as well as therapeutic approaches., (© 2019 The Authors. Transfusion published by Wiley Periodicals, Inc. on behalf of AABB.)
- Published
- 2019
- Full Text
- View/download PDF
43. NLRP3 Inflammasome in Cardioprotective Signaling.
- Author
-
Zuurbier CJ
- Subjects
- Animals, Anti-Inflammatory Agents therapeutic use, CARD Signaling Adaptor Proteins metabolism, Caspase 1 metabolism, Heart Failure immunology, Heart Failure pathology, Heart Failure prevention & control, Humans, Inflammasomes antagonists & inhibitors, Inflammasomes immunology, Inflammation Mediators antagonists & inhibitors, Inflammation Mediators immunology, Myocardial Infarction diagnostic imaging, Myocardial Infarction immunology, Myocardial Infarction pathology, Myocardial Reperfusion Injury drug therapy, Myocardial Reperfusion Injury immunology, Myocardial Reperfusion Injury pathology, Myocardium immunology, Myocardium pathology, NLR Family, Pyrin Domain-Containing 3 Protein antagonists & inhibitors, NLR Family, Pyrin Domain-Containing 3 Protein immunology, Signal Transduction, Heart Failure metabolism, Inflammasomes metabolism, Inflammation Mediators metabolism, Myocardial Infarction metabolism, Myocardial Reperfusion Injury metabolism, Myocardium metabolism, NLR Family, Pyrin Domain-Containing 3 Protein metabolism
- Abstract
The NLRP3 inflammasome may contribute to infarct development during acute cardiac ischemia-reperfusion (IR). Because infarct size strongly correlates with the degree of heart failure in the long term, therapies that reduce reperfusion injury are still needed as first primary care against heart failure development. Inhibition of the NLRP3 inflammasome is currently viewed as such a potential therapy. However, previous research studies directed at inhibition of various inflammatory pathways in acute cardiac IR injury were often disappointing. This is because inflammation is a double-edged sword, detrimental when hyperactive, but beneficial at lower activity, with activity critically dependent on time of reperfusion and cellular location. Moreover, several inflammatory mediators can also mediate cardioprotective signaling. It is reasonable that this also applies to the NLRP3 inflammasome, although current literature has mainly focused on its detrimental effects in the context of acute cardiac IR. Therefore, in this review, we focus on beneficial, cardioprotective properties of the NLRP3 inflammasome and its components NLRP3, ASC, and caspase-1. The results show that (1) NLRP3 deficiency prevents cardioprotection in isolated heart by ischemic preconditioning and in vivo heart by TLR2 activation, associated with impaired STAT3 or Akt signaling, respectively; (2) ASC deficiency also prevents in vivo TLR2-mediated protection; and (3) caspase-1 inhibition results in decreased infarction but impaired protection through the Akt pathway during mild ischemic insults. In conclusion, the NLRP3 inflammasome is not only detrimental, it can also be involved in cardioprotective signaling, thus fueling the future challenge to acquire a full understanding of NLRP3 inflammasome role in cardiac IR before embarking on clinical trials using NLRP3 inhibitors.
- Published
- 2019
- Full Text
- View/download PDF
44. Ketamine-(Dex)Medetomidine, Hyperglycemia, Glycocalyx, and Vascular Permeability.
- Author
-
Zuurbier CJ
- Subjects
- Animals, Capillary Permeability, Glycocalyx, Medetomidine, Permeability, Rats, Hyperglycemia, Ketamine, Shock, Hemorrhagic
- Published
- 2019
- Full Text
- View/download PDF
45. Delayed ischaemic contracture onset by empagliflozin associates with NHE1 inhibition and is dependent on insulin in isolated mouse hearts.
- Author
-
Uthman L, Nederlof R, Eerbeek O, Baartscheer A, Schumacher C, Buchholtz N, Hollmann MW, Coronel R, Weber NC, and Zuurbier CJ
- Subjects
- Animals, Disease Models, Animal, Isolated Heart Preparation, Male, Mice, Inbred C57BL, Myocardial Infarction metabolism, Myocardial Infarction pathology, Myocardial Infarction physiopathology, Myocardial Reperfusion Injury metabolism, Myocardial Reperfusion Injury pathology, Myocardial Reperfusion Injury physiopathology, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Signal Transduction, Sodium-Hydrogen Exchanger 1 metabolism, Time Factors, Benzhydryl Compounds pharmacology, Glucosides pharmacology, Guanidines pharmacology, Insulin pharmacology, Myocardial Contraction drug effects, Myocardial Infarction prevention & control, Myocardial Reperfusion Injury prevention & control, Myocytes, Cardiac drug effects, Sodium-Glucose Transporter 2 Inhibitors pharmacology, Sodium-Hydrogen Exchanger 1 antagonists & inhibitors, Sulfones pharmacology
- Abstract
Aims: Sodium glucose cotransporter 2 (SGLT2) inhibitors have sodium-hydrogen exchanger (NHE) inhibition properties in isolated cardiomyocytes, but it is unknown whether these properties extend to the intact heart during ischaemia-reperfusion (IR) conditions. NHE inhibitors as Cariporide delay time to onset of contracture (TOC) during ischaemia and reduce IR injury. We hypothesized that, in the ex vivo heart, Empagliflozin (Empa) mimics Cariporide during IR by delaying TOC and reducing IR injury. To facilitate translation to in vivo conditions with insulin present, effects were examined in the absence and presence of insulin., Methods and Results: Isolated C57Bl/6NCrl mouse hearts were subjected to 25 min I and 120 min R without and with 50 mU/L insulin. Without insulin, Empa and Cari delayed TOC by 100 and 129 s, respectively, yet only Cariporide reduced IR injury [infarct size (mean ± SEM in %) from 51 ± 6 to 34 ± 5]. Empa did not delay TOC in the presence of the NHE1 inhibitor Eniporide. Insulin perfusion increased tissue glycogen content at baseline (from 2 ± 2 µmol to 42 ± 1 µmol glycosyl units/g heart dry weight), amplified G6P and lactate accumulation at end-ischaemia, thereby decreased mtHKII and exacerbated IR injury. Under these conditions, Empa (1 µM) and Cariporide (10 µM) were without effect on TOC and IR injury. Empa and Cariporide both inhibited NHE activity, in isolated cardiomyocytes, independent of insulin., Conclusions: In the absence of insulin, Empa and Cariporide strongly delayed the time to onset of contracture during ischaemia. In the presence of insulin, both Empa and Cari were without effect on IR, possibly because of severe ischaemic acidification. Insulin exacerbates IR injury through increased glycogen depletion during ischaemia and consequently mtHKII dissociation. The data suggest that also in the ex vivo intact heart Empa exerts direct cardiac effects by inhibiting NHE during ischaemia, but not during reperfusion., (Published on behalf of the European Society of Cardiology. All rights reserved. © The Author(s) 2019. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
46. Innate immunity as a target for acute cardioprotection.
- Author
-
Zuurbier CJ, Abbate A, Cabrera-Fuentes HA, Cohen MV, Collino M, De Kleijn DPV, Downey JM, Pagliaro P, Preissner KT, Takahashi M, and Davidson SM
- Subjects
- Animals, Anti-Inflammatory Agents adverse effects, Cardiovascular Agents adverse effects, Caspase 1 immunology, Caspase 1 metabolism, Caspase Inhibitors therapeutic use, Heart Failure immunology, Heart Failure metabolism, Heart Failure pathology, Humans, Inflammasomes immunology, Inflammasomes metabolism, Molecular Targeted Therapy, Myocardial Reperfusion Injury immunology, Myocardial Reperfusion Injury metabolism, Myocardial Reperfusion Injury pathology, Myocardium metabolism, Myocardium pathology, NLR Family, Pyrin Domain-Containing 3 Protein antagonists & inhibitors, NLR Family, Pyrin Domain-Containing 3 Protein immunology, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Receptors, Immunologic antagonists & inhibitors, Receptors, Immunologic immunology, Receptors, Immunologic metabolism, ST Elevation Myocardial Infarction immunology, ST Elevation Myocardial Infarction metabolism, ST Elevation Myocardial Infarction pathology, Signal Transduction, Anti-Inflammatory Agents therapeutic use, Cardiovascular Agents therapeutic use, Heart Failure prevention & control, Immunity, Innate, Inflammasomes drug effects, Myocardial Reperfusion Injury prevention & control, Myocardium immunology, ST Elevation Myocardial Infarction therapy
- Abstract
Acute obstruction of a coronary artery causes myocardial ischaemia and if prolonged, may result in an ST-segment elevation myocardial infarction (STEMI). First-line treatment involves rapid reperfusion. However, a highly dynamic and co-ordinated inflammatory response is rapidly mounted to repair and remove the injured cells which, paradoxically, can further exacerbate myocardial injury. Furthermore, although cardiac remodelling may initially preserve some function to the heart, it can lead over time to adverse remodelling and eventually heart failure. Since the size of the infarct corresponds to the subsequent risk of developing heart failure, it is important to find ways to limit initial infarct development. In this review, we focus on the role of the innate immune system in the acute response to ischaemia-reperfusion (IR) and specifically its contribution to cell death and myocardial infarction. Numerous danger-associated molecular patterns are released from dying cells in the myocardium, which can stimulate pattern recognition receptors including toll like receptors and NOD-like receptors (NLRs) in resident cardiac and immune cells. Activation of the NLRP3 inflammasome, caspase 1, and pyroptosis may ensue, particularly when the myocardium has been previously aggravated by the presence of comorbidities. Evidence will be discussed that suggests agents targeting innate immunity may be a promising means of protecting the hearts of STEMI patients against acute IR injury. However, the dosing and timing of such agents should be carefully determined because innate immunity pathways may also be involved in cardioprotection. This article is part of a Cardiovascular Research Spotlight Issue entitled 'Cardioprotection Beyond the Cardiomyocyte', and emerged as part of the discussions of the European Union (EU)-CARDIOPROTECTION Cooperation in Science and Technology (COST) Action, CA16225., (Published on behalf of the European Society of Cardiology. All rights reserved. © The Author(s) 2018. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
47. Editorial: Diabetes and Heart Failure: Pathogenesis and Novel Therapeutic Approaches.
- Author
-
de Lucia C, Sardu C, Metzinger L, and Zuurbier CJ
- Published
- 2019
- Full Text
- View/download PDF
48. Empagliflozin and Dapagliflozin Reduce ROS Generation and Restore NO Bioavailability in Tumor Necrosis Factor α-Stimulated Human Coronary Arterial Endothelial Cells.
- Author
-
Uthman L, Homayr A, Juni RP, Spin EL, Kerindongo R, Boomsma M, Hollmann MW, Preckel B, Koolwijk P, van Hinsbergh VWM, Zuurbier CJ, Albrecht M, and Weber NC
- Subjects
- Coronary Vessels cytology, Endothelial Cells cytology, Endothelial Cells drug effects, Endothelial Cells metabolism, Human Umbilical Vein Endothelial Cells, Humans, Intercellular Adhesion Molecule-1 metabolism, Nitric Oxide Synthase Type III metabolism, Permeability drug effects, Signal Transduction drug effects, Sodium-Glucose Transporter 2 genetics, Sodium-Glucose Transporter 2 metabolism, Vascular Cell Adhesion Molecule-1, Benzhydryl Compounds pharmacology, Down-Regulation drug effects, Glucosides pharmacology, Nitric Oxide metabolism, Reactive Oxygen Species metabolism, Tumor Necrosis Factor-alpha pharmacology
- Abstract
Background/aims: Heart failure is characterized by chronic low-grade vascular inflammation, which in itself can lead to endothelial dysfunction. Clinical trials showed reductions in heart failure-related hospitalizations of type 2 diabetic patients using sodium glucose co-transporter 2 inhibitors (SGLT2i's). Whether and how SGLT2i's directly affect the endothelium under inflammatory conditions is not completely understood. The aim of the study was to investigate whether the SGLT2i Empagliflozin (EMPA) and Dapagliflozin (DAPA) reduce tumor necrosis factor α (TNFα) induced endothelial inflammation in vitro., Methods: Human coronary arterial endothelial cells (HCAECs) and human umbilical vein endothelial cells (HUVECs) were (pre-)incubated with 1 µM EMPA or DAPA and subsequently exposed to 10 ng/ml TNFα. ROS and NO were measured using live cell imaging. Target proteins were either determined by infrared western blotting or fluorescence activated cell sorting (FACS). The connection between Cav-1 and eNOS was determined by co-immunoprecipitation., Results: Nitric oxide (NO) bioavailability was reduced by TNFα and both EMPA and DAPA restored NO levels in TNFα-stimulated HCAECs. Intracellular ROS was increased by TNFα, and this increase was completely abolished by EMPA and DAPA in HCAECs by means of live cell imaging. eNOS signaling was significantly disturbed after 24 h when cells were exposed to TNFα for 24h, yet the presence of both SGLT2is did not prevent this disruption. TNFα-induced enhanced permeability at t=24h was unaffected in HUVECs by EMPA. Similarly, adhesion molecule expression (VCAM-1 and ICAM-1) was elevated after 4h TNFα (1.5-5.5 fold increase of VCAM-1 and 4-12 fold increase of ICAM-1) but were unaffected by EMPA and DAPA in both cell types. Although we detected expression of SGLT2 protein levels, the fact that we could not silence this expression by means of siRNA and the mRNA levels of SGLT2 were not detectable in HCAECs, suggests aspecificity or our SGLT2 antibody and absence of SGLT2 in our cells., Conclusion: These data suggest that EMPA and DAPA rather restore NO bioavailability by inhibiting ROS generation than by affecting eNOS expression or signaling, barrier function and adhesion molecules expression in TNFα-induced endothelial cells. Furthermore, the observed effects cannot be ascribed to the inhibition of SGLT2 in endothelial cells., Competing Interests: The authors have no conflicts of interest to declare., (© Copyright by the Author(s). Published by Cell Physiol Biochem Press.)
- Published
- 2019
- Full Text
- View/download PDF
49. Washing or filtering of blood products does not improve outcome in a rat model of trauma and multiple transfusion.
- Author
-
Wirtz MR, Jurgens J, Zuurbier CJ, Roelofs JJTH, Spinella PC, Muszynski JA, Carel Goslings J, and Juffermans NP
- Subjects
- Animals, Blood Component Transfusion methods, Disease Models, Animal, Erythrocyte Transfusion methods, Extracellular Vesicles, Male, Platelet Transfusion methods, Rats, Rats, Sprague-Dawley, Resuscitation methods, Blood Transfusion methods, Wounds and Injuries therapy
- Abstract
Background: Transfusion is associated with organ failure and nosocomial infection in trauma patients, which may be mediated by soluble bioactive substances in blood products, including extracellular vesicles (EVs). We hypothesize that removing EVs, by washing or filtering of blood products, reduces organ failure and improves host immune response., Materials and Methods: Blood products were prepared from syngeneic rat blood. EVs were removed from RBCs and platelets by washing. Plasma was filtered through a 0.22-μm filter. Rats were traumatized by crush injury to the intestines and liver, and a femur was fractured. Rats were hemorrhaged until a mean arterial pressure of 40 mm Hg and randomized to receive resuscitation with standard or washed/filtered blood products, in a 1:1:1 ratio. Sham controls were not resuscitated. Ex vivo whole blood stimulation tests were performed and histopathology was done., Results: Washing of blood products improved quality metrics compared to standard products. Also, EV levels reduced by 12% to 77%. The coagulation status, as assessed by thromboelastometry, was deranged in both groups and normalized during transfusion, without significant differences. Use of washed/filtered products did not reduce organ failure, as assessed by histopathologic score and biochemical measurements. Immune response ex vivo was decreased following transfusion compared to sham but did not differ between transfusion groups., Conclusion: Filtering or washing of blood products improved biochemical properties and reduced EV counts, while maintaining coagulation abilities. However, in this trauma and transfusion model, the use of optimized blood components did not attenuate organ injury or immune suppression., (© 2018 The Authors. Transfusion published by Wiley Periodicals, Inc. on behalf of AABB.)
- Published
- 2019
- Full Text
- View/download PDF
50. Divergent Effects of Hypertonic Fluid Resuscitation on Renal Pathophysiological and Structural Parameters in Rat Model of Lower Body Ischemia/Reperfusion-Induced Sterile Inflammation.
- Author
-
Ergin B, Zuurbier CJ, Kapucu A, and Ince C
- Subjects
- Acute Kidney Injury therapy, Animals, Fluid Therapy methods, Hemodynamics drug effects, Hyaluronic Acid metabolism, Interleukin-6 metabolism, Kidney drug effects, Kidney metabolism, Kidney Function Tests, Male, Microcirculation drug effects, Oxygen Consumption physiology, Rats, Rats, Wistar, Resuscitation methods, Saline Solution, Hypertonic therapeutic use, Tumor Necrosis Factor-alpha metabolism, Inflammation etiology, Inflammation therapy, Ischemia complications, Reperfusion Injury complications
- Abstract
The pathogenesis of acute kidney injury (AKI) is characterized by the deterioration of tissue perfusion and oxygenation and enhanced inflammation. The purpose of this study was to investigate whether or not the hemodynamic and inflammatory effects of hypertonic saline (HS) protect the kidney by promoting renal microcirculatory oxygenation and possible deleterious effects of HS due to its high sodium content on renal functional and structural injury following ischemia/reperfusion. Mechanically ventilated and anesthetized rats were randomly divided into four groups (n = 6 per group): a sham-operated control group; a group subjected to renal ischemia for 45 min by supra-aortic occlusion followed by 2 h of reperfusion (I/R); and I/R group treated with a continuous i.v. infusion (5 mL/kg/h) of either % 0.9 NaCl (IR+NS) or %10 NaCl (I/R+HS) after releasing the clamp. Systemic and renal hemodynamic, renal cortical (CμPO2), and medullar microcirculatory pO2 (MμPO2) are measured by the oxygen-dependent quenching of the phosphorescence lifetime technique. Renal functional, inflammatory, and tissues damage parameters were also assessed. HS, but not NS, treatment restored I/R-induced reduced mean arterial pressure, CμPO2, renal oxygen deliver (DO2ren), and consumption (VO2ren). HS caused a decrease in tubular sodium reabsorption (TNa) that correlated with an elevation of fractional sodium excretion (EFNa) and urine output. HS had an anti-inflammatory effect by reducing the levels TNF-α, IL-6, and hyaluronic acid in the renal tissue samples as compared with the I/R and I/R+NS groups (P < 0.05). HS treatment was also associated with mild acidosis and an increased renal tubular damage. Despite HS resuscitation improving the systemic hemodynamics, microcirculatory oxygenation, and renal oxygen consumption as well as inflammation, it should be limited or strictly controlled for long-term use because of provoking widespread renal structural damage.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.