66 results on '"Dewenter M"'
Search Results
2. OP0116 THE AP-1 TRANSCRIPTION FACTOR FOSL-2 DRIVES CARDIAC FIBROSIS AND ARRHYTHMIAS UNDER IMMUNOFIBROTIC CONDITIONS IN SYSTEMIC SCLEROSIS
- Author
-
Stellato, M., primary, Dewenter, M., additional, Rudnik, M., additional, Hukara, A., additional, Cagla, O., additional, Renoux, F., additional, Pachera, E., additional, Gantenbein, F., additional, Seebeck, P., additional, Uhtjaerv, S., additional, Osto, E., additional, Razansky, D., additional, Klingel, K., additional, Henes, J., additional, Distler, O., additional, Błyszczuk, P., additional, and Kania, G., additional
- Published
- 2023
- Full Text
- View/download PDF
3. Vareniclin – Erhöhtes kardiovaskuläres Risiko bei der Raucherentwöhnung?
- Author
-
Dewenter, M. and El-Armouche, A.
- Published
- 2012
- Full Text
- View/download PDF
4. Genetic findings in miscarriages and their relation to the number of previous miscarriages
- Author
-
Gomez, R., primary, Hafezi, N., additional, Amrani, M., additional, Schweiger, S., additional, Dewenter, M. K., additional, Thomas, P., additional, Lieb, C., additional, Hasenburg, A., additional, and Skala, C., additional
- Published
- 2020
- Full Text
- View/download PDF
5. Aquaponics using Asian leafy vegetables – potential and challenge
- Author
-
Böhme, M.H., primary, Dewenter, M., additional, and Gohlke, A., additional
- Published
- 2020
- Full Text
- View/download PDF
6. S1631 UNEXPECTED DIFFERENCES IN CARDIAC IRON METABOLISM AND SURVIVAL IN HAMP-KO AND FPN(C326S) MICE
- Author
-
Altamura, S., primary, Müdder, K., additional, Dewenter, M., additional, Garbowski, M., additional, Galy, B., additional, Vaulont, S., additional, Backs, J., additional, Gröne, H.-J., additional, Hentze, M., additional, and Muckenthaler, M., additional
- Published
- 2019
- Full Text
- View/download PDF
7. Calcium/Calmodulin-Dependent Protein Kinase II Activity Persists During Chronic -Adrenoceptor Blockade in Experimental and Human Heart Failure
- Author
-
Dewenter, M, Neef, S, Vettel, C, Laemmle, S, Beushausen, C, Zelarayan, LC, Katz, S, von der Lieth, A, Meyer-Roxlau, S, Weber, S, Wieland, T, Sossalla, S, Backs, J, Brown, JH, Maier, LS, and El-Armouche, A
- Subjects
beta-adrenergic blockers ,calcium ,heart failure ,calmodulin-dependent protein kinase type 2 - Published
- 2017
8. P514Investigating the mechanistic role of PDE2 in cardiac arrhythmia
- Author
-
Fahmi, M, primary, Richter, K, additional, Dybkova, N, additional, Vettel, C, additional, Dewenter, M, additional, Fischmeister, R, additional, Piorkowski, C, additional, El-Armouche, A, additional, Sossalla, S, additional, and Wagner, M, additional
- Published
- 2018
- Full Text
- View/download PDF
9. THE AP-1 TRANSCRIPTION FACTOR FOSL-2 DRIVES CARDIAC FIBROSIS AND ARRHYTHMIAS UNDER IMMUNOFIBROTIC CONDITIONS IN SYSTEMIC SCLEROSIS.
- Author
-
Stellato, M., Dewenter, M., Rudnik, M., Hukara, A., Cagla, O., Renoux, F., Pachera, E., Gantenbein, F., Seebeck, P., Uhtjaerv, S., Osto, E., Razansky, D., Klingel, K., Henes, J., Distler, O., Błyszczuk, P., and Kania, G.
- Published
- 2023
- Full Text
- View/download PDF
10. 1982Molecular mechanisms of arrhythmia upon TBX5 loss
- Author
-
Rathjens, F.R., primary, Renger, A., additional, Dewenter, M., additional, Toischer, K., additional, El-Armouche, A., additional, Zimmermann, W.H., additional, Zelarayan, L.C., additional, and Zafiriou, M.P., additional
- Published
- 2017
- Full Text
- View/download PDF
11. Tofacitinib
- Author
-
Döker, S., additional, Dewenter, M., additional, and El-Armouche, A., additional
- Published
- 2014
- Full Text
- View/download PDF
12. PDE2-mediated cAMP hydrolysis accelerates cardiac fibroblast to myofibroblast conversion and is antagonized by exogenous activation of cGMP signaling pathways
- Author
-
Vettel, C., primary, Lämmle, S., additional, Ewens, S., additional, Cervirgen, C., additional, Emons, J., additional, Ongherth, A., additional, Dewenter, M., additional, Lindner, D., additional, Westermann, D., additional, Nikolaev, V. O., additional, Lutz, S., additional, Zimmermann, W. H., additional, and El-Armouche, A., additional
- Published
- 2014
- Full Text
- View/download PDF
13. Losmapimod
- Author
-
Dewenter, M., additional, Vettel, C., additional, and El-Armouche, A., additional
- Published
- 2012
- Full Text
- View/download PDF
14. Effektivität und Sicherheit von Vareniclin zur Raucherentwöhnung
- Author
-
Christalla, P., additional, Dewenter, M., additional, and El-Armouche, A., additional
- Published
- 2012
- Full Text
- View/download PDF
15. Tofacitinib.
- Author
-
Döker, S., Dewenter, M., and El-Armouche, A.
- Published
- 2014
- Full Text
- View/download PDF
16. Phosphodiesterase-2 is upregulated in failing hearts and blunts beta-AR mediated chronotropic and inotropic effects
- Author
-
Vettel, C., Mehel, H., Emons, J., Wittkoepper, K., Dewenter, M., Laemmle, S., Seppelt, D., Riedel, M., Napiany, S., Lutz, S., Sossalla, S., Lechene, P., Maier, L., Nikolaev, V. O., Zimmermann, W. H., Eschenhagen, T., Vandecasteele, G., RODOLPHE FISCHMEISTER, and El-Armouche, A.
17. Phosphodiesterase 2 regulates resting heart rate and protects against arrhythmias and beta-adrenergic overstimulation
- Author
-
Vettel, C., Dewenter, M., Linder, M., Riedel, M., Laemmle, S., Mason, F., Meinecke, S., Wieland, T., Vandecasteele, G., Geerts, A., Wunder, F., Sossalla, S., RODOLPHE FISCHMEISTER, and El Armouche, A.
18. Ca2+/calmodulin-dependent kinase IIδC-induced chronic heart failure does not depend on sarcoplasmic reticulum Ca2+ leak.
- Author
-
Dewenter M, Seitz T, Steinbrecher JH, Westenbrink BD, Ling H, Lehnart SE, Wehrens XHT, Backs J, Brown JH, Maier LS, and Neef S
- Subjects
- Animals, Mice, Phosphorylation, Disease Models, Animal, Calcium Signaling physiology, Chronic Disease, Heart Failure metabolism, Sarcoplasmic Reticulum metabolism, Calcium-Calmodulin-Dependent Protein Kinase Type 2 metabolism, Calcium-Calmodulin-Dependent Protein Kinase Type 2 genetics, Ryanodine Receptor Calcium Release Channel metabolism, Ryanodine Receptor Calcium Release Channel genetics, Calcium metabolism, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Mice, Transgenic
- Abstract
Aims: Hyperactivity of Ca
2+ /calmodulin-dependent protein kinase II (CaMKII) has emerged as a central cause of pathologic remodelling in heart failure. It has been suggested that CaMKII-induced hyperphosphorylation of the ryanodine receptor 2 (RyR2) and consequently increased diastolic Ca2+ leak from the sarcoplasmic reticulum (SR) is a crucial mechanism by which increased CaMKII activity leads to contractile dysfunction. We aim to evaluate the relevance of CaMKII-dependent RyR2 phosphorylation for CaMKII-induced heart failure development in vivo., Methods and Results: We crossbred CaMKIIδC overexpressing [transgenic (TG)] mice with RyR2-S2814A knock-in mice that are resistant to CaMKII-dependent RyR2 phosphorylation. Ca2+ -spark measurements on isolated ventricular myocytes confirmed the severe diastolic SR Ca2+ leak previously reported in CaMKIIδC TG [4.65 ± 0.73 mF/F0 vs. 1.88 ± 0.30 mF/F0 in wild type (WT)]. Crossing in the S2814A mutation completely prevented SR Ca2+ -leak induction in the CaMKIIδC TG, both regarding Ca2+ -spark size and frequency, demonstrating that the CaMKIIδC-induced SR Ca2+ leak entirely depends on the CaMKII-specific RyR2-S2814 phosphorylation. Yet, the RyR2-S2814A mutation did not affect the massive contractile dysfunction (ejection fraction = 12.17 ± 2.05% vs. 45.15 ± 3.46% in WT), cardiac hypertrophy (heart weight/tibia length = 24.84 ± 3.00 vs. 9.81 ± 0.50 mg/mm in WT), or severe premature mortality (median survival of 12 weeks) associated with cardiac CaMKIIδC overexpression. In the face of a prevented SR Ca2+ leak, the phosphorylation status of other critical CaMKII downstream targets that can drive heart failure, including transcriptional regulator histone deacetylase 4, as well as markers of pathological gene expression including Xirp2, Il6, and Col1a1, was equally increased in hearts from CaMKIIδC TG on a RyR WT and S2814A background., Conclusions: S2814 phosphoresistance of RyR2 prevents the CaMKII-dependent SR Ca2+ leak induction but does not prevent the cardiomyopathic phenotype caused by enhanced CaMKIIδC activity. Our data indicate that additional mechanisms-independent of SR Ca2+ leak-are critical for the maladaptive effects of chronically increased CaMKIIδC activity with respect to heart failure., (© 2024 The Authors. ESC Heart Failure published by John Wiley & Sons Ltd on behalf of European Society of Cardiology.)- Published
- 2024
- Full Text
- View/download PDF
19. Transverse aortic constriction multi-omics analysis uncovers pathophysiological cardiac molecular mechanisms.
- Author
-
Gjerga E, Dewenter M, Britto-Borges T, Grosso J, Stein F, Eschenbach J, Rettel M, Backs J, and Dieterich C
- Subjects
- Animals, Mice, Heart Failure metabolism, Heart Failure genetics, Transcriptome genetics, Aorta metabolism, Gene Expression Profiling, Multiomics, Proteomics methods
- Abstract
Time-course multi-omics data of a murine model of progressive heart failure (HF) induced by transverse aortic constriction (TAC) provide insights into the molecular mechanisms that are causatively involved in contractile failure and structural cardiac remodelling. We employ Illumina-based transcriptomics, Nanopore sequencing and mass spectrometry-based proteomics on samples from the left ventricle (LV) and right ventricle (RV, RNA only) of the heart at 1, 7, 21 and 56 days following TAC and Sham surgery. Here, we present Transverse Aortic COnstriction Multi-omics Analysis (TACOMA), as an interactive web application that integrates and visualizes transcriptomics and proteomics data collected in a TAC time-course experiment. TACOMA enables users to visualize the expression profile of known and novel genes and protein products thereof. Importantly, we capture alternative splicing events by assessing differential transcript and exon usage as well. Co-expression-based clustering algorithms and functional enrichment analysis revealed overrepresented annotations of biological processes and molecular functions at the protein and gene levels. To enhance data integration, TACOMA synchronizes transcriptomics and proteomics profiles, enabling cross-omics comparisons. With TACOMA (https://shiny.dieterichlab.org/app/tacoma), we offer a rich web-based resource to uncover molecular events and biological processes implicated in contractile failure and cardiac hypertrophy. For example, we highlight: (i) changes in metabolic genes and proteins in the time course of hypertrophic growth and contractile impairment; (ii) identification of RNA splicing changes in the expression of Tpm2 isoforms between RV and LV; and (iii) novel transcripts and genes likely contributing to the pathogenesis of HF. We plan to extend these data with additional environmental and genetic models of HF to decipher common and distinct molecular changes in heart diseases of different aetiologies. Database URL: https://shiny.dieterichlab.org/app/tacoma., (© The Author(s) 2024. Published by Oxford University Press.)
- Published
- 2024
- Full Text
- View/download PDF
20. Acral lamellar ichthyosis with amino acid substitution in the C-terminus of keratin 2.
- Author
-
Frommherz L, Komlosi K, Hewel C, Kopp J, Dewenter M, Zimmer A, Bartsch O, Linke M, Technau-Hafsi K, Gerber S, Fischer J, and Has C
- Subjects
- Humans, Phenotype, Amino Acid Substitution genetics, Ichthyosis, Lamellar genetics, Keratin-2 genetics
- Abstract
Background: Most cases of hereditary ichthyoses present with generalized scaling and skin dryness. However, in some cases skin involvement is restricted to particular body regions as in acral lamellar ichthyosis., Objectives: We report on the genetic basis of acral ichthyosis in two families presenting with a similar phenotype., Methods: Genetic testing was performed by targeted next generation sequencing and whole-exome sequencing. For identity-by-descent analysis, the parents were genotyped and data analysis was performed with the Chromosome Analysis Suite Software. RT-PCR with RNA extracted from skin samples was used to analyse the effect of variants on splicing., Results: Genetic testing identified a few heterozygous variants, but only the variant in KRT2 c.1912 T > C, p.Phe638Leu segregated with the disease and remained the strongest candidate. Pairwise identity-by-descent analysis revealed no indication of family relationship. Phenylalanine 638 is the second last amino acid upstream of the termination codon in the tail of K2, and substitution to leucine is predicted as probably damaging. Assessment of the variant is difficult, in part due to the lack of crystal structures of this region., Conclusions: Altogether, we show that a type of autosomal dominant acral ichthyosis is most probably caused by an amino acid substitution in the C-terminus of keratin 2., (© 2022 The Authors. Journal of the European Academy of Dermatology and Venereology published by John Wiley & Sons Ltd on behalf of European Academy of Dermatology and Venereology.)
- Published
- 2023
- Full Text
- View/download PDF
21. Author Correction: The AP-1 transcription factor Fosl-2 drives cardiac fibrosis and arrhythmias under immunofibrotic conditions.
- Author
-
Stellato M, Dewenter M, Rudnik M, Hukara A, Özsoy Ç, Renoux F, Pachera E, Gantenbein F, Seebeck P, Uhtjaerv S, Osto E, Razansky D, Klingel K, Henes J, Distler O, Błyszczuk P, and Kania G
- Published
- 2023
- Full Text
- View/download PDF
22. CNP Promotes Antiarrhythmic Effects via Phosphodiesterase 2.
- Author
-
Cachorro E, Günscht M, Schubert M, Sadek MS, Siegert J, Dutt F, Bauermeister C, Quickert S, Berning H, Nowakowski F, Lämmle S, Firneburg R, Luo X, Künzel SR, Klapproth E, Mirtschink P, Mayr M, Dewenter M, Vettel C, Heijman J, Lorenz K, Guan K, El-Armouche A, Wagner M, and Kämmerer S
- Subjects
- Mice, Animals, Humans, Signal Transduction, Catecholamines metabolism, Arrhythmias, Cardiac drug therapy, Arrhythmias, Cardiac etiology, Arrhythmias, Cardiac prevention & control, Anti-Arrhythmia Agents pharmacology, Anti-Arrhythmia Agents therapeutic use, Anti-Arrhythmia Agents metabolism, Cyclic GMP metabolism, Natriuretic Peptide, C-Type pharmacology, Phosphoric Diester Hydrolases metabolism, Myocytes, Cardiac metabolism
- Abstract
Background: Ventricular arrhythmia and sudden cardiac death are the most common lethal complications after myocardial infarction. Antiarrhythmic pharmacotherapy remains a clinical challenge and novel concepts are highly desired. Here, we focus on the cardioprotective CNP (C-type natriuretic peptide) as a novel antiarrhythmic principle. We hypothesize that antiarrhythmic effects of CNP are mediated by PDE2 (phosphodiesterase 2), which has the unique property to be stimulated by cGMP to primarily hydrolyze cAMP. Thus, CNP might promote beneficial effects of PDE2-mediated negative crosstalk between cAMP and cGMP signaling pathways., Methods: To determine antiarrhythmic effects of cGMP-mediated PDE2 stimulation by CNP, we analyzed arrhythmic events and intracellular trigger mechanisms in mice in vivo, at organ level and in isolated cardiomyocytes as well as in human-induced pluripotent stem cell-derived cardiomyocytes., Results: In ex vivo perfused mouse hearts, CNP abrogated arrhythmia after ischemia/reperfusion injury. Upon high-dose catecholamine injections in mice, PDE2 inhibition prevented the antiarrhythmic effect of CNP. In mouse ventricular cardiomyocytes, CNP blunted the catecholamine-mediated increase in arrhythmogenic events as well as in I
CaL , INaL , and Ca2+ spark frequency. Mechanistically, this was driven by reduced cellular cAMP levels and decreased phosphorylation of Ca2+ handling proteins. Key experiments were confirmed in human iPSC-derived cardiomyocytes. Accordingly, the protective CNP effects were reversed by either specific pharmacological PDE2 inhibition or cardiomyocyte-specific PDE2 deletion., Conclusions: CNP shows strong PDE2-dependent antiarrhythmic effects. Consequently, the CNP-PDE2 axis represents a novel and attractive target for future antiarrhythmic strategies.- Published
- 2023
- Full Text
- View/download PDF
23. The AP-1 transcription factor Fosl-2 drives cardiac fibrosis and arrhythmias under immunofibrotic conditions.
- Author
-
Stellato M, Dewenter M, Rudnik M, Hukara A, Özsoy Ç, Renoux F, Pachera E, Gantenbein F, Seebeck P, Uhtjaerv S, Osto E, Razansky D, Klingel K, Henes J, Distler O, Błyszczuk P, and Kania G
- Subjects
- Animals, Humans, Mice, Arrhythmias, Cardiac genetics, Fibrosis, Mice, Transgenic, Cardiomyopathies, Transcription Factor AP-1
- Abstract
Fibrotic changes in the myocardium and cardiac arrhythmias represent fatal complications in systemic sclerosis (SSc), however the underlying mechanisms remain elusive. Mice overexpressing transcription factor Fosl-2 (Fosl-2
tg ) represent animal model of SSc. Fosl-2tg mice showed interstitial cardiac fibrosis, disorganized connexin-43/40 in intercalated discs and deregulated expression of genes controlling conduction system, and developed higher heart rate (HR), prolonged QT intervals, arrhythmias with prevalence of premature ventricular contractions, ventricular tachycardias, II-degree atrio-ventricular blocks and reduced HR variability. Following stimulation with isoproterenol Fosl-2tg mice showed impaired HR response. In contrast to Fosl-2tg , immunodeficient Rag2-/- Fosl-2tg mice were protected from enhanced myocardial fibrosis and ECG abnormalities. Transcriptomics analysis demonstrated that Fosl-2-overexpression was responsible for profibrotic signature of cardiac fibroblasts, whereas inflammatory component in Fosl-2tg mice activated their fibrotic and arrhythmogenic phenotype. In human cardiac fibroblasts FOSL-2-overexpression enhanced myofibroblast signature under proinflammatory or profibrotic stimuli. These results demonstrate that under immunofibrotic conditions transcription factor Fosl-2 exaggerates myocardial fibrosis, arrhythmias and aberrant response to stress., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
24. Endothelial deletion of the cytochrome P450 reductase leads to cardiac remodelling.
- Author
-
Lopez M, Malacarne PF, Ramanujam DP, Warwick T, Müller N, Hu J, Dewenter M, Weigert A, Günther S, Gilsbach R, Engelhardt S, Brandes RP, and Rezende F
- Abstract
The cytochrome P450 reductase (POR) transfers electrons to all microsomal cytochrome P450 enzymes (CYP450) thereby driving their activity. In the vascular system, the POR/CYP450 system has been linked to the production of epoxyeicosatrienoic acids (EETs) but also to the generation of reactive oxygen species. In cardiac myocytes (CMs), EETs have been shown to modulate the cardiac function and have cardioprotective effects. The functional importance of the endothelial POR/CYP450 system in the heart is unclear and was studied here using endothelial cell-specific, inducible knockout mice of POR (ecPOR
-/- ). RNA sequencing of murine cardiac cells revealed a cell type-specific expression of different CYP450 homologues. Cardiac endothelial cells mainly expressed members of the CYP2 family which produces EETs, and of the CYP4 family that generates omega fatty acids. Tamoxifen-induced endothelial deletion of POR in mice led to cardiac remodelling under basal conditions, as shown by an increase in heart weight to body weight ratio and an increased CM area as compared to control animals. Endothelial deletion of POR was associated with a significant increase in endothelial genes linked to protein synthesis with no changes in genes of the oxidative stress response. CM of ecPOR-/- mice exhibited attenuated expression of genes linked to mitochondrial function and an increase in genes related to cardiac myocyte contractility. In a model of pressure overload (transverse aortic constriction, TAC with O-rings), ecPOR-/- mice exhibited an accelerated reduction in cardiac output (CO) and stroke volume (SV) as compared to control mice. These results suggest that loss of endothelial POR along with a reduction in EETs leads to an increase in vascular stiffness and loss in cardioprotection, resulting in cardiac remodelling., 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 © 2022 Lopez, Malacarne, Ramanujam, Warwick, Müller, Hu, Dewenter, Weigert, Günther, Gilsbach, Engelhardt, Brandes and Rezende.)- Published
- 2022
- Full Text
- View/download PDF
25. Deep phenotyping of two preclinical mouse models and a cohort of RBM20 mutation carriers reveals no sex-dependent disease severity in RBM20 cardiomyopathy.
- Author
-
Lennermann DC, Pepin ME, Grosch M, Konrad L, Kemmling E, Hartmann J, Nolte JL, Clauder-Münster S, Kayvanpour E, Sedaghat-Hamedani F, Haas J, Meder B, van den Boogaard M, Amin AS, Dewenter M, Krüger M, Steinmetz LM, Backs J, and van den Hoogenhof MMG
- Subjects
- Mice, Male, Female, Animals, Arrhythmias, Cardiac genetics, Mutation, Mice, Knockout, Severity of Illness Index, RNA-Binding Proteins genetics, Cardiomyopathies
- Abstract
RBM20 cardiomyopathy is an arrhythmogenic form of dilated cardiomyopathy caused by mutations in the splicing factor RBM20. A recent study found a more severe phenotype in male patients with RBM20 cardiomyopathy patients than in female patients. Here, we aim to determine sex differences in an animal model of RBM20 cardiomyopathy and investigate potential underlying mechanisms. In addition, we aim to determine sex and gender differences in clinical parameters in a novel RBM20 cardiomyopathy patient cohort. We characterized an Rbm20 knockout (KO) mouse model, and show that splicing of key RBM20 targets, cardiac function, and arrhythmia susceptibility do not differ between sexes. Next, we performed deep phenotyping of these mice, and show that male and female Rbm20 -KO mice possess transcriptomic and phosphoproteomic differences. Hypothesizing that these differences may influence the heart's ability to compensate for stress, we exposed Rbm20 -KO mice to acute catecholaminergic stimulation and again found no functional differences. We also replicate the lack of functional differences in a mouse model with the Rbm20 -R636Q mutation. Lastly, we present a patient cohort of 33 RBM20 cardiomyopathy patients and show that these patients do not possess sex and gender differences in disease severity. Current mouse models of RBM20 cardiomyopathy show more pronounced changes in gene expression and phosphorylation of cardiac proteins in male mice, but no sex differences in cardiac morphology and function. Moreover, other than reported before, male RBM20 cardiomyopathy patients do not present with worse cardiac function in a patient cohort from Germany and the Netherlands. NEW & NOTEWORTHY Optimal management of the cardiac disease is increasingly personalized, partly because of differences in outcomes between sexes. RBM20 cardiomyopathy has been described to be more severe in male patients, and this carries the risk that male patients are more scrutinized in the clinic than female patients. Our findings do not support this observation and suggest that treatment should not differ between male and female RBM20 cardiomyopathy patients, but instead should focus on the underlying disease mechanism.
- Published
- 2022
- Full Text
- View/download PDF
26. Forebrain corticosteroid receptors promote post-myocardial infarction depression and mortality.
- Author
-
Bruns B, Daub R, Schmitz T, Hamze-Sinno M, Spaich S, Dewenter M, Schwale C, Gass P, Vogt M, Katus H, Herzog W, Friederich HC, Frey N, Schultz JH, and Backs J
- Subjects
- Animals, Humans, Mice, Myocardium pathology, Prosencephalon metabolism, Receptors, Glucocorticoid metabolism, Depression, Myocardial Infarction pathology
- Abstract
Myocardial infarction (MI) with subsequent depression is associated with increased cardiac mortality. Impaired central mineralocorticoid (MR) and glucocorticoid receptor (GR) equilibrium has been suggested as a key mechanism in the pathogenesis of human depression. Here, we investigate if deficient central MR/GR signaling is causative for a poor outcome after MI in mice. Mice with an inducible forebrain-specific MR/GR knockout (MR/GR-KO) underwent baseline and follow-up echocardiography every 2 weeks after MI or sham operation. Behavioral testing at 4 weeks confirmed significant depressive-like behavior and, strikingly, a higher mortality after MI, while cardiac function and myocardial damage remained unaffected. Telemetry revealed cardiac autonomic imbalance with marked bradycardia and ventricular tachycardia (VT) upon MI in MR/GR-KO. Mechanistically, we found a higher responsiveness to atropine, pointing to impaired parasympathetic tone of 'depressive' mice after MI. Serum corticosterone levels were increased but-in line with the higher vagal tone-plasma and cardiac catecholamines were decreased. MR/GR deficiency in the forebrain led to significant depressive-like behavior and a higher mortality after MI. This was accompanied by increased vagal tone, depleted catecholaminergic compensatory capacity and VTs. Thus, limbic MR/GR disequilibrium may contribute to the impaired outcome of depressive patients after MI and possibly explain the lack of anti-depressive treatment benefit., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
27. Further characterization of Borjeson-Forssman-Lehmann syndrome in females due to de novo variants in PHF6.
- Author
-
Gerber CB, Fliedner A, Bartsch O, Berland S, Dewenter M, Haug M, Hayes I, Marin-Reina P, Mark PR, Martinez-Castellano F, Maystadt I, Karadurmus D, Steindl K, Wiesener A, Zweier M, Sticht H, and Zweier C
- Subjects
- Epilepsy, Face abnormalities, Female, Fingers abnormalities, Growth Disorders, Humans, Male, Obesity, Hypogonadism genetics, Intellectual Disability complications, Mental Retardation, X-Linked genetics, Musculoskeletal Abnormalities complications, Repressor Proteins genetics
- Abstract
While inherited hemizygous variants in PHF6 cause X-linked recessive Borjeson-Forssman-Lehmann syndrome (BFLS) in males, de novo heterozygous variants in females are associated with an overlapping but distinct phenotype, including moderate to severe intellectual disability, characteristic facial dysmorphism, dental, finger and toe anomalies, and linear skin pigmentation. By personal communication with colleagues, we assembled 11 additional females with BFLS due to variants in PHF6. We confirm the distinct phenotype to include variable intellectual disability, recognizable facial dysmorphism and other anomalies. We observed skewed X-inactivation in blood and streaky skin pigmentation compatible with functional mosaicism. Variants occurred de novo in 10 individuals, of whom one was only mildly affected and transmitted it to her more severely affected daughter. The mutational spectrum comprises a two-exon deletion, five truncating, one splice-site and three missense variants, the latter all located in the PHD2 domain and predicted to severely destabilize the domain structure. This observation supports the hypothesis of more severe variants in females contributing to gender-specific phenotypes in addition to or in combination with effects of X-inactivation and functional mosaicism. Therefore, our findings further delineate the clinical and mutational spectrum of female BFLS and provide further insights into possible genotype-phenotype correlations between females and males., (© 2022 The Authors. Clinical Genetics published by John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
28. Ketone body oxidation increases cardiac endothelial cell proliferation.
- Author
-
Weis EM, Puchalska P, Nelson AB, Taylor J, Moll I, Hasan SS, Dewenter M, Hagenmüller M, Fleming T, Poschet G, Hotz-Wagenblatt A, Backs J, Crawford PA, and Fischer A
- Subjects
- Animals, Cell Proliferation, Glucose metabolism, Mice, Myocytes, Cardiac metabolism, Endothelial Cells metabolism, Ketone Bodies metabolism
- Abstract
Blood vessel formation is dependent on metabolic adaption in endothelial cells. Glucose and fatty acids are essential substrates for ATP and biomass production; however, the metabolism of other substrates remains poorly understood. Ketone bodies are important nutrients for cardiomyocytes during starvation or consumption of carbohydrate-restrictive diets. This raises the question whether cardiac endothelial cells would not only transport ketone bodies but also consume some of these to achieve their metabolic needs. Here, we report that cardiac endothelial cells are able to oxidize ketone bodies and that this enhances cell proliferation, migration, and vessel sprouting. Mechanistically, this requires succinyl-CoA:3-oxoacid-CoA transferase, a key enzyme of ketone body oxidation. Targeted metabolite profiling revealed that carbon from ketone bodies got incorporated into tricarboxylic acid cycle intermediates as well as other metabolites fueling biomass production. Elevation of ketone body levels by a high-fat, low-carbohydrate ketogenic diet transiently increased endothelial cell proliferation in mouse hearts. Notably, in a mouse model of heart hypertrophy, ketogenic diet prevented blood vessel rarefication. This suggests a potential beneficial role of dietary intervention in heart diseases., (© 2022 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2022
- Full Text
- View/download PDF
29. Chronic isoprenaline/phenylephrine vs. exclusive isoprenaline stimulation in mice: critical contribution of alpha 1 -adrenoceptors to early cardiac stress responses.
- Author
-
Dewenter M, Pan J, Knödler L, Tzschöckel N, Henrich J, Cordero J, Dobreva G, Lutz S, Backs J, Wieland T, and Vettel C
- Subjects
- Animals, Isoproterenol pharmacology, Mice, Phenylephrine pharmacology, Receptors, Adrenergic, beta, Heart, Receptors, Adrenergic, alpha-1 genetics, Receptors, Adrenergic, alpha-1 metabolism
- Abstract
Hyperactivity of the sympathetic nervous system is a major driver of cardiac remodeling, exerting its effects through both α-, and β-adrenoceptors (α-, β-ARs). As the relative contribution of subtype α
1 -AR to cardiac stress responses remains poorly investigated, we subjected mice to either subcutaneous perfusion with the β-AR agonist isoprenaline (ISO, 30 mg/kg × day) or to a combination of ISO and the stable α1 -AR agonist phenylephrine (ISO/PE, 30 mg/kg × day each). Telemetry analysis revealed similar hemodynamic responses under both ISO and ISO/PE treatment i.e., permanently increased heart rates and only transient decreases in mean blood pressure during the first 24 h. Echocardiography and single cell analysis after 1 week of exposure showed that ISO/PE-, but not ISO-treated animals established α1 -AR-mediated inotropic responsiveness to acute adrenergic stimulation. Morphologically, additional PE perfusion limited concentric cardiomyocyte growth and enhanced cardiac collagen deposition during 7 days of treatment. Time-course analysis demonstrated a diverging development in transcriptional patterns at day 4 of treatment i.e., increased expression of selected marker genes Xirp2, Nppa, Tgfb1, Col1a1, Postn under chronic ISO/PE treatment which was either less pronounced or absent in the ISO group. Transcriptome analyses at day 4 via RNA sequencing demonstrated that additional PE treatment caused a marked upregulation of genes allocated to extracellular matrix and fiber organization along with a more pronounced downregulation of genes involved in metabolic processes, muscle adaptation and cardiac electrophysiology. Consistently, transcriptome changes under ISO/PE challenge more effectively recapitulated early transcriptional alterations in pressure overload-induced experimental heart failure and in human hypertrophic cardiomyopathy., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
30. Preclinical evidence for the therapeutic value of TBX5 normalization in arrhythmia control.
- Author
-
Rathjens FS, Blenkle A, Iyer LM, Renger A, Syeda F, Noack C, Jungmann A, Dewenter M, Toischer K, El-Armouche A, Müller OJ, Fabritz L, Zimmermann WH, Zelarayan LC, and Zafeiriou MP
- Subjects
- Animals, Arrhythmias, Cardiac genetics, Arrhythmias, Cardiac metabolism, Arrhythmias, Cardiac physiopathology, Chromatin Immunoprecipitation Sequencing, Death, Sudden, Cardiac etiology, Disease Models, Animal, Gene Expression Profiling, Genetic Therapy, Heart Rate, Heart Ventricles physiopathology, Hypertrophy, Left Ventricular genetics, Hypertrophy, Left Ventricular metabolism, Hypertrophy, Left Ventricular physiopathology, Isolated Heart Preparation, Mice, Inbred C57BL, Mice, Knockout, RNA-Seq, T-Box Domain Proteins genetics, Transcription, Genetic, Transcriptome, Ventricular Dysfunction, Left genetics, Ventricular Dysfunction, Left metabolism, Ventricular Dysfunction, Left physiopathology, Ventricular Function, Left, Ventricular Remodeling, Mice, Arrhythmias, Cardiac prevention & control, Death, Sudden, Cardiac prevention & control, Heart Ventricles metabolism, Hypertrophy, Left Ventricular therapy, T-Box Domain Proteins metabolism, Ventricular Dysfunction, Left therapy
- Abstract
Aims: Arrhythmias and sudden cardiac death (SCD) occur commonly in patients with heart failure. We found T-box 5 (TBX5) dysregulated in ventricular myocardium from heart failure patients and thus we hypothesized that TBX5 reduction contributes to arrhythmia development in these patients. To understand the underlying mechanisms, we aimed to reveal the ventricular TBX5-dependent transcriptional network and further test the therapeutic potential of TBX5 level normalization in mice with documented arrhythmias., Methods and Results: We used a mouse model of TBX5 conditional deletion in ventricular cardiomyocytes. Ventricular (v) TBX5 loss in mice resulted in mild cardiac dysfunction and arrhythmias and was associated with a high mortality rate (60%) due to SCD. Upon angiotensin stimulation, vTbx5KO mice showed exacerbated cardiac remodelling and dysfunction suggesting a cardioprotective role of TBX5. RNA-sequencing of a ventricular-specific TBX5KO mouse and TBX5 chromatin immunoprecipitation was used to dissect TBX5 transcriptional network in cardiac ventricular tissue. Overall, we identified 47 transcripts expressed under the control of TBX5, which may have contributed to the fatal arrhythmias in vTbx5KO mice. These included transcripts encoding for proteins implicated in cardiac conduction and contraction (Gja1, Kcnj5, Kcng2, Cacna1g, Chrm2), in cytoskeleton organization (Fstl4, Pdlim4, Emilin2, Cmya5), and cardiac protection upon stress (Fhl2, Gpr22, Fgf16). Interestingly, after TBX5 loss and arrhythmia development in vTbx5KO mice, TBX5 protein-level normalization by systemic adeno-associated-virus (AAV) 9 application, re-established TBX5-dependent transcriptome. Consequently, cardiac dysfunction was ameliorated and the propensity of arrhythmia occurrence was reduced., Conclusions: This study uncovers a novel cardioprotective role of TBX5 in the adult heart and provides preclinical evidence for the therapeutic value of TBX5 protein normalization in the control of arrhythmia., (© The Author(s) 2020. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2021
- Full Text
- View/download PDF
31. Adaptive versus maladaptive cardiac remodelling in response to sustained β-adrenergic stimulation in a new 'ISO on/off model'.
- Author
-
Werhahn SM, Kreusser JS, Hagenmüller M, Beckendorf J, Diemert N, Hoffmann S, Schultz JH, Backs J, and Dewenter M
- Subjects
- Animals, Mice, Calcium-Binding Proteins metabolism, Calcium-Binding Proteins genetics, Cardiomegaly metabolism, Cardiomegaly chemically induced, Cardiomegaly physiopathology, Male, Heart Failure metabolism, Heart Failure physiopathology, Adaptation, Physiological drug effects, Receptors, Adrenergic, beta metabolism, Calcium-Calmodulin-Dependent Protein Kinase Type 2 metabolism, Atrial Natriuretic Factor metabolism, Mice, Inbred C57BL, Phosphorylation drug effects, Myosin Heavy Chains metabolism, Myosin Heavy Chains genetics, Myocardial Contraction drug effects, Signal Transduction drug effects, Disease Models, Animal, Histone Deacetylases, Isoproterenol pharmacology, Ventricular Remodeling drug effects, Adrenergic beta-Agonists pharmacology
- Abstract
On the one hand, sustained β-adrenergic stress is a hallmark of heart failure (HF) and exerts maladaptive cardiac remodelling. On the other hand, acute β-adrenergic stimulation maintains cardiac function under physiological stress. However, it is still incompletely understood to what extent the adaptive component of β-adrenergic signaling contributes to the maintenance of cardiac function during chronic β-adrenergic stress. We developed an experimental catecholamine-based protocol to distinguish adaptive from maladaptive effects. Mice were for 28 days infused with 30 mg/kg body weight/day isoproterenol (ISO) by subcutaneously implanted osmotic minipumps ('ISO on'). In a second and third group, ISO infusion was stopped after 26 days and the mice were observed for additional two or seven days without further ISO infusion ('ISO off short', 'ISO off long'). In this setup, 'ISO on' led to cardiac hypertrophy and slightly improved cardiac contractility. In stark contrast, 'ISO off' mice displayed progressive worsening of left ventricular ejection fraction that dropped down below 40%. While fetal and pathological gene expression (increase in Nppa, decrease in Myh6/Myh7 ratios, increase in Xirp2) was not induced in 'ISO on', it was activated in 'ISO off' mice. After ISO withdrawal, phosphorylation of phospholamban (PLN) at the protein kinase A (PKA) phosphorylation site Ser-16 dropped down to 20% as compared to only 50% at the Ca2+/Calmodulin-dependent kinase II (CaMKII) phosphorylation site Thr-17 in 'ISO off' mice. PKA-dependent cardioprotective production of the N-terminal proteolytic product of histone deacetylase 4 (HDAC4-NT) was reduced in 'ISO off' as compared to 'ISO on'. Taken together, these data indicate that chronic ISO infusion induces besides maladaptive remodelling also adaptive PKA signalling to maintain cardiac function. The use of the 'ISO on/off' model will further enable the separation of the underlying adaptive from maladaptive components of β-adrenergic signalling and may help to better define and test therapeutic targets downstream of β-adrenergic receptors., Competing Interests: Johannes Backs and Matthias Dewenter collaborate with the Lead Discovery Center (LDC) Dortmund to develop CaMKII (Calmodulin-dependent Kinase II)-HDAC4 (histone deacetylase 4) inhibitory compounds. Johannes Backs is scientific cofounder of Artemes Bio, holds a patent on “ABHD5 and partial HDAC4 fragments and variants as a therapeutic approach for the treatment of cardiovascular diseases“, and received personal fees from Bayer outside the submitted work. This does not alter our adherence to PLOS ONE policies on sharing data and materials.
- Published
- 2021
- Full Text
- View/download PDF
32. Loss of CASK Accelerates Heart Failure Development.
- Author
-
Mustroph J, Sag CM, Bähr F, Schmidtmann AL, Gupta SN, Dietz A, Islam MMT, Lücht C, Beuthner BE, Pabel S, Baier MJ, El-Armouche A, Sossalla S, Anderson ME, Möllmann J, Lehrke M, Marx N, Mohler PJ, Bers DM, Unsöld B, He T, Dewenter M, Backs J, Maier LS, and Wagner S
- Subjects
- Animals, Calcium metabolism, Calcium-Calmodulin-Dependent Protein Kinase Type 2 metabolism, Cells, Cultured, Glucagon-Like Peptide-1 Receptor metabolism, Guanylate Kinases genetics, Heart Failure genetics, Heart Failure physiopathology, Heart Rate, Humans, Male, Mice, Mice, Inbred C57BL, Myocardial Contraction, Myocytes, Cardiac metabolism, Guanylate Kinases metabolism, Heart Failure metabolism
- Abstract
[Figure: see text].
- Published
- 2021
- Full Text
- View/download PDF
33. CaM Kinase II-δ Is Required for Diabetic Hyperglycemia and Retinopathy but Not Nephropathy.
- Author
-
Chen J, Fleming T, Katz S, Dewenter M, Hofmann K, Saadatmand A, Kronlage M, Werner MP, Pokrandt B, Schreiter F, Lin J, Katz D, Morgenstern J, Elwakiel A, Sinn P, Gröne HJ, Hammes HP, Nawroth PP, Isermann B, Sticht C, Brügger B, Katus HA, Hagenmueller M, and Backs J
- Subjects
- Animals, Calcium-Calmodulin-Dependent Protein Kinase Type 2 genetics, Diabetes Mellitus, Type 2 genetics, Diabetic Nephropathies genetics, Diabetic Retinopathy genetics, Gene Expression, Hyperglycemia genetics, Mice, Mice, Knockout, Receptors, Leptin genetics, Receptors, Leptin metabolism, Calcium-Calmodulin-Dependent Protein Kinase Type 2 metabolism, Diabetes Mellitus, Type 2 metabolism, Diabetic Nephropathies metabolism, Diabetic Retinopathy metabolism, Hyperglycemia metabolism
- Abstract
Type 2 diabetes has become a pandemic and leads to late diabetic complications of organs, including kidney and eye. Lowering hyperglycemia is the typical therapeutic goal in clinical medicine. However, hyperglycemia may only be a symptom of diabetes but not the sole cause of late diabetic complications; instead, other diabetes-related alterations could be causative. Here, we studied the role of CaM kinase II-δ (CaMKIIδ), which is known to be activated through diabetic metabolism. CaMKIIδ is expressed ubiquitously and might therefore affect several different organ systems. We crossed diabetic leptin receptor-mutant mice to mice lacking CaMKIIδ globally. Remarkably, CaMKIIδ-deficient diabetic mice did not develop hyperglycemia. As potential underlying mechanisms, we provide evidence for improved insulin sensing with increased glucose transport into skeletal muscle and also reduced hepatic glucose production. Despite normoglycemia, CaMKIIδ-deficient diabetic mice developed the full picture of diabetic nephropathy, but diabetic retinopathy was prevented. We also unmasked a retina-specific gene expression signature that might contribute to CaMKII-dependent retinal diabetic complications. These data challenge the clinical concept of normalizing hyperglycemia in diabetes as a causative treatment strategy for late diabetic complications and call for a more detailed analysis of intracellular metabolic signals in different diabetic organs., (© 2020 by the American Diabetes Association.)
- Published
- 2021
- Full Text
- View/download PDF
34. Nitro-fatty acids suppress ischemic ventricular arrhythmias by preserving calcium homeostasis.
- Author
-
Mollenhauer M, Mehrkens D, Klinke A, Lange M, Remane L, Friedrichs K, Braumann S, Geißen S, Simsekyilmaz S, Nettersheim FS, Lee S, Peinkofer G, Geisler AC, Geis B, Schwoerer AP, Carrier L, Freeman BA, Dewenter M, Luo X, El-Armouche A, Wagner M, Adam M, Baldus S, and Rudolph V
- Subjects
- Animals, Calcium-Calmodulin-Dependent Protein Kinase Type 2 metabolism, Catecholamines pharmacology, Dietary Supplements, Homeostasis drug effects, Isoproterenol pharmacology, Male, Mice, Inbred Strains, Myocardial Ischemia complications, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Phosphorylation drug effects, Ryanodine Receptor Calcium Release Channel metabolism, Tachycardia, Ventricular etiology, Tachycardia, Ventricular prevention & control, Anti-Arrhythmia Agents pharmacology, Arrhythmias, Cardiac drug therapy, Arrhythmias, Cardiac metabolism, Calcium metabolism, Nitro Compounds pharmacology, Oleic Acids pharmacology
- Abstract
Nitro-fatty acids are electrophilic anti-inflammatory mediators which are generated during myocardial ischemic injury. Whether these species exert anti-arrhythmic effects in the acute phase of myocardial ischemia has not been investigated so far. Herein, we demonstrate that pretreatment of mice with 9- and 10-nitro-octadec-9-enoic acid (nitro-oleic acid, NO
2 -OA) significantly reduced the susceptibility to develop acute ventricular tachycardia (VT). Accordingly, epicardial mapping revealed a markedly enhanced homogeneity in ventricular conduction. NO2 -OA treatment of isolated cardiomyocytes lowered the number of spontaneous contractions upon adrenergic isoproterenol stimulation and nearly abolished ryanodine receptor type 2 (RyR2)-dependent sarcoplasmic Ca2+ leak. NO2 -OA also significantly reduced RyR2-phosphorylation by inhibition of increased CaMKII activity. Thus, NO2 -OA might be a novel pharmacological option for the prevention of VT development.- Published
- 2020
- Full Text
- View/download PDF
35. Age-related hearing loss influences functional connectivity of auditory cortex for the McGurk illusion.
- Author
-
Rosemann S, Smith D, Dewenter M, and Thiel CM
- Subjects
- Acoustic Stimulation, Aged, Auditory Perception, Humans, Photic Stimulation, Visual Perception, Auditory Cortex, Hearing Loss, Illusions, Speech Perception
- Abstract
Age-related hearing loss affects hearing at high frequencies and is associated with difficulties in understanding speech. Increased audio-visual integration has recently been found in age-related hearing impairment, the brain mechanisms that contribute to this effect are however unclear. We used functional magnetic resonance imaging in elderly subjects with normal hearing and mild to moderate uncompensated hearing loss. Audio-visual integration was studied using the McGurk task. In this task, an illusionary fused percept can occur if incongruent auditory and visual syllables are presented. The paradigm included unisensory stimuli (auditory only, visual only), congruent audio-visual and incongruent (McGurk) audio-visual stimuli. An illusionary precept was reported in over 60% of incongruent trials. These McGurk illusion rates were equal in both groups of elderly subjects and correlated positively with speech-in-noise perception and daily listening effort. Normal-hearing participants showed an increased neural response in left pre- and postcentral gyri and right middle frontal gyrus for incongruent stimuli (McGurk) compared to congruent audio-visual stimuli. Activation patterns were however not different between groups. Task-modulated functional connectivity differed between groups showing increased connectivity from auditory cortex to visual, parietal and frontal areas in hard of hearing participants as compared to normal-hearing participants when comparing incongruent stimuli (McGurk) with congruent audio-visual stimuli. These results suggest that changes in functional connectivity of auditory cortex rather than activation strength during processing of audio-visual McGurk stimuli accompany age-related hearing loss., Competing Interests: Declaration of Competing Interest The authors declare no competing interests., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
36. CaMKII does not control mitochondrial Ca 2+ uptake in cardiac myocytes.
- Author
-
Nickel AG, Kohlhaas M, Bertero E, Wilhelm D, Wagner M, Sequeira V, Kreusser MM, Dewenter M, Kappl R, Hoth M, Dudek J, Backs J, and Maack C
- Subjects
- Animals, Calcium, Mice, Reactive Oxygen Species, Sarcoplasmic Reticulum, Calcium-Calmodulin-Dependent Protein Kinase Type 2, Myocytes, Cardiac
- Abstract
Key Points: Mitochondrial Ca
2+ uptake stimulates the Krebs cycle to regenerate the reduced forms of pyridine nucleotides (NADH, NADPH and FADH2 ) required for ATP production and reactive oxygen species (ROS) elimination. Ca2+ /calmodulin-dependent protein kinase II (CaMKII) has been proposed to regulate mitochondrial Ca2+ uptake via mitochondrial Ca2+ uniporter phosphorylation. We used two mouse models with either global deletion of CaMKIIδ (CaMKIIδ knockout) or cardiomyocyte-specific deletion of CaMKIIδ and γ (CaMKIIδ/γ double knockout) to interrogate whether CaMKII controls mitochondrial Ca2+ uptake in isolated mitochondria and during β-adrenergic stimulation in cardiac myocytes. CaMKIIδ/γ did not control Ca2+ uptake, respiration or ROS emission in isolated cardiac mitochondria, nor in isolated cardiac myocytes, during β-adrenergic stimulation and pacing. The results of the present study do not support a relevant role of CaMKII for mitochondrial Ca2+ uptake in cardiac myocytes under physiological conditions., Abstract: Mitochondria are the main source of ATP and reactive oxygen species (ROS) in cardiac myocytes. Furthermore, activation of the mitochondrial permeability transition pore (mPTP) induces programmed cell death. These processes are essentially controlled by Ca2+ , which is taken up into mitochondria via the mitochondrial Ca2+ uniporter (MCU). It was recently proposed that Ca2+ /calmodulin-dependent protein kinase II (CaMKII) regulates Ca2+ uptake by interacting with the MCU, thereby affecting mPTP activation and programmed cell death. In the present study, we investigated the role of CaMKII under physiological conditions in which mitochondrial Ca2+ uptake matches energy supply to the demand of cardiac myocytes. Accordingly, we measured mitochondrial Ca2+ uptake in isolated mitochondria and cardiac myocytes harvested from cardiomyocyte-specific CaMKII δ and γ double knockout (KO) (CaMKIIδ/γ DKO) and global CaMKIIδ KO mice. To simulate a physiological workload increase, cardiac myocytes were subjected to β-adrenergic stimulation (by isoproterenol superfusion) and an increase in stimulation frequency (from 0.5 to 5 Hz). No differences in mitochondrial Ca2+ accumulation were detected in isolated mitochondria or cardiac myocytes from both CaMKII KO models compared to wild-type littermates. Mitochondrial redox state and ROS production were unchanged in CaMKIIδ/γ DKO, whereas we observed a mild oxidation of mitochondrial redox state and an increase in H2 O2 emission from CaMKIIδ KO cardiac myocytes exposed to an increase in workload. In conclusion, the results obtained in the present study do not support the regulation of mitochondrial Ca2+ uptake via the MCU or mPTP activation by CaMKII in cardiac myocytes under physiological conditions., (© 2019 The Authors. The Journal of Physiology © 2019 The Physiological Society.)- Published
- 2020
- Full Text
- View/download PDF
37. The lipid droplet-associated protein ABHD5 protects the heart through proteolysis of HDAC4.
- Author
-
Jebessa ZH, Shanmukha KD, Dewenter M, Lehmann LH, Xu C, Schreiter F, Siede D, Gong XM, Worst BC, Federico G, Sauer SW, Fischer T, Wechselberger L, Müller OJ, Sossalla S, Dieterich C, Most P, Gröne HJ, Moro C, Oberer M, Haemmerle G, Katus HA, Tyedmers J, and Backs J
- Subjects
- 3T3-L1 Cells, Animals, Heart Failure prevention & control, Humans, Mice, Protein Binding, Proteolysis, Serine Proteases metabolism, 1-Acylglycerol-3-Phosphate O-Acyltransferase metabolism, Histone Deacetylases metabolism, Lipid Droplets, Repressor Proteins metabolism
- Abstract
Catecholamines stimulate the first step of lipolysis by PKA-dependent release of the lipid droplet-associated protein ABHD5 from perilipin to co-activate the lipase ATGL. Here, we unmask a yet unrecognized proteolytic and cardioprotective function of ABHD5. ABHD5 acts in vivo and in vitro as a serine protease cleaving HDAC4. Through the production of an N-terminal polypeptide of HDAC4 (HDAC4-NT), ABHD5 inhibits MEF2-dependent gene expression and thereby controls glucose handling. ABHD5-deficiency leads to neutral lipid storage disease in mice. Cardiac-specific gene therapy of HDAC4-NT does not protect from intra-cardiomyocyte lipid accumulation but strikingly from heart failure, thereby challenging the concept of lipotoxicity-induced heart failure. ABHD5 levels are reduced in failing human hearts and murine transgenic ABHD5 expression protects from pressure-overload induced heart failure. These findings represent a conceptual advance by connecting lipid with glucose metabolism through HDAC4 proteolysis and enable new translational approaches to treat cardiometabolic disease., Competing Interests: Competing Interests Statement Z.H.J., L.H.L, O.J.M, H.A.K. and J.B. filed a patent on HDAC4-NT and ABHD5 gene therapy (US9914912B2). All other authors declare no conflict of interest.
- Published
- 2019
- Full Text
- View/download PDF
38. CaM kinase II regulates cardiac hemoglobin expression through histone phosphorylation upon sympathetic activation.
- Author
-
Saadatmand AR, Sramek V, Weber S, Finke D, Dewenter M, Sticht C, Gretz N, Wüstemann T, Hagenmueller M, Kuenzel SR, Meyer-Roxlau S, Kramer M, Sossalla S, Lehmann LH, Kämmerer S, Backs J, and El-Armouche A
- Subjects
- Adrenergic beta-Antagonists pharmacology, Adult, Animals, Catecholamines pharmacology, Cells, Cultured, Female, Heart Failure genetics, Hemoglobins metabolism, Humans, Male, Mice, Middle Aged, Phosphorylation, Rats, Sympathetic Nervous System drug effects, Calcium-Calmodulin-Dependent Protein Kinase Type 2 metabolism, Heart Failure metabolism, Hemoglobins genetics, Histone Code, Histones metabolism, Myocardium metabolism, Sympathetic Nervous System physiology
- Abstract
Sympathetic activation of β-adrenoreceptors (β-AR) represents a hallmark in the development of heart failure (HF). However, little is known about the underlying mechanisms of gene regulation. In human ventricular myocardium from patients with end-stage HF, we found high levels of phosphorylated histone 3 at serine-28 (H3S28p). H3S28p was increased by inhibition of the catecholamine-sensitive protein phosphatase 1 and decreased by β-blocker pretreatment. By a series of in vitro and in vivo experiments, we show that the β-AR downstream protein kinase CaM kinase II (CaMKII) directly binds and phosphorylates H3S28. Whereas, in CaMKII-deficient myocytes, acute catecholaminergic stimulation resulted in some degree of H3S28p, sustained catecholaminergic stimulation almost entirely failed to induce H3S28p. Genome-wide analysis of CaMKII-mediated H3S28p in response to chronic β-AR stress by chromatin immunoprecipitation followed by massive genomic sequencing led to the identification of CaMKII-dependent H3S28p target genes. Forty percent of differentially H3S28p-enriched genomic regions were associated with differential, mostly increased expression of the nearest genes, pointing to CaMKII-dependent H3S28p as an activating histone mark. Remarkably, the adult hemoglobin genes showed an H3S28p enrichment close to their transcriptional start or end sites, which was associated with increased messenger RNA and protein expression. In summary, we demonstrate that chronic β-AR activation leads to CaMKII-mediated H3S28p in cardiomyocytes. Thus, H3S28p-dependent changes may play an unexpected role for cardiac hemoglobin regulation in the context of sympathetic activation. These data also imply that CaMKII may be a yet unrecognized stress-responsive regulator of hematopoesis., Competing Interests: The authors declare no competing interest.
- Published
- 2019
- Full Text
- View/download PDF
39. O-GlcNAcylation of Histone Deacetylase 4 Protects the Diabetic Heart From Failure.
- Author
-
Kronlage M, Dewenter M, Grosso J, Fleming T, Oehl U, Lehmann LH, Falcão-Pires I, Leite-Moreira AF, Volk N, Gröne HJ, Müller OJ, Sickmann A, Katus HA, and Backs J
- Subjects
- Animals, Animals, Newborn, Cells, Cultured, Diabetes Mellitus, Experimental pathology, Diabetes Mellitus, Type 1 pathology, Heart Failure pathology, Humans, Male, Mice, Mice, 129 Strain, Mice, Inbred C57BL, Mice, Knockout, Rats, Rats, Wistar, Serine metabolism, Diabetes Mellitus, Experimental metabolism, Diabetes Mellitus, Type 1 metabolism, Heart Failure metabolism, Heart Failure prevention & control, Histone Deacetylases metabolism, Repressor Proteins metabolism
- Abstract
Background: Worldwide, diabetes mellitus and heart failure represent frequent comorbidities with high socioeconomic impact and steadily growing incidence, calling for a better understanding of how diabetic metabolism promotes cardiac dysfunction. Paradoxically, some glucose-lowering drugs have been shown to worsen heart failure, raising the question of how glucose mediates protective versus detrimental cardiac signaling. Here, we identified a histone deacetylase 4 (HDAC4) subdomain as a molecular checkpoint of adaptive and maladaptive signaling in the diabetic heart., Methods: A conditional HDAC4 allele was used to delete HDAC4 specifically in cardiomyocytes (HDAC4-knockout). Mice were subjected to diabetes mellitus either by streptozotocin injections (type 1 diabetes mellitus model) or by crossing into mice carrying a leptin receptor mutation (db/db; type 2 diabetes mellitus model) and monitored for remodeling and cardiac function. Effects of glucose and the posttranslational modification by β-linked N-acetylglucosamine (O-GlcNAc) on HDAC4 were investigated in vivo and in vitro by biochemical and cellular assays., Results: We show that the cardio-protective N-terminal proteolytic fragment of HDAC4 is enhanced in vivo in patients with diabetes mellitus and mouse models, as well as in vitro under high-glucose and high-O-GlcNAc conditions. HDAC4-knockout mice develop heart failure in models of type 1 and type 2 diabetes mellitus, whereas wild-type mice do not develop clear signs of heart failure, indicating that HDAC4 protects the diabetic heart. Reexpression of the N-terminal fragment of HDAC4 prevents HDAC4-dependent diabetic cardiomyopathy. Mechanistically, the posttranslational modification of HDAC4 at serine (Ser)-642 by O-GlcNAcylation is an essential step for production of the N-terminal fragment of HDAC4, which was attenuated by Ca
2+ /calmodulin-dependent protein kinase II-mediated phosphorylation at Ser-632. Preventing O-GlcNAcylation at Ser-642 not only entirely precluded production of the N-terminal fragment of HDAC4 but also promoted Ca2+ /calmodulin-dependent protein kinase II-mediated phosphorylation at Ser-632, pointing to a mutual posttranslational modification cross talk of (cardio-detrimental) phosphorylation at Ser-632 and (cardio-protective) O-GlcNAcylation at Ser-642., Conclusions: In this study, we found that O-GlcNAcylation of HDAC4 at Ser-642 is cardio-protective in diabetes mellitus and counteracts pathological Ca2+ /calmodulin-dependent protein kinase II signaling. We introduce a molecular model explaining how diabetic metabolism possesses important cardio-protective features besides its known detrimental effects. A deeper understanding of the here-described posttranslational modification cross talk may lay the groundwork for the development of specific therapeutic concepts to treat heart failure in the context of diabetes mellitus.- Published
- 2019
- Full Text
- View/download PDF
40. A proteolytic fragment of histone deacetylase 4 protects the heart from failure by regulating the hexosamine biosynthetic pathway.
- Author
-
Lehmann LH, Jebessa ZH, Kreusser MM, Horsch A, He T, Kronlage M, Dewenter M, Sramek V, Oehl U, Krebs-Haupenthal J, von der Lieth AH, Schmidt A, Sun Q, Ritterhoff J, Finke D, Völkers M, Jungmann A, Sauer SW, Thiel C, Nickel A, Kohlhaas M, Schäfer M, Sticht C, Maack C, Gretz N, Wagner M, El-Armouche A, Maier LS, Londoño JEC, Meder B, Freichel M, Gröne HJ, Most P, Müller OJ, Herzig S, Furlong EEM, Katus HA, and Backs J
- Subjects
- Animals, Epigenesis, Genetic, Gene Transfer Techniques, Heart Failure genetics, Histone Deacetylases genetics, Mice, Mice, Knockout, Myocardium enzymology, Nuclear Receptor Subfamily 4, Group A, Member 1 genetics, Nuclear Receptor Subfamily 4, Group A, Member 1 metabolism, Physical Conditioning, Animal, Proteolysis, Stromal Interaction Molecule 1 metabolism, Heart Failure metabolism, Hexosamines biosynthesis, Histone Deacetylases metabolism, Myocardial Contraction
- Abstract
The stress-responsive epigenetic repressor histone deacetylase 4 (HDAC4) regulates cardiac gene expression. Here we show that the levels of an N-terminal proteolytically derived fragment of HDAC4, termed HDAC4-NT, are lower in failing mouse hearts than in healthy control hearts. Virus-mediated transfer of the portion of the Hdac4 gene encoding HDAC4-NT into the mouse myocardium protected the heart from remodeling and failure; this was associated with decreased expression of Nr4a1, which encodes a nuclear orphan receptor, and decreased NR4A1-dependent activation of the hexosamine biosynthetic pathway (HBP). Conversely, exercise enhanced HDAC4-NT levels, and mice with a cardiomyocyte-specific deletion of Hdac4 show reduced exercise capacity, which was characterized by cardiac fatigue and increased expression of Nr4a1. Mechanistically, we found that NR4A1 negatively regulated contractile function in a manner that depended on the HBP and the calcium sensor STIM1. Our work describes a new regulatory axis in which epigenetic regulation of a metabolic pathway affects calcium handling. Activation of this axis during intermittent physiological stress promotes cardiac function, whereas its impairment in sustained pathological cardiac stress leads to heart failure.
- Published
- 2018
- Full Text
- View/download PDF
41. Atropine augments cardiac contractility by inhibiting cAMP-specific phosphodiesterase type 4.
- Author
-
Perera RK, Fischer TH, Wagner M, Dewenter M, Vettel C, Bork NI, Maier LS, Conti M, Wess J, El-Armouche A, Hasenfuß G, and Nikolaev VO
- Subjects
- Animals, Anti-Arrhythmia Agents administration & dosage, Atropine administration & dosage, Cyclic AMP metabolism, Fluorescence Resonance Energy Transfer, Humans, Mice, Mice, Knockout, Myocytes, Cardiac physiology, Phosphodiesterase 4 Inhibitors administration & dosage, Anti-Arrhythmia Agents pharmacology, Atropine pharmacology, Cyclic Nucleotide Phosphodiesterases, Type 4 metabolism, Myocytes, Cardiac drug effects, Phosphodiesterase 4 Inhibitors pharmacology
- Abstract
Atropine is a clinically relevant anticholinergic drug, which blocks inhibitory effects of the parasympathetic neurotransmitter acetylcholine on heart rate leading to tachycardia. However, many cardiac effects of atropine cannot be adequately explained solely by its antagonism at muscarinic receptors. In isolated mouse ventricular cardiomyocytes expressing a Förster resonance energy transfer (FRET)-based cAMP biosensor, we confirmed that atropine inhibited acetylcholine-induced decreases in cAMP. Unexpectedly, even in the absence of acetylcholine, after G-protein inactivation with pertussis toxin or in myocytes from M
2 - or M1/3 -muscarinic receptor knockout mice, atropine increased cAMP levels that were pre-elevated with the β-adrenergic agonist isoproterenol. Using the FRET approach and in vitro phosphodiesterase (PDE) activity assays, we show that atropine acts as an allosteric PDE type 4 (PDE4) inhibitor. In human atrial myocardium and in both intact wildtype and M2 or M1/3 -receptor knockout mouse Langendorff hearts, atropine led to increased contractility and heart rates, respectively. In vivo, the atropine-dependent prolongation of heart rate increase was blunted in PDE4D but not in wildtype or PDE4B knockout mice. We propose that inhibition of PDE4 by atropine accounts, at least in part, for the induction of tachycardia and the arrhythmogenic potency of this drug.- Published
- 2017
- Full Text
- View/download PDF
42. Calcium Signaling and Transcriptional Regulation in Cardiomyocytes.
- Author
-
Dewenter M, von der Lieth A, Katus HA, and Backs J
- Subjects
- Animals, Cardiovascular Agents therapeutic use, Cell Nucleus metabolism, Cytosol metabolism, Excitation Contraction Coupling, Gene Expression Regulation, Heart Diseases drug therapy, Heart Diseases physiopathology, Humans, Kinetics, Myocardial Contraction, Myocytes, Cardiac drug effects, Calcium Signaling drug effects, Heart Diseases genetics, Heart Diseases metabolism, Myocytes, Cardiac metabolism, Transcription, Genetic drug effects
- Abstract
Calcium (Ca
2+ ) is a universal regulator of various cellular functions. In cardiomyocytes, Ca2+ is the central element of excitation-contraction coupling, but also impacts diverse signaling cascades and influences the regulation of gene expression, referred to as excitation-transcription coupling. Disturbances in cellular Ca2+ -handling and alterations in Ca2+ -dependent gene expression patterns are pivotal characteristics of failing cardiomyocytes, with several excitation-transcription coupling pathways shown to be critically involved in structural and functional remodeling processes. Thus, targeting Ca2+ -dependent transcriptional pathways might offer broad therapeutic potential. In this article, we (1) review cytosolic and nuclear Ca2+ dynamics in cardiomyocytes with respect to their impact on Ca2+ -dependent signaling, (2) give an overview on Ca2+ -dependent transcriptional pathways in cardiomyocytes, and (3) discuss implications of excitation-transcription coupling in the diseased heart., (© 2017 American Heart Association, Inc.)- Published
- 2017
- Full Text
- View/download PDF
43. Chronic loss of inhibitor-1 diminishes cardiac RyR2 phosphorylation despite exaggerated CaMKII activity.
- Author
-
Neef S, Heijman J, Otte K, Dewenter M, Saadatmand AR, Meyer-Roxlau S, Antos CL, Backs J, Dobrev D, Wagner M, Maier LS, and El-Armouche A
- Subjects
- Adrenergic beta-1 Receptor Agonists, Animals, Dobutamine, Dogs, Echocardiography, Stress, Guanine Nucleotide Exchange Factors metabolism, Mice, Knockout, Myocytes, Cardiac metabolism, Phosphorylation, Protein Phosphatase 1 metabolism, Rats, Stress, Physiological, Calcium-Calmodulin-Dependent Protein Kinase Type 2 metabolism, Myocardium metabolism, Proteins genetics, Ryanodine Receptor Calcium Release Channel metabolism
- Abstract
Inhibitor-1 (I-1) modulates protein phosphatase 1 (PP1) activity and thereby counteracts the phosphorylation by kinases. I-1 is downregulated and deactivated in failing hearts, but whether its role is beneficial or detrimental remains controversial, and opposing therapeutic strategies have been proposed. Overactivity of Ca
2+ /calmodulin-dependent protein kinase II (CaMKII) with hyperphosphorylation of ryanodine receptors (RyR2) at the CaMKII-site is recognized to be central for heart failure and arrhythmias. Using an I-1-deficient mouse line as well as transfected cell lines, we investigated the effects of acute and chronic modulation of I-1 on CaMKII activity and RyR2 phosphorylation. We demonstrate that I-1 acutely modulates CaMKII by regulating PP1 activity. However, while ablation of I-1 should thus limit CaMKII-activation, we unexpectedly found exaggerated CaMKII-activation under β-adrenergic stress upon chronic loss of I-1 in knockout mice. We unraveled that this is due to chronic upregulation of the exchange protein activated by cAMP (EPAC) leading to augmented CaMKII activation, and using computational modeling validated that an increase in EPAC expression can indeed explain our experimental findings. Interestingly, at the level of RyR2, the increase in PP1 activity more than outweighed the increase in CaMKII activity, resulting in reduced RyR phosphorylation at Ser-2814. Exaggerated CaMKII activation due to counterregulatory mechanisms upon loss of I-1 is an important caveat with respect to suggested therapeutic I-1-inhibition, as CaMKII overactivity has been heavily implicated in several cardiac pathologies.- Published
- 2017
- Full Text
- View/download PDF
44. Calcium/Calmodulin-Dependent Protein Kinase II Activity Persists During Chronic β-Adrenoceptor Blockade in Experimental and Human Heart Failure.
- Author
-
Dewenter M, Neef S, Vettel C, Lämmle S, Beushausen C, Zelarayan LC, Katz S, von der Lieth A, Meyer-Roxlau S, Weber S, Wieland T, Sossalla S, Backs J, Brown JH, Maier LS, and El-Armouche A
- Subjects
- Adrenergic beta-Antagonists pharmacology, Animals, Disease Models, Animal, Echocardiography, Heart Failure diagnosis, Heart Failure drug therapy, Humans, Immunoblotting, Mice, Mice, Transgenic, Calcium-Calmodulin-Dependent Protein Kinase Type 2 metabolism, Heart Failure metabolism, Metoprolol pharmacology
- Abstract
Background: Considerable evidence suggests that calcium/calmodulin-dependent protein kinase II (CaMKII) overactivity plays a crucial role in the pathophysiology of heart failure (HF), a condition characterized by excessive β-adrenoceptor (β-AR) stimulation. Recent studies indicate a significant cross talk between β-AR signaling and CaMKII activation presenting CaMKII as a possible downstream mediator of detrimental β-AR signaling in HF. In this study, we investigated the effect of chronic β-AR blocker treatment on CaMKII activity in human and experimental HF., Methods and Results: Immunoblot analysis of myocardium from end-stage HF patients (n=12) and non-HF subjects undergoing cardiac surgery (n=12) treated with β-AR blockers revealed no difference in CaMKII activity when compared with non-β-AR blocker-treated patients. CaMKII activity was judged by analysis of CaMKII expression, autophosphorylation, and oxidation and by investigating the phosphorylation status of CaMKII downstream targets. To further evaluate these findings, CaMKIIδ
C transgenic mice were treated with the β1 -AR blocker metoprolol (270 mg/kg*d). Metoprolol significantly reduced transgene-associated mortality (n≥29; P <0.001), attenuated the development of cardiac hypertrophy (-14±6% heart weight/tibia length; P <0.05), and strongly reduced ventricular arrhythmias (-70±22% premature ventricular contractions; P <0.05). On a molecular level, metoprolol expectedly decreased protein kinase A-dependent phospholamban and ryanodine receptor 2 phosphorylation (-42±9% for P-phospholamban-S16 and -22±7% for P-ryanodine receptor 2-S2808; P <0.05). However, this was paralled neither by a reduction in CaMKII autophosphorylation, oxidation, and substrate binding nor a change in the phosphorylation of CaMKII downstream target proteins (n≥11). The lack of CaMKII modulation by β-AR blocker treatment was confirmed in healthy wild-type mice receiving metoprolol., Conclusions: Chronic β-AR blocker therapy in patients and in a mouse model of CaMKII-induced HF is not associated with a change in CaMKII activity. Thus, our data suggest that the molecular effects of β-AR blockers are not based on a modulation of CaMKII. Directly targeting CaMKII may, therefore, further improve HF therapy in addition to β-AR blockade., (© 2017 American Heart Association, Inc.)- Published
- 2017
- Full Text
- View/download PDF
45. Phosphodiesterase 2 Protects Against Catecholamine-Induced Arrhythmia and Preserves Contractile Function After Myocardial Infarction.
- Author
-
Vettel C, Lindner M, Dewenter M, Lorenz K, Schanbacher C, Riedel M, Lämmle S, Meinecke S, Mason FE, Sossalla S, Geerts A, Hoffmann M, Wunder F, Brunner FJ, Wieland T, Mehel H, Karam S, Lechêne P, Leroy J, Vandecasteele G, Wagner M, Fischmeister R, and El-Armouche A
- Subjects
- Animals, Arrhythmias, Cardiac chemically induced, Arrhythmias, Cardiac prevention & control, Catecholamines toxicity, Cyclic Nucleotide Phosphodiesterases, Type 2 antagonists & inhibitors, Dogs, Female, Imidazoles pharmacology, Male, Mice, Mice, Transgenic, Myocardial Contraction drug effects, Myocardial Infarction physiopathology, Triazines pharmacology, Arrhythmias, Cardiac metabolism, Cardiotonic Agents metabolism, Cyclic Nucleotide Phosphodiesterases, Type 2 biosynthesis, Isoproterenol toxicity, Myocardial Contraction physiology, Myocardial Infarction metabolism
- Abstract
Rationale: Phosphodiesterase 2 is a dual substrate esterase, which has the unique property to be stimulated by cGMP, but primarily hydrolyzes cAMP. Myocardial phosphodiesterase 2 is upregulated in human heart failure, but its role in the heart is unknown., Objective: To explore the role of phosphodiesterase 2 in cardiac function, propensity to arrhythmia, and myocardial infarction., Methods and Results: Pharmacological inhibition of phosphodiesterase 2 (BAY 60-7550, BAY) led to a significant positive chronotropic effect on top of maximal β-adrenoceptor activation in healthy mice. Under pathological conditions induced by chronic catecholamine infusions, BAY reversed both the attenuated β-adrenoceptor-mediated inotropy and chronotropy. Conversely, ECG telemetry in heart-specific phosphodiesterase 2-transgenic (TG) mice showed a marked reduction in resting and in maximal heart rate, whereas cardiac output was completely preserved because of greater cardiac contraction. This well-tolerated phenotype persisted in elderly TG with no indications of cardiac pathology or premature death. During arrhythmia provocation induced by catecholamine injections, TG animals were resistant to triggered ventricular arrhythmias. Accordingly, Ca
2+ -spark analysis in isolated TG cardiomyocytes revealed remarkably reduced Ca2+ leakage and lower basal phosphorylation levels of Ca2+ -cycling proteins including ryanodine receptor type 2. Moreover, TG demonstrated improved cardiac function after myocardial infarction., Conclusions: Endogenous phosphodiesterase 2 contributes to heart rate regulation. Greater phosphodiesterase 2 abundance protects against arrhythmias and improves contraction force after severe ischemic insult. Activating myocardial phosphodiesterase 2 may, thus, represent a novel intracellular antiadrenergic therapeutic strategy protecting the heart from arrhythmia and contractile dysfunction., (© 2016 American Heart Association, Inc.)- Published
- 2017
- Full Text
- View/download PDF
46. LATITUDE-TIMI: is there still hope for anti-inflammatory therapy in acute myocardial infaction?
- Author
-
Dewenter M, Wagner M, and El-Armouche A
- Abstract
Competing Interests: The authors have no conflicts of interest to declare.
- Published
- 2016
- Full Text
- View/download PDF
47. Physiologic force-frequency response in engineered heart muscle by electromechanical stimulation.
- Author
-
Godier-Furnémont AF, Tiburcy M, Wagner E, Dewenter M, Lämmle S, El-Armouche A, Lehnart SE, Vunjak-Novakovic G, and Zimmermann WH
- Subjects
- Animals, Calcium metabolism, Cells, Cultured, Electric Stimulation methods, Heart Rate, Mechanical Phenomena, Myocardial Contraction, Rats, Myocardium cytology, Myocytes, Cardiac cytology, Tissue Engineering methods
- Abstract
A hallmark of mature mammalian ventricular myocardium is a positive force-frequency relationship (FFR). Despite evidence of organotypic structural and molecular maturation, a positive FFR has not been observed in mammalian tissue engineered heart muscle. We hypothesized that concurrent mechanical and electrical stimulation at frequencies matching physiological heart rate will result in functional maturation. We investigated the role of biomimetic mechanical and electrical stimulation in functional maturation in engineered heart muscle (EHM). Following tissue consolidation, EHM were subjected to electrical field stimulation at 0, 2, 4, or 6 Hz for 5 days, while strained on flexible poles to facilitate auxotonic contractions. EHM stimulated at 2 and 4 Hz displayed a similarly enhanced inotropic reserve, but a clearly diverging FFR. The positive FFR in 4 Hz stimulated EHM was associated with reduced calcium sensitivity, frequency-dependent acceleration of relaxation, and enhanced post-rest potentiation. This was paralleled on the cellular level with improved calcium storage and release capacity of the sarcoplasmic reticulum and enhanced T-tubulation. We conclude that electro-mechanical stimulation at a physiological frequency supports functional maturation in mammalian EHM. The observed positive FFR in EHM has important implications for the applicability of EHM in cardiovascular research., (Copyright © 2015. Published by Elsevier Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
48. The Janus face of BNP therapy in chronic heart failure: beneficial effects unmasked by β blockers?
- Author
-
Dewenter M, Vettel C, and El-Armouche A
- Subjects
- Animals, Male, Adrenergic beta-Antagonists administration & dosage, Heart Failure complications, Natriuretic Peptide, Brain administration & dosage, Tachycardia, Ventricular drug therapy, Ventricular Remodeling drug effects
- Published
- 2014
- Full Text
- View/download PDF
49. [Tofacitinib].
- Author
-
Döker S, Dewenter M, and El-Armouche A
- Subjects
- Administration, Oral, Arthritis, Rheumatoid blood, Clinical Trials, Phase III as Topic, Drug Approval, Europe, Humans, Piperidines adverse effects, Piperidines pharmacokinetics, Protein Kinase Inhibitors adverse effects, Protein Kinase Inhibitors pharmacokinetics, Pyrimidines adverse effects, Pyrimidines pharmacokinetics, Pyrroles adverse effects, Pyrroles pharmacokinetics, Treatment Outcome, Arthritis, Rheumatoid drug therapy, Piperidines therapeutic use, Protein Kinase Inhibitors therapeutic use, Pyrimidines therapeutic use, Pyrroles therapeutic use
- Abstract
Tofacitinib is the first Janus kinase inhibitor which was approved for the therapy of rheumatoid arthritis in the USA. Several phase III studies proved the efficacy of Tofacitinib as monotherapy or in combination with established medication. This article discusses the therapeutic potential of this new pharmacological approach and the current data on efficacy and safety of Tofacitinib therapy with special emphasis on a prospective approval in the EU., (© Georg Thieme Verlag KG Stuttgart · New York.)
- Published
- 2014
- Full Text
- View/download PDF
50. PDE2-mediated cAMP hydrolysis accelerates cardiac fibroblast to myofibroblast conversion and is antagonized by exogenous activation of cGMP signaling pathways.
- Author
-
Vettel C, Lämmle S, Ewens S, Cervirgen C, Emons J, Ongherth A, Dewenter M, Lindner D, Westermann D, Nikolaev VO, Lutz S, Zimmermann WH, and El-Armouche A
- Subjects
- Animals, Animals, Newborn, Atrial Natriuretic Factor pharmacology, Cells, Cultured, Cyclic Nucleotide Phosphodiesterases, Type 2 genetics, Fibroblasts drug effects, Fibroblasts enzymology, Fibroblasts physiology, Gene Expression, Hydrolysis, Myocytes, Cardiac enzymology, Nitric Oxide Donors pharmacology, Nitroprusside pharmacology, Rats, Receptors, Adrenergic, beta physiology, Cyclic AMP metabolism, Cyclic GMP physiology, Cyclic Nucleotide Phosphodiesterases, Type 2 physiology, Myocardium cytology, Myofibroblasts physiology, Signal Transduction physiology
- Abstract
Recent studies suggest that the signal molecules cAMP and cGMP have antifibrotic effects by negatively regulating pathways associated with fibroblast to myofibroblast (MyoCF) conversion. The phosphodiesterase 2 (PDE2) has the unique property to be stimulated by cGMP, which leads to a remarkable increase in cAMP hydrolysis and thus mediates a negative cross-talk between both pathways. PDE2 has been recently investigated in cardiomyocytes; here we specifically addressed its role in fibroblast conversion and cardiac fibrosis. PDE2 is abundantly expressed in both neonatal rat cardiac fibroblasts (CFs) and cardiomyocytes. The overexpression of PDE2 in CFs strongly reduced basal and isoprenaline-induced cAMP synthesis, and this decrease was sufficient to induce MyoCF conversion even in the absence of exogenous profibrotic stimuli. Functional stress-strain experiments with fibroblast-derived engineered connective tissue (ECT) demonstrated higher stiffness in ECTs overexpressing PDE2. In regard to cGMP, neither basal nor atrial natriuretic peptide-induced cGMP levels were affected by PDE2, whereas the response to nitric oxide donor sodium nitroprusside was slightly but significantly reduced. Interestingly, despite persistently depressed cAMP levels, both cGMP-elevating stimuli were able to completely prevent the PDE2-induced MyoCF phenotype, arguing for a double-tracked mechanism. In conclusion, PDE2 accelerates CF to MyoCF conversion, which leads to greater stiffness in ECTs. Atrial natriuretic peptide- and sodium nitroprusside-mediated cGMP synthesis completely reverses PDE2-induced fibroblast conversion. Thus PDE2 may augment cardiac remodeling, but this effect can also be overcome by enhanced cGMP. The redundant role of cAMP and cGMP as antifibrotic meditators may be viewed as a protective mechanism in heart failure.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.