96 results on '"Schroen B"'
Search Results
2. The microRNA-146a - DLST axis: a new therapeutic target in heart failure.: 736
- Author
-
Ward Heggermont, W A, Papageorgiou, A P, Quaegebeur, A, Deckx, S, Carai, P, Schroen, B, Van Bilsen, M, Pinto, Y, Carmeliet, P, and Heymans, S
- Published
- 2016
3. RNA Profiling in Human and Murine Transplanted Hearts: Identification and Validation of Therapeutic Targets for Acute Cardiac and Renal Allograft Rejection
- Author
-
Van Aelst, L. N. L., Summer, G., Li, S., Gupta, S. K., Heggermont, W., De Vusser, K., Carai, P., Naesens, M., Van Cleemput, J., Van de Werf, F., Vanhaecke, J., Thum, T., Waer, M., Papageorgiou, A.-P., Schroen, B., and Heymans, S.
- Published
- 2016
- Full Text
- View/download PDF
4. Genomic instability in the naturally and prematurely aged myocardium. Proceedings of the National Academy of Sciences of the United States of America
- Author
-
De Majo, F., Martens, L., Hegenbarth, J. C., Rühle, F., Hamczyk, M. R., Nevado, R. M., Andrés, V., Hilbold, E., Bär, C., Thum, T., de Boer, M., Duncker, D. J., Schroen, B., Armand, A. S., Stoll, M., & De Windt, L. J.
- Published
- 2021
- Full Text
- View/download PDF
5. 545MiR-139 expression is detrimental during pressure overload-induced heart failure
- Author
-
Schroen, B, Peters, T, Verhesen, W, Derks, W, Zentlini, L, Zacchigna, S, Giacca, M, Van Der Velden, J, De Windt, L, and Heymans, S
- Published
- 2014
- Full Text
- View/download PDF
6. P69The microRNA-221/222 family is differentially regulated in cardiac disease and counteracts pressure overload-induced cardiac remodeling in mice
- Author
-
Peters, T, Bijnen, M, Rech, M, Van Leeuwen, R, Derks, W, De Windt, LJ, Heymans, S, and Schroen, B
- Published
- 2014
- Full Text
- View/download PDF
7. Dichotomy between the transcriptomic landscape of naturally versus accelerated aged murine hearts
- Author
-
De Majo, F. (Federica), Hegenbarth, J.-C. (Jana-Charlotte), Rühle, F. (Frank), Bär, C. (Christian), Thum, T. (Thomas), Boer, M. (Martine) de, Duncker, D.J.G.M. (Dirk), Schroen, B. (Blanche), Armand, A.S. (Anne-Sophie), Stoll, M. (Monika), Windt, L.J. (Leon) de, De Majo, F. (Federica), Hegenbarth, J.-C. (Jana-Charlotte), Rühle, F. (Frank), Bär, C. (Christian), Thum, T. (Thomas), Boer, M. (Martine) de, Duncker, D.J.G.M. (Dirk), Schroen, B. (Blanche), Armand, A.S. (Anne-Sophie), Stoll, M. (Monika), and Windt, L.J. (Leon) de
- Abstract
We investigated the transcriptomic landscape of the murine myocardium along the course of natural aging and in three distinct mouse models of premature aging with established aging-related cardiac dysfunction. Genome-wide total RNA-seq was performed and the expression patterns of protein-coding genes and non-coding RNAs were compared between hearts from naturally aging mice, mice with cardiac-specific deficiency of a component of the DNA repair machinery, mice with reduced mitochondrial antioxidant capacity and mice with reduced telomere length. Our results demonstrate that no dramatic changes are evident in the transcriptomes of naturally senescent murine hearts until two years of age, in contrast to the transcriptome of accelerated aged mice. Additionally, these mice displayed model-specific alterations of the expression levels of protein-coding and non-coding genes with hardly any overlap with age-related signatures. Our data demonstrate very limited similarities between the transcriptomes of all our murine aging models and question their reliability to study human cardiovascular senescence.
- Published
- 2020
- Full Text
- View/download PDF
8. Dichotomy between the transcriptomic landscape of naturally versus accelerated aged murine hearts
- Author
-
De Majo, F, Hegenbarth, JC, Ruhle, F, Bar, C, Thum, T, Boer, Meta, Duncker, Dirk-jan, Schroen, B, Armand, AS, Stoll, M, De Windt, LJ, De Majo, F, Hegenbarth, JC, Ruhle, F, Bar, C, Thum, T, Boer, Meta, Duncker, Dirk-jan, Schroen, B, Armand, AS, Stoll, M, and De Windt, LJ
- Published
- 2020
9. 253Absence of microRNA-155 protects against adverse cardiac inflammation and hypertrophy during pressure overload and prevents heart failure
- Author
-
Schroen, B., Corsten, M., Verhesen, W., De Windt, L., Pinto, Y.M., Zacchigna, S., Thum, T., Carmeliet, P., Papageorgiou, A., and Heymans, S.
- Published
- 2012
10. P97MicroRNA-155 mediates cardiac inflammation and injury in acute viral myocarditis
- Author
-
Corsten, M.F., Verhesen, W., Papageorgiou, A.P., Carai, P., Lindow, M., Obad, S., Summer, G., De Rijck, L., Coort, S., Hazebroek, M., Hazebroek, M., Van Leeuwen, R., Gijbels, M., De Winther, M.P.J., Stassen, F.R.M., Kauppinen, S., Schroen, B., and Heymans, S.
- Published
- 2012
11. Peer Review #2 of "Sex differences in the response to angiotensin II receptor blockade in a rat model of eccentric cardiac hypertrophy (v0.1)"
- Author
-
Schroen, B, additional
- Published
- 2019
- Full Text
- View/download PDF
12. 397tRNA fragments are novel obesity-regulated components of the small cardiac RNAome
- Author
-
Liapi, E, primary, Verhesen, W, additional, Derks, K, additional, Van Leeuwen, REW, additional, and Schroen, B, additional
- Published
- 2018
- Full Text
- View/download PDF
13. P68Cardiomyocyte cell cycle activity and function is under the control of miR-125a
- Author
-
Verjans, R, primary, Braga, L, additional, Rech, M, additional, Beijnsberger, S, additional, Carai, P, additional, Spatjens, R, additional, Van Bilsen, M, additional, Giacca, M, additional, and Schroen, B, additional
- Published
- 2018
- Full Text
- View/download PDF
14. Assessing fatty acid oxidation flux in rodent cardiomyocyte models
- Author
-
Rech, M., primary, Luiken, J. J. F. P., additional, Glatz, J. F. C., additional, van Bilsen, M., additional, Schroen, B., additional, and Nabben, M., additional
- Published
- 2018
- Full Text
- View/download PDF
15. MicroRNA-18 and microRNA-19 regulate CTGF and TSP-1 expression in age-related heart failure
- Author
-
Almen, G.C. van, Verhesen, W., Leeuwen, R.E. van, Vrie, M. van de, Eurlings, C., Schellings, M.W., Swinnen, M., Cleutjens, J., Zandvoort, M.A. van, Heymans, S., Schroen, B., RS: CARIM School for Cardiovascular Diseases, Cardiologie, Pathologie, and Biomedische Technologie
- Subjects
Adult ,Male ,Aging ,Biopsy ,Thrombospondin 1 ,Mice ,connective tissue growth factor ,Animals ,Humans ,Myocytes, Cardiac ,thrombospondin-1 ,GeneralLiterature_REFERENCE(e.g.,dictionaries,encyclopedias,glossaries) ,Aged ,Heart Failure ,miR-18 ,microRNA ,Gene Expression Regulation, Developmental ,Heart ,Original Articles ,Middle Aged ,miR-19 ,Fibrosis ,Extracellular Matrix ,Rats ,matricellular proteins ,Mice, Inbred C57BL ,MicroRNAs ,Rats, Inbred Lew ,Multigene Family ,Collagen ,Immune Regulation Auto-immunity, transplantation and immunotherapy [NCMLS 2] ,cardiac aging - Abstract
To understand the process of cardiac aging, it is of crucial importance to gain insight into the age-related changes in gene expression in the senescent failing heart. Age-related cardiac remodeling is known to be accompanied by changes in extracellular matrix (ECM) gene and protein levels. Small noncoding microRNAs regulate gene expression in cardiac development and disease and have been implicated in the aging process and in the regulation of ECM proteins. However, their role in age-related cardiac remodeling and heart failure is unknown. In this study, we investigated the aging-associated microRNA cluster 17-92, which targets the ECM proteins connective tissue growth factor (CTGF) and thrombospondin-1 (TSP-1). We employed aged mice with a failure-resistant (C57Bl6) and failure-prone (C57Bl6 x 129Sv) genetic background and extrapolated our findings to human age-associated heart failure. In aging-associated heart failure, we linked an aging-induced increase in the ECM proteins CTGF and TSP-1 to a decreased expression of their targeting microRNAs 18a, 19a, and 19b, all members of the miR-17-92 cluster. Failure-resistant mice showed an opposite expression pattern for both the ECM proteins and the microRNAs. We showed that these expression changes are specific for cardiomyocytes and are absent in cardiac fibroblasts. In cardiomyocytes, modulation of miR-18/19 changes the levels of ECM proteins CTGF and TSP-1 and collagens type 1 and 3. Together, our data support a role for cardiomyocyte-derived miR-18/19 during cardiac aging, in the fine-tuning of cardiac ECM protein levels. During aging, decreased miR-18/19 and increased CTGF and TSP-1 levels identify the failure-prone heart.
- Published
- 2011
16. Absence of thrombospondin-2 causes age-related dilated cardiomyopathy
- Author
-
Swinnen, M., Vanhoutte, D., Van Almen, G., Hamdani, N., Schellings, M., D'hooge, J., Van Der Velden, J., Weaver, M., Sage, E., Bornstein, P., Verheyen, F., Chuah, Marinee, Westermann, D., Van De Werf, F., Schroen, B., Carmeliet, P., Pinto, Y., Heymans, S., VandenDriessche, Thierry, Cell Biology and Histology, and Division of Gene Therapy & Regenerative Medicine
- Subjects
Male ,Mice, Knockout ,Myocardium/pathology ,endocrine system ,Cardiomyopathy, Dilated/pathology ,mice ,Myocarditis/etiology ,Myocardium/metabolism ,Thrombospondins/genetics ,fibrosis ,Gene Transfer Techniques ,Matrix Metalloproteinase 2/metabolism ,virus diseases ,Cardiomyopathy, Dilated/etiology ,Myocytes, Cardiac/metabolism ,UP-REGULATION ,Enzyme Activation ,cell death ,Thrombospondins/deficiency ,immune system diseases ,Cardiomyopathy, Dilated/mortality ,Proto-Oncogene Proteins c-akt/metabolism ,Female ,Myocytes, Cardiac/ultrastructure ,Cardiomyopathy, Dilated/prevention & control - Abstract
BACKGROUND: The progressive shift from a young to an aged heart is characterized by alterations in the cardiac matrix. The present study investigated whether the matricellular protein thrombospondin-2 (TSP-2) may affect cardiac dimensions and function with physiological aging of the heart. METHODS AND RESULTS: TSP-2 knockout (KO) and wild-type mice were followed up to an age of 60 weeks. Survival rate, cardiac function, and morphology did not differ at a young age in TSP-2 KO compared with wild-type mice. However, >55% of the TSP-2 KO mice died between 24 and 60 weeks of age, whereas CONCLUSIONS: TSP-2 expression in the heart protects against age-dependent dilated cardiomyopathy.
- Published
- 2009
17. Genome-wide profiling of the cardiac transcriptome after myocardial infarction identifies novel heart-specific long non-coding RNAs
- Author
-
Ounzain, S., primary, Micheletti, R., additional, Beckmann, T., additional, Schroen, B., additional, Alexanian, M., additional, Pezzuto, I., additional, Crippa, S., additional, Nemir, M., additional, Sarre, A., additional, Johnson, R., additional, Dauvillier, J., additional, Burdet, F., additional, Ibberson, M., additional, Guigo, R., additional, Xenarios, I., additional, Heymans, S., additional, and Pedrazzini, T., additional
- Published
- 2014
- Full Text
- View/download PDF
18. MicroRNA involvement in immune activation during heart failure.
- Author
-
Vrie, M. van de, Heymans, S., Schroen, B., Vrie, M. van de, Heymans, S., and Schroen, B.
- Abstract
Item does not contain fulltext
- Published
- 2011
19. The microRNA 221/222 cluster controls CVB3-induced myocarditis: could tiny microRNAs explain adverse inflammation in the heart?
- Author
-
Heggermont, W. A., primary, Corsten, M. F., additional, Papageorgiou, A. P., additional, Thibaut, H. J., additional, Verhesen, W., additional, Carai, P., additional, Van Leeuwen, R., additional, Neyts, J., additional, Schroen, B., additional, and Heymans, S., additional
- Published
- 2013
- Full Text
- View/download PDF
20. Oral abstract presentations & Young Investigators Competition
- Author
-
Leone, A., primary, Aquila, I., additional, Vicinanza, C., additional, Iaconetti, C., additional, Bochicchio, A., additional, Ottolenghi, S., additional, Indolfi, C., additional, Nadal-Ginard, B., additional, Ellison, G. M., additional, Torella, D., additional, Mias, C., additional, Genet, G., additional, Guilbeau-Frugier, C., additional, Pathak, A., additional, Senard, J. M., additional, Gales, C., additional, Egorova, A. D., additional, Khedoe, P. S. J., additional, Goumans, M. T. H., additional, Nauli, S. M., additional, Ten Dijke, P., additional, Poelmann, R. E., additional, Hierck, B. P., additional, Miragoli, M., additional, Lab, M. J., additional, Singh, A., additional, Sikkel, M., additional, Lyon, A., additional, Gorelik, J., additional, Cheung, C., additional, Bernardo, A. S., additional, Trotter, M. W., additional, Pedersen, R. A., additional, Sinha, S., additional, Mioulane, M., additional, Foldes, G., additional, Harding, S. E., additional, Reglin, B., additional, Secomb, T. W., additional, Pries, A. R., additional, Buckingham, M., additional, Lescroart, F., additional, Meilhac, S., additional, Le Garrec, J.-F., additional, Rozmaritsa, N., additional, Christ, T., additional, Wettwer, E., additional, Knaut, M., additional, Ravens, U., additional, Tokar, S., additional, Schobesberger, S., additional, Wright, P. T., additional, Lyon, A. R., additional, Van Mil, A., additional, Grundmann, S., additional, Goumans, M.-J., additional, Jaksani, S., additional, Doevendans, P. A., additional, Sluijter, J. P., additional, Tijsen, A. J., additional, Amin, A. S., additional, Giudicessi, J. R., additional, Tanck, M. W., additional, Bezzina, C. R., additional, Creemers, E. E., additional, Wilde, A. M., additional, Ackerman, M. J., additional, Pinto, Y. M., additional, Gedicke-Hornung, C., additional, Behrens-Gawlik, V., additional, Khajetoorians, D., additional, Mearini, G., additional, Reischmann, S., additional, Geertz, B., additional, Voit, T., additional, Dreyfus, P., additional, Eschenhagen, T., additional, Carrier, L., additional, Duerr, G. D., additional, Heinemann, J. C., additional, Wenzel, D., additional, Ghanem, A., additional, Alferink, J. C., additional, Zimmer, A., additional, Lutz, B., additional, Welz, A., additional, Fleischmann, B. K., additional, Dewald, O., additional, Sbroggio', M., additional, Bertero, A., additional, Giuliano, L., additional, Brancaccio, M., additional, Tarone, G., additional, Meiser, M., additional, Kohlhaas, M., additional, Chen, Y., additional, Csordas, G., additional, Dorn, G., additional, Maack, C., additional, Stapel, B., additional, Hoch, M., additional, Haghikia, A., additional, Fischer, P., additional, Hilfiker-Kleiner, D., additional, Schroen, B., additional, Corsten, M., additional, Verhesen, W., additional, De Windt, L., additional, Zacchigna, S., additional, Thum, T., additional, Carmeliet, P., additional, Papageorgiou, A., additional, Heymans, S., additional, Lunde, I. G., additional, Finsen, A. V., additional, Florholmen, G., additional, Skrbic, B., additional, Kvaloy, H., additional, Jarstadmarken, H. O., additional, Sjaastad, I., additional, Tonnessen, T., additional, Carlson, C. R., additional, Christensen, G., additional, Paavola, J., additional, Schliffke, S., additional, Rossetti, S., additional, Kuo, I., additional, Yuan, S., additional, Sun, Z., additional, Harris, P., additional, Torres, V., additional, Ehrlich, B., additional, Robinson, P., additional, Adams, K., additional, Zhang, Y.-H., additional, Casadei, B., additional, Watkins, H., additional, Redwood, C., additional, Seneviratne, A. N., additional, Cole, J. E., additional, Goddard, M. E., additional, Mohri, Z., additional, Cross, A. J., additional, Krams, R., additional, Monaco, C., additional, Everaert, B. R., additional, Van Laere, S. J., additional, Hoymans, V. Y., additional, Timmermans, J. P., additional, and Vrints, C. J., additional
- Published
- 2012
- Full Text
- View/download PDF
21. Small but smart--microRNAs in the centre of inflammatory processes during cardiovascular diseases, the metabolic syndrome, and ageing
- Author
-
Schroen, B., primary and Heymans, S., additional
- Published
- 2011
- Full Text
- View/download PDF
22. Biologically relevant effects of mRNA amplification on gene expression profiles
- Author
-
Smits Jos FM, Smeets Hubert JM, Debets Jacques JM, van Erk Arie, Janssen Ben JA, Schroen Blanche, van Haaften Rachel IM, van den Wijngaard Arthur, Pinto Yigal M, and Evelo Chris TA
- Subjects
Computer applications to medicine. Medical informatics ,R858-859.7 ,Biology (General) ,QH301-705.5 - Abstract
Abstract Background Gene expression microarray technology permits the analysis of global gene expression profiles. The amount of sample needed limits the use of small excision biopsies and/or needle biopsies from human or animal tissues. Linear amplification techniques have been developed to increase the amount of sample derived cDNA. These amplified samples can be hybridised on microarrays. However, little information is available whether microarrays based on amplified and unamplified material yield comparable results. In the present study we compared microarray data obtained from amplified mRNA derived from biopsies of rat cardiac left ventricle and non-amplified mRNA derived from the same organ. Biopsies were linearly amplified to acquire enough material for a microarray experiment. Both amplified and unamplified samples were hybridized to the Rat Expression Set 230 Array of Affymetrix. Results Analysis of the microarray data showed that unamplified material of two different left ventricles had 99.6% identical gene expression. Gene expression patterns of two biopsies obtained from the same parental organ were 96.3% identical. Similarly, gene expression pattern of two biopsies from dissimilar organs were 92.8% identical to each other. Twenty-one percent of reporters called present in parental left ventricular tissue disappeared after amplification in the biopsies. Those reporters were predominantly seen in the low intensity range. Sequence analysis showed that reporters that disappeared after amplification had a GC-content of 53.7+/-4.0%, while reporters called present in biopsy- and whole LV-samples had an average GC content of 47.8+/-5.5% (P Conclusion This study establishes that the gene expression profile obtained after amplification of mRNA of left ventricular biopsies is representative for the whole left ventricle of the rat heart. However, specific gene transcripts present in parental tissues were undetectable in the minute left ventricular biopsies. Transcripts that were lost due to the amplification process were not randomly distributed, but had higher GC-content and hairpins in the sequence and were mainly found in the lower intensity range which includes many transcription factors from specific signalling pathways.
- Published
- 2006
- Full Text
- View/download PDF
23. 545 MiR-139 expression is detrimental during pressure overload-induced heart failure.
- Author
-
Schroen, B, Peters, T, Verhesen, W, Derks, W, Zentlini, L, Zacchigna, S, Giacca, M, Van Der Velden, J, De Windt, L, and Heymans, S
- Subjects
- *
HEART failure , *MICRORNA , *GENE expression , *HEART cells , *PROTEIN kinases , *PHOSPHODIESTERASES , *HYPERTROPHY - Abstract
Cardiac hypertrophy and consequent contractile dysfunction continue to burden Western society. Cardiomyocyte cyclic AMP (cAMP) and calcium are driving forces behind cardiomyocyte contraction. Distortion of their balance may induce HF, but specific therapies aiming at restoring physiological cAMP/calcium signaling are lacking.We recently identified microRNA-139 (miR-139) to be downregulated in failing human hearts. MiR-139 resides in the phosphodiesterase gene PDE2A and is predicted to target several phosphodiesterase messengers. In view of the central role of phosphodiesterases in controlling cardiac cAMP and calcium signaling, we hypothesized that miR-139 may affect HF progression by fine-tuning cAMP and calcium balances.Adeno-associated virus serotype 9 (AAV9), either empty control or expressing pre-miR-139, was administered to male C57Bl/6J mice. After allowing transgene expression for 3 weeks, mice were subjected to sham treatment or 4 weeks of pressure overload by subcutaneous Angiotensin II infusion (AngII, 2,5 mg/(kg·d). MiR-139 overexpression mildly aggravated HF development upon AngII with echocardiographically measured fractional shortening decreasing by 24±7% in AAV9-control AngII and by 46±5% in AAV9-pre-miR-139 AngII (n>11/group; p=0.14). In AAV9-control mice, AngII infusion led to concentric hypertrophy with an increased relative wall thickness (RWT) of 35±6%, whereas mice overexpressing miR-139 showed a rather eccentric form of hypertrophy with an increased RWT of 14±7% (p<0.05). The fraction of unphosphorylated cardiac troponin I (cTnI), a substrate of the cAMP dependent protein kinase A (PKA), tended to increase upon AngII infusion only in mice overexpressing miR-139 (n=4/group; p=0.11), indicating a reduced relaxation rate due to increased calcium sensitivity, a feature commonly observed in HF. Complimentary to these data, in vivo knockdown of mir-139 by cholesterol-tagged antagomiRs (20mg/kg) on three consecutive days before start of AngII infusion dampened the development of pressure overload-induced cardiac hypertrophy (increase in HW/TL: ctrl: 48±10%, n=4; antagomiR: 25±7%, n=7; p=0.16).In conclusion, cardiac downregulation of miR-139 upon pressure overload is a protective response to preserve cardiac function. AAV9-mediated overexpression of miR-139 promotes cardiac dilation and predisposes to HF. Upcoming experiments will aim at defining the molecular mechanism by which miR-139/phosphodiesterase signaling affects cardiac pathophysiology. [ABSTRACT FROM PUBLISHER]
- Published
- 2014
- Full Text
- View/download PDF
24. P69 The microRNA-221/222 family is differentially regulated in cardiac disease and counteracts pressure overload-induced cardiac remodeling in mice.
- Author
-
Peters, T, Bijnen, M, Rech, M, Van Leeuwen, R, Derks, W, De Windt, LJ, Heymans, S, and Schroen, B
- Subjects
MICRORNA ,VENTRICULAR remodeling ,HEART failure ,HEART diseases ,THERAPEUTICS ,MEDICAL innovations ,LABORATORY mice - Abstract
Purpose: Despite major advances in the treatment of cardiovascular diseases, heart failure (HF) remains one of the top causes of death world-wide. The implications of microRNAs in this process are well accepted but still only incompletely understood. The microRNAs 221-3p and 222-3p are processed from a common precursor and share the same seed sequence and thus form the microRNA-221/222 family (miR-221/222). Both microRNAs were found to be involved in myoblast differentiation and are upregulated after aortic banding in mice. We therefore hypothesized that the miR-221/222 family is involved in the pathophysiology of cardiac hypertrophy and failure upon pressure overload.Methods and results: In a genome wide screen for microRNAs regulated in human dilated cardiomyopathy, we found miR-222 levels to be significantly decreased (p<0.01). MiR-221/222 were also downregulated in neonatal rat cardiomyocytes (nRCMs) upon stimulation with the pro-hypertrophic compound phenylephrine (PE) (p<0.05). Interestingly, the overexpression of these miRs in nRCMs using mimics significantly blunted the induction of the hypertrophy markers Bnp and skeletal alpha actin (Acta1) in nRCMs upon stimulation with PE.To investigate the role of miR-221/222 in pressure overload-induced heart failure, we simultaneously injected anti-miR-221 and anti-miR-222 antisense oligonucleotides (ASOs) or scrambled control oligonucleotides (SCOs) in male C57BL/6 mice 3 days before implanting angiotensin II-filled osmotic minipumps (AngII, 2.5 mg/(kg d)). After 4 weeks, we assessed cardiac function and histology as well as molecular changes in the left ventricle. Surprisingly, we did not find an effect of miR-221/222 inhibition on overall cardiac hypertrophy after AngII infusion (HW/TL: 8.04 vs 7.92 mg/mm, p>0.05). However, interstitial fibrosis was significantly increased upon AngII stimulation in mice that received miR-221/222 ASOs as compared to SCOs (6.1 vs 3.7% LV area, p<0.05). On the mRNA level, these mice also showed a 2.9-fold higher induction of Anp upon AngII stimulation (p~0.10), in line with anti-hypertrophic effects of miR-221/222 mimics shown in vitro.Conclusions: Taken together, our results indicate a protective effect of the microRNA-221/222 family in the stressed heart. Inhibition of miR-221/222 prior to pressure overload in mice led to increased fibrosis indicating adverse remodeling. In vitro, a direct effect of miR-221/222 overexpression on the hypertrophic response of nRCMs could be shown. Further experiments will aim at identifying the function of the miR-221/222 family both in cardiac fibroblast and cardiomyocytes. [ABSTRACT FROM PUBLISHER]
- Published
- 2014
- Full Text
- View/download PDF
25. Commonalities of platelet dysfunction in heart failure with preserved ejection fraction and underlying comorbidities.
- Author
-
D'Italia G, Schroen B, and Cosemans JMEM
- Abstract
Heart failure with preserved ejection fraction (HFpEF) is characterized by a lack of a specific targeted treatment and a complex, partially unexplored pathophysiology. Common comorbidities associated with HFpEF are hypertension, atrial fibrillation, obesity and diabetes. These comorbidities, combined with advanced age, play a crucial role in the initiation and development of the disease through the promotion of systemic inflammation and consequent changes in cardiac phenotype. In this context, we suggest platelets as important players due to their emerging role in vascular inflammation. This review provides an overview of the role of platelets in HFpEF and its associated comorbidities, including hypertension, atrial fibrillation, obesity and diabetes mellitus, as well as the impact of age and sex on platelet function. These major HFpEF-associated comorbidities present alterations in platelet behaviour and in features linked to platelet size, content and reactivity. The resulting dysfunctional platelets can contribute to further increase inflammation, oxidative stress and endothelial dysfunction, suggesting an active role of these cells in the initiation and progression of HFpEF. Recent evidence shows that reduced platelet count and elevated mean platelet volume are associated with worsening heart failure in HFpEF patients. However, the specific mechanisms by which platelets contribute to HFpEF development and progression are still largely unexplored, with only a few studies investigating platelet function in HFpEF. We discuss the limited yet significant body of research investigating platelet function in HFpEF, emphasizing the need for more comprehensive studies. Additionally, we explore the potential mechanisms through which platelets may influence HFpEF, such as their interactions with the vascular endothelium and the secretion of bioactive molecules like cytokines, chemokines and RNA molecules. These interactions and secretions may play a role in modulating vascular inflammation and contributing to the pathophysiological landscape of HFpEF. The review underscores the necessity for future research to elucidate the precise contributions of platelets to HFpEF, aiming to potentially identify novel therapeutic targets and improve patient outcomes. The evidence presented herein supports the hypothesis that platelets are not merely passive bystanders but active participants in the pathophysiology of HFpEF and its comorbidities., (© 2024 The Author(s). ESC Heart Failure published by John Wiley & Sons Ltd on behalf of European Society of Cardiology.)
- Published
- 2024
- Full Text
- View/download PDF
26. Guidelines for mitochondrial RNA analysis.
- Author
-
Jusic A, Erpapazoglou Z, Dalgaard LT, Lakkisto P, de Gonzalo-Calvo D, Benczik B, Ágg B, Ferdinandy P, Fiedorowicz K, Schroen B, Lazou A, and Devaux Y
- Abstract
Mitochondria are the energy-producing organelles of mammalian cells with critical involvement in metabolism and signaling. Studying their regulation in pathological conditions may lead to the discovery of novel drugs to treat, for instance, cardiovascular or neurological diseases, which affect high-energy-consuming cells such as cardiomyocytes, hepatocytes, or neurons. Mitochondria possess both protein-coding and noncoding RNAs, such as microRNAs, long noncoding RNAs, circular RNAs, and piwi-interacting RNAs, encoded by the mitochondria or the nuclear genome. Mitochondrial RNAs are involved in anterograde-retrograde communication between the nucleus and mitochondria and play an important role in physiological and pathological conditions. Despite accumulating evidence on the presence and biogenesis of mitochondrial RNAs, their study continues to pose significant challenges. Currently, there are no standardized protocols and guidelines to conduct deep functional characterization and expression profiling of mitochondrial RNAs. To overcome major obstacles in this emerging field, the EU-CardioRNA and AtheroNET COST Action networks summarize currently available techniques and emphasize critical points that may constitute sources of variability and explain discrepancies between published results. Standardized methods and adherence to guidelines to quantify and study mitochondrial RNAs in normal and disease states will improve research outputs, their reproducibility, and translation potential to clinical application., Competing Interests: A.J. is employed by HAYA Therapeutics SA, Switzerland. Y.D. holds patents related to diagnostic and therapeutic applications of RNAs and is member of the Scientific Advisory Board of Firalis SA. P.F. is the founder and CEO of Pharmahungary Group, a group of research and development (R&D) companies (www.pharmahungary.com). B.Á. is employed by Pharmahungary Group., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
27. Plasma Extracellular Vesicles as Liquid Biopsy to Unravel the Molecular Mechanisms of Cardiac Reverse Remodeling Following Resynchronization Therapy?
- Author
-
van Nieuwenhoven FA, Schroen B, Barile L, van Middendorp L, Prinzen FW, and Auricchio A
- Abstract
Cardiac resynchronization therapy (CRT) has become a valuable addition to the treatment options for heart failure, in particular for patients with disturbances in electrical conduction that lead to regionally different contraction patterns (dyssynchrony). Dyssynchronous hearts show extensive molecular and cellular remodeling, which has primarily been investigated in experimental animals. Evidence showing that at least several miRNAs play a role in this remodeling is increasing. A comparison of results from measurements in plasma and myocardial tissue suggests that plasma levels of miRNAs may reflect the expression of these miRNAs in the heart. Because many miRNAs released in the plasma are included in extracellular vesicles (EVs), which protect them from degradation, measurement of myocardium-derived miRNAs in peripheral blood EVs may open new avenues to investigate and monitor (reverse) remodeling in dyssynchronous and resynchronized hearts of patients.
- Published
- 2023
- Full Text
- View/download PDF
28. High-Sensitivity Troponin-T and Cardiovascular Outcomes in the Community: Differences Between Women and Men.
- Author
-
Suthahar N, Meems LMG, van Veldhuisen DJ, Walter JE, Gansevoort RT, Heymans S, Schroen B, van der Harst P, Kootstra-Ros JE, van Empel V, Mueller C, Bakker SJL, and de Boer RA
- Subjects
- Cardiovascular Diseases blood, Cardiovascular Diseases mortality, Female, Heart Failure mortality, Humans, Incidence, Independent Living statistics & numerical data, Male, Middle Aged, Mortality, Proportional Hazards Models, Prospective Studies, Sex Factors, Heart Failure blood, Troponin T blood
- Abstract
Objective: To evaluate associations of high-sensitivity cardiac troponin-T (cTnT) with cardiovascular disease (CVD), heart failure (HF), and mortality in community-dwelling women and men., Participants and Methods: A total of 8226 adults from the Prevention of Renal and Vascular End-stage Disease (PREVEND) cohort (1997-1998) were enrolled in a prospective observational study (mean age: 49 years; 50.2% women). Sex-specific associations of cTnT levels with future clinical outcomes were evaluated using adjusted Cox-regression models., Results: Measurable cTnT levels (≥3 ng/L) were detected in 1102 women (26.7%) and in 2396 men (58.5%). Baseline cTnT levels were associated with a greater risk of developing CVD in women than men [Hazard ratio (HR
women ), 1.48 per unit increase in log2 -cTnT; 95% CI, 1.21 to 1.81 vs HRmen , 1.20; 95% CI, 1.07 to 1.35; Pinteraction <.001]. Similar sex-related differences were observed for HF (Pinteraction = .005) and mortality (Pinteraction = .008). Further, compared with referent category (cTnT <3 ng/L), women with cTnT levels greater than or equal to 6 ng/L had a significantly increased risk for CVD (HR, 2.30; 95% CI, 1.45 to 3.64), HF (HR, 2.86; 95% CI, 1.41 to 5.80), and mortality (HR, 2.65; 95% CI, 1.52 to 4.61), whereas men with cTnT levels greater than or equal to 6 ng/L had a significantly increased risk only for CVD (HR, 1.51; 95% CI, 1.07 to 2.13)., Conclusion: Baseline cTnT levels were associated with future CVD, HF, and mortality in both sexes, and these associations were stronger in women. Future studies are needed to determine the value of cTnT in early diagnosis of CVD, particularly in women., (Copyright © 2020 Mayo Foundation for Medical Education and Research. Published by Elsevier Inc. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
29. Dichotomy between the transcriptomic landscape of naturally versus accelerated aged murine hearts.
- Author
-
De Majo F, Hegenbarth JC, Rühle F, Bär C, Thum T, de Boer M, Duncker DJ, Schroen B, Armand AS, Stoll M, and De Windt LJ
- Subjects
- Aging metabolism, Aging, Premature metabolism, Aging, Premature physiopathology, Animals, Female, Humans, Ichthyosis, Lamellar genetics, Ichthyosis, Lamellar metabolism, Ichthyosis, Lamellar physiopathology, Male, Mice, Mitochondria genetics, Mitochondria metabolism, Proteins metabolism, Telomere genetics, Telomere metabolism, Telomere Shortening, Transcriptome, Aging genetics, Aging, Premature genetics, Heart growth & development, Myocardium metabolism, Proteins genetics
- Abstract
We investigated the transcriptomic landscape of the murine myocardium along the course of natural aging and in three distinct mouse models of premature aging with established aging-related cardiac dysfunction. Genome-wide total RNA-seq was performed and the expression patterns of protein-coding genes and non-coding RNAs were compared between hearts from naturally aging mice, mice with cardiac-specific deficiency of a component of the DNA repair machinery, mice with reduced mitochondrial antioxidant capacity and mice with reduced telomere length. Our results demonstrate that no dramatic changes are evident in the transcriptomes of naturally senescent murine hearts until two years of age, in contrast to the transcriptome of accelerated aged mice. Additionally, these mice displayed model-specific alterations of the expression levels of protein-coding and non-coding genes with hardly any overlap with age-related signatures. Our data demonstrate very limited similarities between the transcriptomes of all our murine aging models and question their reliability to study human cardiovascular senescence.
- Published
- 2020
- Full Text
- View/download PDF
30. A Year in the Life of the EU-CardioRNA COST Action: CA17129 Catalysing Transcriptomics Research in Cardiovascular Disease.
- Author
-
Robinson EL, Gomes CPDC, Potočnjak I, Hellemans J, Betsou F, de Gonzalo-Calvo D, Stoll M, Yilmaz MB, Ágg B, Beis D, Carmo-Fonseca M, Enguita FJ, Dogan S, Tuna BG, Schroen B, Ammerlaan W, Kuster GM, Carpusca I, Pedrazzini T, Emanueli C, Martelli F, and Devaux Y
- Abstract
The EU-CardioRNA Cooperation in Science and Technology (COST) Action is a European-wide consortium established in 2018 with 31 European country members and four associate member countries to build bridges between translational researchers from academia and industry who conduct research on non-coding RNAs, cardiovascular diseases and similar research areas. EU-CardioRNA comprises four core working groups (WG1-4). In the first year since its launch, EU-CardioRNA met biannually to exchange and discuss recent findings in related fields of scientific research, with scientific sessions broadly divided up according to WG. These meetings are also an opportunity to establish interdisciplinary discussion groups, brainstorm ideas and make plans to apply for joint research grants and conduct other scientific activities, including knowledge transfer. Following its launch in Brussels in 2018, three WG meetings have taken place. The first of these in Lisbon, Portugal, the second in Istanbul, Turkey, and the most recent in Maastricht, The Netherlands. Each meeting includes a scientific session from each WG. This meeting report briefly describes the highlights and key take-home messages from each WG session in this first successful year of the EU-CardioRNA COST Action., Competing Interests: No potential conflict of interest was reported by any of the authors. If you are interested in learning more about the EU-CardioRNA COST Action or to apply to join a Working Group, please contact the corresponding authors.
- Published
- 2020
- Full Text
- View/download PDF
31. tRNAs and tRNA fragments as modulators of cardiac and skeletal muscle function.
- Author
-
Liapi E, van Bilsen M, Verjans R, and Schroen B
- Subjects
- Gene Expression Regulation genetics, Heart physiopathology, Humans, Muscle, Skeletal metabolism, Muscle, Skeletal pathology, Muscular Diseases metabolism, Muscular Diseases pathology, Myocardium pathology, RNA Processing, Post-Transcriptional genetics, Heart growth & development, Muscular Diseases genetics, Myocardium metabolism, RNA, Transfer genetics
- Abstract
Transfer RNAs (tRNAs) and their processing enzymes have long-recognized roles in cardiac and skeletal muscle pathophysiology. Recently, tRNA fragments have emerged as a new class of non-coding RNAs involved in the regulation of cell function. In this review, we provide a synopsis of the molecular processes that regulate the biogenesis, post-transcriptional regulation and functional roles of tRNAs in cardiac and skeletal muscle. In addition, we list the (dys)regulated expression profiles and putative functional roles of tRNA-derived small RNAs in the heart and skeletal muscle. Finally, the technical challenges surrounding tRNA research are discussed alongside suggestions to advance research in this field., Competing Interests: Conflict of interest None., (Copyright © 2019 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
32. Reviewing the Limitations of Adult Mammalian Cardiac Regeneration: Noncoding RNAs as Regulators of Cardiomyogenesis.
- Author
-
Verjans R, van Bilsen M, and Schroen B
- Subjects
- Animals, Cell Differentiation genetics, Cell Proliferation genetics, Heart Failure genetics, Humans, Mammals genetics, Mammals metabolism, MicroRNAs genetics, Myocytes, Cardiac metabolism, RNA, Long Noncoding genetics, RNA, Long Noncoding metabolism, RNA, Untranslated metabolism, Regeneration genetics, Signal Transduction genetics, Muscle Development genetics, Myocardium metabolism, RNA, Untranslated genetics
- Abstract
The adult mammalian heart is incapable of regeneration following cardiac injury, leading to a decline in function and eventually heart failure. One of the most evident barriers limiting cardiac regeneration is the inability of cardiomyocytes to divide. It has recently become clear that the mammalian heart undergoes limited cardiomyocyte self-renewal throughout life and is even capable of modest regeneration early after birth. These exciting findings have awakened the goal to promote cardiomyogenesis of the human heart to repair cardiac injury or treat heart failure. We are still far from understanding why adult mammalian cardiomyocytes possess only a limited capacity to proliferate. Identifying the key regulators may help to progress towards such revolutionary therapy. Specific noncoding RNAs control cardiomyocyte division, including well explored microRNAs and more recently emerged long noncoding RNAs. Elucidating their function and molecular mechanisms during cardiomyogenesis is a prerequisite to advance towards therapeutic options for cardiac regeneration. In this review, we present an overview of the molecular basis of cardiac regeneration and describe current evidence implicating microRNAs and long noncoding RNAs in this process. Current limitations and future opportunities regarding how these regulatory mechanisms can be harnessed to study myocardial regeneration will be addressed., Competing Interests: The authors declare no conflict of interest.
- Published
- 2020
- Full Text
- View/download PDF
33. Regulatory RNAs in Heart Failure.
- Author
-
Gomes CPC, Schroen B, Kuster GM, Robinson EL, Ford K, Squire IB, Heymans S, Martelli F, Emanueli C, and Devaux Y
- Subjects
- Animals, Heart Failure genetics, Heart Failure pathology, Heart Failure therapy, Humans, RNA, Messenger genetics, RNA, Untranslated genetics, Heart Failure metabolism, RNA, Messenger metabolism, RNA, Untranslated metabolism
- Abstract
Cardiovascular disease is an enormous socioeconomic burden worldwide and remains a leading cause of mortality and disability despite significant efforts to improve treatments and personalize healthcare. Heart failure is the main manifestation of cardiovascular disease and has reached epidemic proportions. Heart failure follows a loss of cardiac homeostasis, which relies on a tight regulation of gene expression. This regulation is under the control of multiple types of RNA molecules, some encoding proteins (the so-called messenger RNAs) and others lacking protein-coding potential, named noncoding RNAs. In this review article, we aim to revisit the notion of regulatory RNA, which has been thus far mainly confined to noncoding RNA. Regulatory RNA, which we propose to abbreviate as regRNA, can include both protein-coding RNAs and noncoding RNAs, as long as they contribute, directly or indirectly, to the regulation of gene expression. We will address the regulation and functional role of messenger RNAs, microRNAs, long noncoding RNAs, and circular RNAs (ie, regRNAs) in heart failure. We will debate the utility of regRNAs to diagnose, prognosticate, and treat heart failure, and we will provide directions for future work.
- Published
- 2020
- Full Text
- View/download PDF
34. Long Noncoding RNA-Enriched Vesicles Secreted by Hypoxic Cardiomyocytes Drive Cardiac Fibrosis.
- Author
-
Kenneweg F, Bang C, Xiao K, Boulanger CM, Loyer X, Mazlan S, Schroen B, Hermans-Beijnsberger S, Foinquinos A, Hirt MN, Eschenhagen T, Funcke S, Stojanovic S, Genschel C, Schimmel K, Just A, Pfanne A, Scherf K, Dehmel S, Raemon-Buettner SM, Fiedler J, and Thum T
- Abstract
Long non-coding RNAs (lncRNAs) have potential as novel therapeutic targets in cardiovascular diseases, but detailed information about the intercellular lncRNA shuttling mechanisms in the heart is lacking. Here, we report an important novel crosstalk between cardiomyocytes and fibroblasts mediated by the transfer of lncRNA-enriched extracellular vesicles (EVs) in the context of cardiac ischemia. lncRNA profiling identified two hypoxia-sensitive lncRNAs: ENSMUST00000122745 was predominantly found in small EVs, whereas lncRNA Neat1 was enriched in large EVs in vitro and in vivo. Vesicles were taken up by fibroblasts, triggering expression of profibrotic genes. In addition, lncRNA Neat1 was transcriptionally regulated by P53 under basal conditions and by HIF2A during hypoxia. The function of Neat1 was further elucidated in vitro and in vivo. Silencing of Neat1 in vitro revealed that Neat1 was indispensable for fibroblast and cardiomyocyte survival and affected fibroblast functions (reduced migration capacity, stalled cell cycle, and decreased expression of fibrotic genes). Of translational importance, genetic loss of Neat1 in vivo resulted in an impaired heart function after myocardial infarction highlighting its translational relevance., (Copyright © 2019 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
35. Functional Screening Identifies MicroRNAs as Multi-Cellular Regulators of Heart Failure.
- Author
-
Verjans R, Derks WJA, Korn K, Sönnichsen B, van Leeuwen REW, Schroen B, van Bilsen M, and Heymans S
- Subjects
- Animals, Animals, Newborn, Cardiomegaly genetics, Cardiomegaly immunology, Cells, Cultured, Fibroblasts, Fibrosis, Gene Expression Profiling, Gene Expression Regulation, Heart Failure immunology, Heart Failure pathology, Humans, Macrophage Activation genetics, Macrophage Activation immunology, Macrophages, Mice, Myocarditis genetics, Myocarditis pathology, Myocardium cytology, Myocardium immunology, Myocytes, Cardiac, Primary Cell Culture, Rats, Cardiomegaly pathology, Heart Failure genetics, MicroRNAs metabolism, Myocarditis immunology, Myocardium pathology
- Abstract
Heart failure (HF) is the leading cause of death in the Western world. Pathophysiological processes underlying HF development, including cardiac hypertrophy, fibrosis and inflammation, are controlled by specific microRNAs (miRNAs). Whereas most studies investigate miRNA function in one particular cardiac cell type, their multicellular function is poorly investigated. The present study probed 194 miRNAs -differentially expressed in cardiac inflammatory disease - for regulating cardiomyocyte size, cardiac fibroblasts collagen content, and macrophage polarization. Of the tested miRNAs, 13%, 26%, and 41% modulated cardiomyocyte size, fibroblast collagen production, and macrophage polarization, respectively. Seventeen miRNAs affected all three cellular processes, including miRNAs with established (miR-210) and unknown roles in cardiac pathophysiology (miR-145-3p). These miRNAs with a multi-cellular function commonly target various genes. In-depth analysis in vitro of previously unstudied miRNAs revealed that the observed phenotypical alterations concurred with changes in transcript and protein levels of hypertrophy-, fibrosis- and inflammation-related genes. MiR-145-3p and miR-891a-3p were identified to regulate the fibrotic response, whereas miR-223-3p, miR-486-3p, and miR-488-5p modulated macrophage activation and polarisation. In conclusion, miRNAs are multi-cellular regulators of different cellular processes underlying cardiac disease. We identified previously undescribed roles of miRNAs in hypertrophy, fibrosis, and inflammation, and attribute new cellular effects to various well-known miRNAs.
- Published
- 2019
- Full Text
- View/download PDF
36. AntagomiR-103 and -107 Treatment Affects Cardiac Function and Metabolism.
- Author
-
Rech M, Kuhn AR, Lumens J, Carai P, van Leeuwen R, Verhesen W, Verjans R, Lecomte J, Liu Y, Luiken JJFP, Mohren R, Cillero-Pastor B, Heymans S, Knoops K, van Bilsen M, and Schroen B
- Abstract
MicroRNA-103/107 regulate systemic glucose metabolism and insulin sensitivity. For this reason, inhibitory strategies for these microRNAs are currently being tested in clinical trials. Given the high metabolic demands of the heart and the abundant cardiac expression of miR-103/107, we questioned whether antagomiR-mediated inhibition of miR-103/107 in C57BL/6J mice impacts on cardiac function. Notably, fractional shortening decreased after 6 weeks of antagomiR-103 and -107 treatment. This was paralleled by a prolonged systolic radial and circumferential time to peak and by a decreased global strain rate. Histology and electron microscopy showed reduced cardiomyocyte area and decreased mitochondrial volume and mitochondrial cristae density following antagomiR-103 and -107. In line, antagomiR-103 and -107 treatment decreased mitochondrial OXPHOS complexes' protein levels compared to scrambled, as assessed by mass spectrometry-based label-free quantitative proteomics. MiR-103/107 inhibition in primary cardiomyocytes did not affect glycolysis rates, but it decreased mitochondrial reserve capacity, reduced mitochondrial membrane potential, and altered mitochondrial network morphology, as assessed by live-cell imaging. Our data indicate that antagomiR-103 and -107 decrease cardiac function, cardiomyocyte size, and mitochondrial oxidative capacity in the absence of pathological stimuli. These data raise concern about the possible cardiac implications of the systemic use of antagomiR-103 and -107 in the clinical setting, and careful cardiac phenotyping within ongoing trials is highly recommended., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
37. Targeted HFpEF therapy based on matchmaking of human and animal models.
- Author
-
Barandiarán Aizpurua A, Schroen B, van Bilsen M, and van Empel V
- Subjects
- Animals, Heart Failure etiology, Heart Failure physiopathology, Humans, Disease Models, Animal, Heart Failure therapy, Stroke Volume, Translational Research, Biomedical methods
- Abstract
The diversity in clinical phenotypes and poor understanding of the underlying pathophysiology of heart failure with preserved ejection fraction (HFpEF) is the main reason why no effective treatments have been found yet. Targeted, instead of one size fits all, treatment seems the only promising approach for treating HFpEF. To be able to design a targeted, phenotype-specific HFpEF treatment, the matrix relating clinical phenotypes and underlying pathophysiological mechanisms has to be clarified. This review discusses the opportunities for additional evaluation of the underlying pathophysiological processes, e.g., to evaluate biological phenotypes on top of clinical routine, to guide us toward a phenotype-specific HFpEF treatment. Moreover, a translational approach with matchmaking of animal models to biological HFpEF phenotypes will be a valuable step to test the effectiveness of novel, targeted interventions in HFpEF. Listen to this article's corresponding podcast at https://ajpheart.podbean.com/e/personalized-medicine-in-hfpef/ .
- Published
- 2018
- Full Text
- View/download PDF
38. MicroRNA-155 Amplifies Nitric Oxide/cGMP Signaling and Impairs Vascular Angiotensin II Reactivity in Septic Shock.
- Author
-
Vasques-Nóvoa F, Laundos TL, Cerqueira RJ, Quina-Rodrigues C, Soares-Dos-Reis R, Baganha F, Ribeiro S, Mendonça L, Gonçalves F, Reguenga C, Verhesen W, Carneiro F, Paiva JA, Schroen B, Castro-Chaves P, Pinto-do-Ó P, Nascimento DS, Heymans S, Leite-Moreira AF, and Roncon-Albuquerque R Jr
- Subjects
- Animals, Blood Vessels metabolism, Blood Vessels physiopathology, Cells, Cultured, Endothelial Cells, Heart physiopathology, Humans, Male, Mice, Mice, Inbred C57BL, Myocardium metabolism, Prospective Studies, Random Allocation, Shock, Septic genetics, Signal Transduction, Angiotensin II physiology, Cyclic GMP physiology, MicroRNAs physiology, Nitric Oxide physiology, Shock, Septic complications
- Abstract
Objectives: Septic shock is a life-threatening clinical situation associated with acute myocardial and vascular dysfunction, whose pathophysiology is still poorly understood. Herein, we investigated microRNA-155-dependent mechanisms of myocardial and vascular dysfunction in septic shock., Design: Prospective, randomized controlled experimental murine study and clinical cohort analysis., Setting: University research laboratory and ICU at a tertiary-care center., Patients: Septic patients, ICU controls, and healthy controls. Postmortem myocardial samples from septic and nonseptic patients. Ex vivo evaluation of arterial rings from patients undergoing coronary artery bypass grafting., Subjects: C57Bl/6J and genetic background-matched microRNA-155 knockout mice., Interventions: Two mouse models of septic shock were used. Genetic deletion and pharmacologic inhibition of microRNA-155 were performed. Ex vivo myographic studies were performed using mouse and human arterial rings., Measurements and Main Results: We identified microRNA-155 as a highly up-regulated multifunctional mediator of sepsis-associated cardiovascular dysfunction. In humans, plasma and myocardial microRNA-155 levels correlate with sepsis-related mortality and cardiac injury, respectively, whereas in murine models, microRNA-155 deletion and pharmacologic inhibition attenuate sepsis-associated cardiovascular dysfunction and mortality. MicroRNA-155 up-regulation in septic myocardium was found to be mostly supported by microvascular endothelial cells. This promoted myocardial microvascular permeability and edema, bioenergetic deterioration, contractile dysfunction, proinflammatory, and nitric oxide-cGMP-protein kinase G signaling overactivation. In isolate cardiac microvascular endothelial cells, microRNA-155 up-regulation significantly contributes to LPS-induced proinflammatory cytokine up-regulation, leukocyte adhesion, and nitric oxide overproduction. Furthermore, we identified direct targeting of CD47 by microRNA-155 as a novel mechanism of myocardial and vascular contractile depression in sepsis, promoting microvascular endothelial cell and vascular insensitivity to thrombospondin-1-mediated inhibition of nitric oxide production and nitric oxide-mediated vasorelaxation, respectively. Additionally, microRNA-155 directly targets angiotensin type 1 receptor, decreasing vascular angiotensin II reactivity. Deletion of microRNA-155 restored angiotensin II and thrombospondin-1 vascular reactivity in LPS-exposed arterial rings., Conclusions: Our study demonstrates multiple new microRNA-155-mediated mechanisms of sepsis-associated cardiovascular dysfunction, supporting the translational potential of microRNA-155 inhibition in human septic shock.
- Published
- 2018
- Full Text
- View/download PDF
39. Sex-specific associations of obesity and N-terminal pro-B-type natriuretic peptide levels in the general population.
- Author
-
Suthahar N, Meijers WC, Ho JE, Gansevoort RT, Voors AA, van der Meer P, Bakker SJL, Heymans S, van Empel V, Schroen B, van der Harst P, van Veldhuisen DJ, and de Boer RA
- Subjects
- Adult, Body Mass Index, Female, Follow-Up Studies, Heart Failure etiology, Humans, Incidence, Male, Middle Aged, Netherlands epidemiology, Obesity complications, Obesity epidemiology, Prognosis, Prospective Studies, Risk Factors, Sex Distribution, Sex Factors, Heart Failure blood, Natriuretic Peptide, Brain blood, Obesity blood, Peptide Fragments blood, Population Surveillance
- Abstract
Background: Obese subjects have lower natriuretic peptide levels, but males and females have different anthropometric characteristics and fat distribution. Whether obesity-associated lowering of natriuretic peptides differs among males and females is unknown. Therefore, we investigated sex-specific associations of obesity and N-terminal pro-B-type natriuretic peptide (NT-proBNP) levels among adults in the general population., Methods and Results: Using 8260 participants (50.1% females) from the Prevention of REnal and Vascular ENd-stage Disease (PREVEND) cohort, we evaluated the relationship of NT-proBNP levels with obesity-associated parameters, i.e. waist circumference (WC), body mass index (BMI) and body weight in the overall population, and in males and females separately. NT-proBNP levels were higher in females (median, interquartile range: 50.5, 28.2-87.0 ng/L) than in males (24.3, 10.1-54.6 ng/L; P < 0.001). In the overall population, NT-proBNP levels were significantly lower in heavier individuals and displayed a 'U-shaped' relationship with increasing WC, but were not associated with BMI. After sex stratification, there was no significant association between NT-proBNP concentrations and anthropometric measures in females. However, in males increasing WC and BMI were associated with higher NT-proBNP levels (P < 0.05) while increasing body weight was associated with slightly lower NT-proBNP levels (P < 0.05). Age strongly confounded the association of NT-proBNP levels with obesity, and age-associated increases in NT-proBNP were significantly higher in males than in females (P < 0.001). In multivariable adjusted analyses, the inverse association of obesity and NT-proBNP levels was also significantly modified by sex: NT-proBNP levels were lower with increasing WC, BMI and body weight among females compared with males (P
interaction < 0.05). After also accounting for BMI, abdominal obesity was associated with lower NT-proBNP levels in females, but not in males (Pinteraction < 0.001)., Conclusions: Natriuretic peptide deficiency in obesity mostly pertains to females with abdominal obesity, whereas the relationship between obesity and natriuretic peptides appears to be more complex in males., (© 2018 The Authors. European Journal of Heart Failure © 2018 European Society of Cardiology.)- Published
- 2018
- Full Text
- View/download PDF
40. Pathophysiological understanding of HFpEF: microRNAs as part of the puzzle.
- Author
-
Rech M, Barandiarán Aizpurua A, van Empel V, van Bilsen M, and Schroen B
- Subjects
- Animals, Gene Expression Regulation, Genetic Predisposition to Disease, Heart Failure metabolism, Humans, MicroRNAs metabolism, Multimorbidity, Phenotype, Risk Factors, Signal Transduction, Heart Failure genetics, Heart Failure physiopathology, MicroRNAs genetics, Stroke Volume genetics, Ventricular Function, Left genetics
- Abstract
Half of all heart failure patients have preserved ejection fraction (HFpEF). Comorbidities associated with and contributing to HFpEF include obesity, diabetes and hypertension. Still, the underlying pathophysiological mechanisms of HFpEF are unknown. A preliminary consensus proposes that the multi-morbidity triggers a state of systemic, chronic low-grade inflammation, and microvascular dysfunction, causing reduced nitric oxide bioavailability to adjacent cardiomyocytes. As a result, the cardiomyocyte remodels its contractile elements and fails to relax properly, causing diastolic dysfunction, and eventually HFpEF. HFpEF is a complex syndrome for which currently no efficient therapies exist. This is notably due to the current one-size-fits-all therapy approach that ignores individual patient differences. MicroRNAs have been studied in relation to pathophysiological mechanisms and comorbidities underlying and contributing to HFpEF. As regulators of gene expression, microRNAs may contribute to the pathophysiology of HFpEF. In addition, secreted circulating microRNAs are potential biomarkers and as such, they could help stratify the HFpEF population and open new ways for individualized therapies. In this review, we provide an overview of the ever-expanding world of non-coding RNAs and their contribution to the molecular mechanisms underlying HFpEF. We propose prospects for microRNAs in stratifying the HFpEF population. MicroRNAs add a new level of complexity to the regulatory network controlling cardiac function and hence the understanding of gene regulation becomes a fundamental piece in solving the HFpEF puzzle.
- Published
- 2018
- Full Text
- View/download PDF
41. Long non-coding RNAs in the failing heart and vasculature.
- Author
-
Hermans-Beijnsberger S, van Bilsen M, and Schroen B
- Abstract
Following completion of the human genome, it became evident that the majority of our DNA is transcribed into non-coding RNAs (ncRNAs) instead of protein-coding messenger RNA. Deciphering the function of these ncRNAs, including both small- and long ncRNAs (lncRNAs), is an emerging field of research. LncRNAs have been associated with many disorders and a number have been identified as key regulators in the development and progression of disease, including cardiovascular disease (CVD). CVD causes millions of deaths worldwide, annually. Risk factors include coronary artery disease, high blood pressure and ageing. In this review, we will focus on the roles of lncRNAs in the cellular and molecular processes that underlie the development of CVD: cardiomyocyte hypertrophy, fibrosis, inflammation, vascular disease and ageing. Finally, we discuss the biomarker and therapeutic potential of lncRNAs.
- Published
- 2018
- Full Text
- View/download PDF
42. MicroRNA-221/222 Family Counteracts Myocardial Fibrosis in Pressure Overload-Induced Heart Failure.
- Author
-
Verjans R, Peters T, Beaumont FJ, van Leeuwen R, van Herwaarden T, Verhesen W, Munts C, Bijnen M, Henkens M, Diez J, de Windt LJ, van Nieuwenhoven FA, van Bilsen M, Goumans MJ, Heymans S, González A, and Schroen B
- Subjects
- Animals, Aortic Valve Stenosis complications, Aortic Valve Stenosis metabolism, Cardiomyopathies metabolism, Fibroblasts metabolism, Fibrosis metabolism, Humans, Male, Mice, Mice, Inbred C57BL, Myocardium pathology, Proto-Oncogene Mas, Rats, Signal Transduction, Transforming Growth Factor beta metabolism, Heart Failure metabolism, MicroRNAs metabolism, Myocardium metabolism
- Abstract
Pressure overload causes cardiac fibroblast activation and transdifferentiation, leading to increased interstitial fibrosis formation and subsequently myocardial stiffness, diastolic and systolic dysfunction, and eventually heart failure. A better understanding of the molecular mechanisms underlying pressure overload-induced cardiac remodeling and fibrosis will have implications for heart failure treatment strategies. The microRNA (miRNA)-221/222 family, consisting of miR-221-3p and miR-222-3p, is differentially regulated in mouse and human cardiac pathology and inversely associated with kidney and liver fibrosis. We investigated the role of this miRNA family during pressure overload-induced cardiac remodeling. In myocardial biopsies of patients with severe fibrosis and dilated cardiomyopathy or aortic stenosis, we found significantly lower miRNA-221/222 levels as compared to matched patients with nonsevere fibrosis. In addition, miRNA-221/222 levels in aortic stenosis patients correlated negatively with the extent of myocardial fibrosis and with left ventricular stiffness. Inhibition of both miRNAs during AngII (angiotensin II)-mediated pressure overload in mice led to increased fibrosis and aggravated left ventricular dilation and dysfunction. In rat cardiac fibroblasts, inhibition of miRNA-221/222 derepressed TGF-β (transforming growth factor-β)-mediated profibrotic SMAD2 (mothers against decapentaplegic homolog 2) signaling and downstream gene expression, whereas overexpression of both miRNAs blunted TGF-β-induced profibrotic signaling. We found that the miRNA-221/222 family may target several genes involved in TGF-β signaling, including JNK1 (c-Jun N-terminal kinase 1), TGF-β receptor 1 and TGF-β receptor 2, and ETS-1 (ETS proto-oncogene 1). Our findings show that heart failure-associated downregulation of the miRNA-221/222 family enables profibrotic signaling in the pressure-overloaded heart., (© 2017 American Heart Association, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
43. Cartilage intermediate layer protein 1 (CILP1): A novel mediator of cardiac extracellular matrix remodelling.
- Author
-
van Nieuwenhoven FA, Munts C, Op't Veld RC, González A, Díez J, Heymans S, Schroen B, and van Bilsen M
- Subjects
- Animals, Extracellular Matrix Proteins genetics, Fibroblasts cytology, Fibroblasts metabolism, HEK293 Cells, Humans, Mice, Mice, Inbred C57BL, Myocardium pathology, Pyrophosphatases genetics, Transforming Growth Factor beta metabolism, Extracellular Matrix metabolism, Extracellular Matrix Proteins metabolism, Myocardium metabolism, Pyrophosphatases metabolism
- Abstract
Heart failure is accompanied by extracellular matrix (ECM) remodelling, often leading to cardiac fibrosis. In the present study we explored the significance of cartilage intermediate layer protein 1 (CILP1) as a novel mediator of cardiac ECM remodelling. Whole genome transcriptional analysis of human cardiac tissue samples revealed a strong association of CILP1 with many structural (e.g. COL1A2 r
2 = 0.83) and non-structural (e.g. TGFB3 r2 = 0.75) ECM proteins. Gene enrichment analysis further underscored the involvement of CILP1 in human cardiac ECM remodelling and TGFβ signalling. Myocardial CILP1 protein levels were significantly elevated in human infarct tissue and in aortic valve stenosis patients. CILP1 mRNA levels markedly increased in mouse heart after myocardial infarction, transverse aortic constriction, and angiotensin II treatment. Cardiac fibroblasts were found to be the primary source of cardiac CILP1 expression. Recombinant CILP1 inhibited TGFβ-induced αSMA gene and protein expression in cardiac fibroblasts. In addition, CILP1 overexpression in HEK293 cells strongly (5-fold p < 0.05) inhibited TGFβ signalling activity. In conclusion, our study identifies CILP1 as a new cardiac matricellular protein interfering with pro-fibrotic TGFβ signalling, and as a novel sensitive marker for cardiac fibrosis.- Published
- 2017
- Full Text
- View/download PDF
44. Inhibition of MicroRNA-146a and Overexpression of Its Target Dihydrolipoyl Succinyltransferase Protect Against Pressure Overload-Induced Cardiac Hypertrophy and Dysfunction.
- Author
-
Heggermont WA, Papageorgiou AP, Quaegebeur A, Deckx S, Carai P, Verhesen W, Eelen G, Schoors S, van Leeuwen R, Alekseev S, Elzenaar I, Vinckier S, Pokreisz P, Walravens AS, Gijsbers R, Van Den Haute C, Nickel A, Schroen B, van Bilsen M, Janssens S, Maack C, Pinto Y, Carmeliet P, and Heymans S
- Subjects
- Acyltransferases genetics, Animals, Animals, Newborn, Cardiomegaly genetics, Cardiomegaly prevention & control, Cells, Cultured, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, MicroRNAs genetics, Myocytes, Cardiac metabolism, Rats, Rats, Inbred Lew, Ventricular Dysfunction, Left genetics, Ventricular Dysfunction, Left prevention & control, Acyltransferases biosynthesis, Cardiomegaly metabolism, Gene Expression Regulation, Enzymologic, MicroRNAs antagonists & inhibitors, MicroRNAs biosynthesis, Ventricular Dysfunction, Left metabolism
- Abstract
Background: Cardiovascular diseases remain the predominant cause of death worldwide, with the prevalence of heart failure continuing to increase. Despite increased knowledge of the metabolic alterations that occur in heart failure, novel therapies to treat the observed metabolic disturbances are still lacking., Methods: Mice were subjected to pressure overload by means of angiotensin-II infusion or transversal aortic constriction. MicroRNA-146a was either genetically or pharmacologically knocked out or genetically overexpressed in cardiomyocytes. Furthermore, overexpression of dihydrolipoyl succinyltransferase (DLST) in the murine heart was performed by means of an adeno-associated virus., Results: MicroRNA-146a was upregulated in whole heart tissue in multiple murine pressure overload models. Also, microRNA-146a levels were moderately increased in left ventricular biopsies of patients with aortic stenosis. Overexpression of microRNA-146a in cardiomyocytes provoked cardiac hypertrophy and left ventricular dysfunction in vivo, whereas genetic knockdown or pharmacological blockade of microRNA-146a blunted the hypertrophic response and attenuated cardiac dysfunction in vivo. Mechanistically, microRNA-146a reduced its target DLST-the E2 subcomponent of the α-ketoglutarate dehydrogenase complex, a rate-controlling tricarboxylic acid cycle enzyme. DLST protein levels significantly decreased on pressure overload in wild-type mice, paralleling a decreased oxidative metabolism, whereas DLST protein levels and hence oxidative metabolism were partially maintained in microRNA-146a knockout mice. Moreover, overexpression of DLST in wild-type mice protected against cardiac hypertrophy and dysfunction in vivo., Conclusions: Altogether we show that the microRNA-146a and its target DLST are important metabolic players in left ventricular dysfunction., (© 2017 American Heart Association, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
45. The long noncoding RNA Wisper controls cardiac fibrosis and remodeling.
- Author
-
Micheletti R, Plaisance I, Abraham BJ, Sarre A, Ting CC, Alexanian M, Maric D, Maison D, Nemir M, Young RA, Schroen B, González A, Ounzain S, and Pedrazzini T
- Subjects
- Cardiomyopathies pathology, Fibroblasts metabolism, Fibroblasts pathology, Fibrosis genetics, Fibrosis pathology, Humans, RNA, Long Noncoding physiology, Ventricular Remodeling, Cardiomyopathies genetics, RNA, Long Noncoding genetics
- Abstract
Long noncoding RNAs (lncRNAs) are emerging as powerful regulators of cardiac development and disease. However, our understanding of the importance of these molecules in cardiac fibrosis is limited. Using an integrated genomic screen, we identified Wisper (Wisp2 super-enhancer-associated RNA) as a cardiac fibroblast-enriched lncRNA that regulates cardiac fibrosis after injury. Wisper expression was correlated with cardiac fibrosis both in a murine model of myocardial infarction (MI) and in heart tissue from human patients suffering from aortic stenosis. Loss-of-function approaches in vitro using modified antisense oligonucleotides (ASOs) demonstrated that Wisper is a specific regulator of cardiac fibroblast proliferation, migration, and survival. Accordingly, ASO-mediated silencing of Wisper in vivo attenuated MI-induced fibrosis and cardiac dysfunction. Functionally, Wisper regulates cardiac fibroblast gene expression programs critical for cell identity, extracellular matrix deposition, proliferation, and survival. In addition, its association with TIA1-related protein allows it to control the expression of a profibrotic form of lysyl hydroxylase 2, implicated in collagen cross-linking and stabilization of the matrix. Together, our findings identify Wisper as a cardiac fibroblast-enriched super-enhancer-associated lncRNA that represents an attractive therapeutic target to reduce the pathological development of cardiac fibrosis in response to MI and prevent adverse remodeling in the damaged heart., (Copyright © 2017 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2017
- Full Text
- View/download PDF
46. Transcriptome-wide co-expression analysis identifies LRRC2 as a novel mediator of mitochondrial and cardiac function.
- Author
-
McDermott-Roe C, Leleu M, Rowe GC, Palygin O, Bukowy JD, Kuo J, Rech M, Hermans-Beijnsberger S, Schaefer S, Adami E, Creemers EE, Heinig M, Schroen B, Arany Z, Petretto E, and Geurts AM
- Subjects
- Animals, Heart Failure genetics, Heart Failure metabolism, Heat-Shock Proteins genetics, Heat-Shock Proteins metabolism, Humans, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 genetics, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 metabolism, Mitochondrial Proteins genetics, Mitochondrial Proteins metabolism, Rats, Transcription Factors genetics, Transcription Factors metabolism, Mitochondria metabolism, Myocytes, Cardiac metabolism, Transcriptome genetics
- Abstract
Mitochondrial dysfunction contributes to myriad monogenic and complex pathologies. To understand the underlying mechanisms, it is essential to define the full complement of proteins that modulate mitochondrial function. To identify such proteins, we performed a meta-analysis of publicly available gene expression data. Gene co-expression analysis of a large and heterogeneous compendium of microarray data nominated a sub-population of transcripts that whilst highly correlated with known mitochondrial protein-encoding transcripts (MPETs), are not themselves recognized as generating proteins either localized to the mitochondrion or pertinent to functions therein. To focus the analysis on a medically-important condition with a strong yet incompletely understood mitochondrial component, candidates were cross-referenced with an MPET-enriched module independently generated via genome-wide co-expression network analysis of a human heart failure gene expression dataset. The strongest uncharacterized candidate in the analysis was Leucine Rich Repeat Containing 2 (LRRC2). LRRC2 was found to be localized to the mitochondria in human cells and transcriptionally-regulated by the mitochondrial master regulator Pgc-1α. We report that Lrrc2 transcript abundance correlates with that of β-MHC, a canonical marker of cardiac hypertrophy in humans and experimentally demonstrated an elevation in Lrrc2 transcript in in vitro and in vivo rodent models of cardiac hypertrophy as well as in patients with dilated cardiomyopathy. RNAi-mediated Lrrc2 knockdown in a rat-derived cardiomyocyte cell line resulted in enhanced expression of canonical hypertrophic biomarkers as well as increased mitochondrial mass in the context of increased Pgc-1α expression. In conclusion, our meta-analysis represents a simple yet powerful springboard for the nomination of putative mitochondrially-pertinent proteins relevant to cardiac function and enabled the identification of LRRC2 as a novel mitochondrially-relevant protein and regulator of the hypertrophic response., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2017
- Full Text
- View/download PDF
47. MiRNA Deregulation in Cardiac Aging and Associated Disorders.
- Author
-
Verjans R, van Bilsen M, and Schroen B
- Subjects
- Animals, Apoptosis genetics, Calcium metabolism, Humans, MicroRNAs metabolism, Mitochondria metabolism, Aging genetics, MicroRNAs genetics, Myocardium pathology
- Abstract
The prevalence of age-related diseases is increasing dramatically, among which cardiac disease represents the leading cause of death. Aging of the heart is characterized by various molecular and cellular hallmarks impairing both cardiomyocytes and noncardiomyocytes, and resulting in functional deteriorations of the cardiac system. The aging process includes desensitization of β-adrenergic receptor (βAR)-signaling and decreased calcium handling, altered growth signaling and cardiac hypertrophy, mitochondrial dysfunction and impaired autophagy, increased programmed cell death, low-grade inflammation of noncanonical inflammatory cells, and increased ECM deposition. MiRNAs play a fundamental role in regulating the processes underlying these detrimental changes in the cardiac system, indicating that MiRNAs are crucially involved in aging. Among others, MiR-34, MiR-146a, and members of the MiR-17-92 cluster, are deregulated during senescence and drive cardiac aging processes. It is therefore suggested that MiRNAs form possible therapeutic targets to stabilize the aged failing myocardium., (© 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
48. Long noncoding RNA MALAT1-derived mascRNA is involved in cardiovascular innate immunity.
- Author
-
Gast M, Schroen B, Voigt A, Haas J, Kuehl U, Lassner D, Skurk C, Escher F, Wang X, Kratzer A, Michalik K, Papageorgiou A, Peters T, Loebel M, Wilk S, Althof N, Prasanth KV, Katus H, Meder B, Nakagawa S, Scheibenbogen C, Schultheiss HP, Landmesser U, Dimmeler S, Heymans S, and Poller W
- Subjects
- Humans, Cardiovascular System immunology, Cardiovascular System metabolism, Immunity, Innate genetics, RNA, Long Noncoding genetics, RNA, Long Noncoding metabolism
- Published
- 2016
- Full Text
- View/download PDF
49. Long Non-Coding RNA Malat-1 Is Dispensable during Pressure Overload-Induced Cardiac Remodeling and Failure in Mice.
- Author
-
Peters T, Hermans-Beijnsberger S, Beqqali A, Bitsch N, Nakagawa S, Prasanth KV, de Windt LJ, van Oort RJ, Heymans S, and Schroen B
- Subjects
- Adaptor Proteins, Signal Transducing, Angiotensin II metabolism, Angiotensin II toxicity, Animals, Aorta, Thoracic, Cardiomegaly etiology, Constriction, Pathologic complications, Crosses, Genetic, Fetal Proteins biosynthesis, Fetal Proteins genetics, Gene Expression Regulation genetics, Heart Failure etiology, Heterozygote, Ligation, Mice, Mice, Inbred C57BL, Mice, Inbred CBA, Mice, Knockout, Pressure, Proteins genetics, Proteins metabolism, RNA, Long Noncoding genetics, Cardiomegaly genetics, Heart Failure genetics, RNA Splicing genetics, RNA, Long Noncoding physiology, Ventricular Remodeling genetics
- Abstract
Background: Long non-coding RNAs (lncRNAs) are a class of RNA molecules with diverse regulatory functions during embryonic development, normal life, and disease in higher organisms. However, research on the role of lncRNAs in cardiovascular diseases and in particular heart failure is still in its infancy. The exceptionally well conserved nuclear lncRNA Metastasis associated in lung adenocarcinoma transcript 1 (Malat-1) is a regulator of mRNA splicing and highly expressed in the heart. Malat-1 modulates hypoxia-induced vessel growth, activates ERK/MAPK signaling, and scavenges the anti-hypertrophic microRNA-133. We therefore hypothesized that Malat-1 may act as regulator of cardiac hypertrophy and failure during cardiac pressure overload induced by thoracic aortic constriction (TAC) in mice., Results: Absence of Malat-1 did not affect cardiac hypertrophy upon pressure overload: Heart weight to tibia length ratio significantly increased in WT mice (sham: 5.78±0.55, TAC 9.79±1.82 g/mm; p<0.001) but to a similar extend also in Malat-1 knockout (KO) mice (sham: 6.21±1.12, TAC 8.91±1.74 g/mm; p<0.01) with no significant difference between genotypes. As expected, TAC significantly reduced left ventricular fractional shortening in WT (sham: 38.81±6.53%, TAC: 23.14±11.99%; p<0.01) but to a comparable degree also in KO mice (sham: 37.01±4.19%, TAC: 25.98±9.75%; p<0.05). Histological hallmarks of myocardial remodeling, such as cardiomyocyte hypertrophy, increased interstitial fibrosis, reduced capillary density, and immune cell infiltration, did not differ significantly between WT and KO mice after TAC. In line, the absence of Malat-1 did not significantly affect angiotensin II-induced cardiac hypertrophy, dysfunction, and overall remodeling. Above that, pressure overload by TAC significantly induced mRNA levels of the hypertrophy marker genes Nppa, Nppb and Acta1, to a similar extend in both genotypes. Alternative splicing of Ndrg2 after TAC was apparent in WT (isoform ratio; sham: 2.97±0.26, TAC 1.57±0.40; p<0.0001) and KO mice (sham: 3.64±0.37; TAC: 2.24±0.76; p<0.0001) and interestingly differed between genotypes both at baseline and after pressure overload (p<0.05 each)., Conclusion: These findings confirm a role for the lncRNA Malat-1 in mRNA splicing. However, no critical role for Malat-1 was found in pressure overload-induced heart failure in mice, despite its reported role in vascularization, ERK/MAPK signaling, and regulation of miR-133.
- Published
- 2016
- Full Text
- View/download PDF
50. CARMEN, a human super enhancer-associated long noncoding RNA controlling cardiac specification, differentiation and homeostasis.
- Author
-
Ounzain S, Micheletti R, Arnan C, Plaisance I, Cecchi D, Schroen B, Reverter F, Alexanian M, Gonzales C, Ng SY, Bussotti G, Pezzuto I, Notredame C, Heymans S, Guigó R, Johnson R, and Pedrazzini T
- Subjects
- Animals, Cell Lineage genetics, Enhancer Elements, Genetic genetics, Enhancer of Zeste Homolog 2 Protein, Gene Expression Profiling, Gene Expression Regulation, Developmental, Gene Knockdown Techniques, Humans, Mice, Myocardium pathology, Polycomb Repressive Complex 2 metabolism, RNA, Long Noncoding genetics, Stem Cells cytology, Transcriptome genetics, Body Patterning genetics, Cell Differentiation genetics, Heart embryology, Homeostasis genetics, RNA, Long Noncoding metabolism
- Abstract
Long noncoding RNAs (lncRNAs) are emerging as important regulators of developmental pathways. However, their roles in human cardiac precursor cell (CPC) remain unexplored. To characterize the long noncoding transcriptome during human CPC cardiac differentiation, we profiled the lncRNA transcriptome in CPCs isolated from the human fetal heart and identified 570 lncRNAs that were modulated during cardiac differentiation. Many of these were associated with active cardiac enhancer and super enhancers (SE) with their expression being correlated with proximal cardiac genes. One of the most upregulated lncRNAs was a SE-associated lncRNA that was named CARMEN, (CAR)diac (M)esoderm (E)nhancer-associated (N)oncoding RNA. CARMEN exhibits RNA-dependent enhancing activity and is upstream of the cardiac mesoderm-specifying gene regulatory network. Interestingly, CARMEN interacts with SUZ12 and EZH2, two components of the polycomb repressive complex 2 (PRC2). We demonstrate that CARMEN knockdown inhibits cardiac specification and differentiation in cardiac precursor cells independently of MIR-143 and -145 expression, two microRNAs located proximal to the enhancer sequences. Importantly, CARMEN expression was activated during pathological remodeling in the mouse and human hearts, and was necessary for maintaining cardiac identity in differentiated cardiomyocytes. This study demonstrates therefore that CARMEN is a crucial regulator of cardiac cell differentiation and homeostasis., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.