14 results on '"Versteeg, Danielle"'
Search Results
2. Cardiomyocyte proliferation is suppressed by ARID1A-mediated YAP inhibition during cardiac maturation.
- Author
-
Boogerd, Cornelis J., Perini, Ilaria, Kyriakopoulou, Eirini, Han, Su Ji, La, Phit, van der Swaan, Britt, Berkhout, Jari B., Versteeg, Danielle, Monshouwer-Kloots, Jantine, and van Rooij, Eva
- Subjects
YAP signaling proteins ,CHROMATIN-remodeling complexes ,MYOCARDIAL ischemia ,CARDIAC regeneration ,CORONARY disease ,TRANSCRIPTION factors - Abstract
The inability of adult human cardiomyocytes to proliferate is an obstacle to efficient cardiac regeneration after injury. Understanding the mechanisms that drive postnatal cardiomyocytes to switch to a non-regenerative state is therefore of great significance. Here we show that Arid1a, a subunit of the switching defective/sucrose non-fermenting (SWI/SNF) chromatin remodeling complex, suppresses postnatal cardiomyocyte proliferation while enhancing maturation. Genome-wide transcriptome and epigenome analyses revealed that Arid1a is required for the activation of a cardiomyocyte maturation gene program by promoting DNA access to transcription factors that drive cardiomyocyte maturation. Furthermore, we show that ARID1A directly binds and inhibits the proliferation-promoting transcriptional coactivators YAP and TAZ, indicating ARID1A sequesters YAP/TAZ from their DNA-binding partner TEAD. In ischemic heart disease, Arid1a expression is enhanced in cardiomyocytes of the border zone region. Inactivation of Arid1a after ischemic injury enhanced proliferation of border zone cardiomyocytes. Our study illuminates the pivotal role of Arid1a in cardiomyocyte maturation, and uncovers Arid1a as a crucial suppressor of cardiomyocyte proliferation. Cardiac regeneration is hindered by the limited division of cardiomyocytes. Here, the authors show that Arid1a drives maturation and limits proliferation through interaction with Yap. Suppression of Arid1a enhances proliferation after injury. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
3. Desmosomal protein degradation as an underlying cause of arrhythmogenic cardiomyopathy.
- Author
-
Tsui, Hoyee, van Kampen, Sebastiaan Johannes, Han, Su Ji, Meraviglia, Viviana, van Ham, Willem B., Casini, Simona, van der Kraak, Petra, Vink, Aryan, Yin, Xiaoke, Mayr, Manuel, Bossu, Alexandre, Marchal, Gerard A., Monshouwer-Kloots, Jantine, Eding, Joep, Versteeg, Danielle, de Ruiter, Hesther, Bezstarosti, Karel, Groeneweg, Judith, Klaasen, Sjoerd J., and van Laake, Linda W.
- Subjects
PROTEOLYSIS ,PROTEIN stability ,CARDIOMYOPATHIES ,ADHERENS junctions ,HEART diseases ,PROTEIN expression ,HEART fibrosis - Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited progressive cardiac disease. Many patients with ACM harbor mutations in desmosomal genes, predominantly in plakophilin-2 (PKP2). Although the genetic basis of ACM is well characterized, the underlying disease-driving mechanisms remain unresolved. Explanted hearts from patients with ACM had less PKP2 compared with healthy hearts, which correlated with reduced expression of desmosomal and adherens junction (AJ) proteins. These proteins were also disorganized in areas of fibrotic remodeling. In vitro data from human-induced pluripotent stem cell–derived cardiomyocytes and microtissues carrying the heterozygous PKP2 c.2013delC pathogenic mutation also displayed impaired contractility. Knockin mice carrying the equivalent heterozygous Pkp2 c.1755delA mutation recapitulated changes in desmosomal and AJ proteins and displayed cardiac dysfunction and fibrosis with age. Global proteomics analysis of 4-month-old heterozygous Pkp2 c.1755delA hearts indicated involvement of the ubiquitin-proteasome system (UPS) in ACM pathogenesis. Inhibition of the UPS in mutant mice increased area composita proteins and improved calcium dynamics in isolated cardiomyocytes. Additional proteomics analyses identified lysine ubiquitination sites on the desmosomal proteins, which were more ubiquitinated in mutant mice. In summary, we show that a plakophilin-2 mutation can lead to decreased desmosomal and AJ protein expression through a UPS-dependent mechanism, which preceded cardiac remodeling. These findings suggest that targeting protein degradation and improving desmosomal protein stability may be a potential therapeutic strategy for the treatment of ACM. PKPing up the rhythm: Arrhythmogenic cardiomyopathy (ACM) is a debilitating disease with clear genetic links but variable penetrance and poorly understood mechanisms. Here Tsui and colleagues analyzed heart tissue from patients with ACM along with inducible pluripotent stem cell–derived cardiomyocytes and mice carrying a plakophilin-2 (Pkp2) mutation to reveal that decreased expression of desmosomal and adherens junction proteins correlated with cardiac dysfunction and fibrosis. Proteomics data indicated involvement of the ubiquitin-proteasome system (UPS) in the degradation of these proteins, and inhibition of the UPS improved protein expression and calcium dynamics in isolated mutant cardiomyocytes. These results suggest that therapies aimed at increasing stability of desmosomal proteins may improve cardiac function in patients with ACM.--AW [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
4. Thymosin β4 and prothymosin α promote cardiac regeneration post-ischaemic injury in mice.
- Author
-
Gladka, Monika M, Johansen, Anne Katrine Z, Kampen, Sebastiaan J van, Peters, Marijn M C, Molenaar, Bas, Versteeg, Danielle, Kooijman, Lieneke, Zentilin, Lorena, Giacca, Mauro, and Rooij, Eva van
- Subjects
CARDIAC regeneration ,THYMOSIN ,GENE expression profiling ,MYOCARDIAL ischemia ,HEART diseases - Abstract
Aims The adult mammalian heart is a post-mitotic organ. Even in response to necrotic injuries, where regeneration would be essential to reinstate cardiac structure and function, only a minor percentage of cardiomyocytes undergo cytokinesis. The gene programme that promotes cell division within this population of cardiomyocytes is not fully understood. In this study, we aimed to determine the gene expression profile of proliferating adult cardiomyocytes in the mammalian heart after myocardial ischaemia, to identify factors to can promote cardiac regeneration. Methods and results Here, we demonstrate increased 5-ethynyl-2'deoxyuridine incorporation in cardiomyocytes 3 days post-myocardial infarction in mice. By applying multi-colour lineage tracing, we show that this is paralleled by clonal expansion of cardiomyocytes in the borderzone of the infarcted tissue. Bioinformatic analysis of single-cell RNA sequencing data from cardiomyocytes at 3 days post ischaemic injury revealed a distinct transcriptional profile in cardiomyocytes expressing cell cycle markers. Combinatorial overexpression of the enriched genes within this population in neonatal rat cardiomyocytes and mice at postnatal day 12 (P12) unveiled key genes that promoted increased cardiomyocyte proliferation. Therapeutic delivery of these gene cocktails into the myocardial wall after ischaemic injury demonstrated that a combination of thymosin beta 4 (TMSB4) and prothymosin alpha (PTMA) provide a permissive environment for cardiomyocyte proliferation and thereby attenuated cardiac dysfunction. Conclusion This study reveals the transcriptional profile of proliferating cardiomyocytes in the ischaemic heart and shows that overexpression of the two identified factors, TMSB4 and PTMA, can promote cardiac regeneration. This work indicates that in addition to activating cardiomyocyte proliferation, a supportive environment is a key for regeneration to occur. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
5. Spatial transcriptomics unveils ZBTB11 as a regulator of cardiomyocyte degeneration in arrhythmogenic cardiomyopathy.
- Author
-
Boogerd, Cornelis J, Lacraz, Grégory P A, Vértesy, Ábel, Kampen, Sebastiaan J van, Perini, Ilaria, Ruiter, Hesther de, Versteeg, Danielle, Brodehl, Andreas, van der Kraak, Petra, Giacca, Mauro, Jonge, Nicolaas de, Junker, Jan Philipp, Oudenaarden, Alexander van, Vink, Aryan, and Rooij, Eva van
- Subjects
CARDIOMYOPATHIES ,GENE expression profiling ,CARDIAC arrest ,ZINC-finger proteins ,GENE expression - Abstract
Aims Arrhythmogenic cardiomyopathy (ACM) is an inherited cardiac disorder that is characterized by progressive loss of myocardium that is replaced by fibro-fatty cells, arrhythmias, and sudden cardiac death. While myocardial degeneration and fibro-fatty replacement occur in specific locations, the underlying molecular changes remain poorly characterized. Here, we aim to delineate local changes in gene expression to identify new genes and pathways that are relevant for specific remodelling processes occurring during ACM. Methods and results Using Tomo-Seq, genome-wide transcriptional profiling with high spatial resolution, we created transmural epicardial-to-endocardial gene expression atlases of explanted ACM hearts to gain molecular insights into disease-driving processes. This enabled us to link gene expression profiles to the different regional remodelling responses and allowed us to identify genes that are potentially relevant for disease progression. In doing so, we identified distinct gene expression profiles marking regions of cardiomyocyte degeneration and fibro-fatty remodelling and revealed Zinc finger and BTB domain-containing protein 11 (ZBTB11) to be specifically enriched at sites of active fibro-fatty replacement of myocardium. Immunohistochemistry indicated ZBTB11 to be induced in cardiomyocytes flanking fibro-fatty areas, which could be confirmed in multiple cardiomyopathy patients. Forced overexpression of ZBTB11 induced autophagy and cell death-related gene programmes in human cardiomyocytes, leading to increased apoptosis. Conclusion Our study shows the power of Tomo-Seq to unveil new molecular mechanisms in human cardiomyopathy and uncovers ZBTB11 as a novel driver of cardiomyocyte loss. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
6. Ischemic tolerance and cardiac repair in the spiny mouse (Acomys).
- Author
-
Koopmans, Tim, van Beijnum, Henriette, Roovers, Elke F., Tomasso, Antonio, Malhotra, Divyanshu, Boeter, Jochem, Psathaki, Olympia E., Versteeg, Danielle, van Rooij, Eva, and Bartscherer, Kerstin
- Subjects
MYOCARDIAL ischemia ,CORONARY disease ,CAUSES of death ,MICE ,MYOCARDIAL infarction - Abstract
Ischemic heart disease and by extension myocardial infarction is the primary cause of death worldwide, warranting regenerative therapies to restore heart function. Current models of natural heart regeneration are restricted in that they are not of adult mammalian origin, precluding the study of class-specific traits that have emerged throughout evolution, and reducing translatability of research findings to humans. Here, we present the spiny mouse (Acomys spp.), a murid rodent that exhibits bona fide regeneration of the back skin and ear pinna, as a model to study heart repair. By comparing them to ordinary mice (Mus musculus), we show that the acute injury response in spiny mice is similar, but with an associated tolerance to infarction through superior survivability, improved ventricular conduction, and near-absence of pathological remodeling. Critically, spiny mice display increased vascularization, altered scar organization, and a more immature phenotype of cardiomyocytes, with a corresponding improvement in heart function. These findings present new avenues for mammalian heart research by leveraging unique tissue properties of the spiny mouse. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
7. Gene expression profiling of hypertrophic cardiomyocytes identifies new players in pathological remodelling.
- Author
-
Vigil-Garcia, Marta, Demkes, Charlotte J, Eding, Joep E C, Versteeg, Danielle, Ruiter, Hesther de, Perini, Ilaria, Kooijman, Lieneke, Gladka, Monika M, Asselbergs, Folkert W, Vink, Aryan, Harakalova, Magdalena, Bossu, Alexander, Veen, Toon A B van, Boogerd, Cornelis J, and Rooij, Eva van
- Subjects
GENE expression profiling ,CARDIAC hypertrophy ,HEART failure ,HEART cells ,GENE expression - Abstract
Aims Pathological cardiac remodelling is characterized by cardiomyocyte (CM) hypertrophy and fibroblast activation, which can ultimately lead to maladaptive hypertrophy and heart failure (HF). Genome-wide expression analysis on heart tissue has been instrumental for the identification of molecular mechanisms at play. However, these data were based on signals derived from all cardiac cell types. Here, we aimed for a more detailed view on molecular changes driving maladaptive CM hypertrophy to aid in the development of therapies to reverse pathological remodelling. Methods and results Utilizing CM-specific reporter mice exposed to pressure overload by transverse aortic banding and CM isolation by flow cytometry, we obtained gene expression profiles of hypertrophic CMs in the more immediate phase after stress, and CMs showing pathological hypertrophy. We identified subsets of genes differentially regulated and specific for either stage. Among the genes specifically up-regulated in the CMs during the maladaptive phase we found known stress markers, such as Nppb and Myh7 , but additionally identified a set of genes with unknown roles in pathological hypertrophy, including the platelet isoform of phosphofructokinase (PFKP). Norepinephrine-angiotensin II treatment of cultured human CMs induced the secretion of N-terminal-pro-B-type natriuretic peptide (NT-pro-BNP) and recapitulated the up-regulation of these genes, indicating conservation of the up-regulation in failing CMs. Moreover, several genes induced during pathological hypertrophy were also found to be increased in human HF, with their expression positively correlating to the known stress markers NPPB and MYH7. Mechanistically, suppression of Pfkp in primary CMs attenuated stress-induced gene expression and hypertrophy, indicating that Pfkp is an important novel player in pathological remodelling of CMs. Conclusion Using CM-specific transcriptomic analysis, we identified novel genes induced during pathological hypertrophy that are relevant for human HF, and we show that PFKP is a conserved failure-induced gene that can modulate the CM stress response. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
8. Single-cell transcriptomics following ischemic injury identifies a role for B2M in cardiac repair.
- Author
-
Molenaar, Bas, Timmer, Louk T., Droog, Marjolein, Perini, Ilaria, Versteeg, Danielle, Kooijman, Lieneke, Monshouwer-Kloots, Jantine, de Ruiter, Hesther, Gladka, Monika M., and van Rooij, Eva
- Subjects
MICROGLOBULINS ,HEART cells ,HEART failure ,FIBROBLASTS ,DATA analysis - Abstract
The efficiency of the repair process following ischemic cardiac injury is a crucial determinant for the progression into heart failure and is controlled by both intra- and intercellular signaling within the heart. An enhanced understanding of this complex interplay will enable better exploitation of these mechanisms for therapeutic use. We used single-cell transcriptomics to collect gene expression data of all main cardiac cell types at different time-points after ischemic injury. These data unveiled cellular and transcriptional heterogeneity and changes in cellular function during cardiac remodeling. Furthermore, we established potential intercellular communication networks after ischemic injury. Follow up experiments confirmed that cardiomyocytes express and secrete elevated levels of beta-2 microglobulin in response to ischemic damage, which can activate fibroblasts in a paracrine manner. Collectively, our data indicate phase-specific changes in cellular heterogeneity during different stages of cardiac remodeling and allow for the identification of therapeutic targets relevant for cardiac repair. Molenaar et al. use scRNA-seq to profile cardiac cell gene expression changes at three time points following ischemic injury. They observe that B2M is secreted by cardiomyocytes following injury, promoting scar formation. This data may be useful in finding therapeutic targets for cardiac repair [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
9. Cardiomyocytes stimulate angiogenesis after ischemic injury in a ZEB2-dependent manner.
- Author
-
Gladka, Monika M., Kohela, Arwa, Molenaar, Bas, Versteeg, Danielle, Kooijman, Lieneke, Monshouwer-Kloots, Jantine, Kremer, Veerle, Vos, Harmjan R., Huibers, Manon M. H., Haigh, Jody J., Huylebroeck, Danny, Boon, Reinier A., Giacca, Mauro, and van Rooij, Eva
- Subjects
ZINC-finger proteins ,HEART diseases ,MYOCARDIAL reperfusion ,THYMOSIN ,TRANSCRIPTION factors ,CELL migration ,NEOVASCULARIZATION - Abstract
The disruption in blood supply due to myocardial infarction is a critical determinant for infarct size and subsequent deterioration in function. The identification of factors that enhance cardiac repair by the restoration of the vascular network is, therefore, of great significance. Here, we show that the transcription factor Zinc finger E-box-binding homeobox 2 (ZEB2) is increased in stressed cardiomyocytes and induces a cardioprotective cross-talk between cardiomyocytes and endothelial cells to enhance angiogenesis after ischemia. Single-cell sequencing indicates ZEB2 to be enriched in injured cardiomyocytes. Cardiomyocyte-specific deletion of ZEB2 results in impaired cardiac contractility and infarct healing post-myocardial infarction (post-MI), while cardiomyocyte-specific ZEB2 overexpression improves cardiomyocyte survival and cardiac function. We identified Thymosin β4 (TMSB4) and Prothymosin α (PTMA) as main paracrine factors released from cardiomyocytes to stimulate angiogenesis by enhancing endothelial cell migration, and whose regulation is validated in our in vivo models. Therapeutic delivery of ZEB2 to cardiomyocytes in the infarcted heart induces the expression of TMSB4 and PTMA, which enhances angiogenesis and prevents cardiac dysfunction. These findings reveal ZEB2 as a beneficial factor during ischemic injury, which may hold promise for the identification of new therapies. ZEB2 transcription factor is increased in a subset of cardiomyocytes during stress to induce cardioprotective effects after injury. Here the authors show that therapeutic delivery of ZEB2 prevents cardiac dysfunction after ischemic damage and promotes the activation of pro-angiogenic signals. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
10. Profiling proliferative cells and their progeny in damaged murine hearts.
- Author
-
Kretzschmar, Kai, Post, Yorick, Bannier-Hélaouët, Marie, Mattiotti, Andrea, Drost, Jarno, Basak, Onur, Li, Vivian S. W., van den Born, Maaike, Gunst, Quinn D., Versteeg, Danielle, Kooijman, Lieneke, van der Elst, Stefan, van Es, Johan H., van Rooij, Eva, van den Hoff, Maurice J. B., and Clevers, Hans
- Subjects
CELL proliferation ,MYOCARDIAL infarction ,HEART physiology ,HEART cells ,STEM cells ,FIBROBLASTS ,TRANSCRIPTOMES ,RODENT diseases ,MAMMALS - Abstract
The significance of cardiac stem cell (CSC) populations for cardiac regeneration remains disputed. Here, we apply the most direct definition of stem cell function (the ability to replace lost tissue through cell division) to interrogate the existence of CSCs. By singlecell mRNA sequencing and genetic lineage tracing using two Ki67 knockin mouse models, we map all proliferating cells and their progeny in homoeostatic and regenerating murine hearts. Cycling cardiomyocytes were only robustly observed in the early postnatal growth phase, while cycling cells in homoeostatic and damaged adultmyocardium represented various noncardiomyocyte cell types. Proliferative postdamage fibroblasts expressing follistatin-like protein 1 (FSTL1) closely resemble neonatal cardiac fibroblasts and form the fibrotic scar. Genetic deletion of Fstl1 in cardiac fibroblasts results in postdamage cardiac rupture. We find no evidence for the existence of a quiescent CSC population, for transdifferentiation of other cell types toward cardiomyocytes, or for proliferation of significant numbers of cardiomyocytes in response to cardiac injury. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
11. Single-Cell Sequencing of the Healthy and Diseased Heart Reveals Cytoskeleton-Associated Protein 4 as a New Modulator of Fibroblasts Activation.
- Author
-
Gladka, Monika M., Molenaar, Bas, de Ruiter, Hesther, van der Elst, Stefan, Tsui, Hoyee, Versteeg, Danielle, Lacraz, Grègory P.A., Huibers, Manon M.H., van Oudenaarden, Alexander, van Rooij, Eva, and Lacraz, Grègory P A
- Published
- 2018
- Full Text
- View/download PDF
12. Postnatal Cardiac Gene Editing Using CRISPR/Cas9 With AAV9-Mediated Delivery of Short Guide RNAs Results in Mosaic Gene Disruption.
- Author
-
Johansen, Anne Katrine, Molenaar, Bas, Versteeg, Danielle, Leitoguinho, Ana Rita, Demkes, Charlotte, Spanjaard, Bastiaan, de Ruiter, Hesther, Moqadam, Farhad Akbari, Kooijman, Lieneke, Zentilin, Lorena, Giacca, Mauro, and van Rooij, Eva
- Published
- 2017
- Full Text
- View/download PDF
13. Tomo-Seq Identifies SOX9 as a Key Regulator of Cardiac Fibrosis During Ischemic Injury.
- Author
-
Lacraz, Grégory P. A., Junker, Jan Philipp, Gladka, Monika M., Molenaar, Bas, Scholman, Koen T., Vigil-Garcia, Marta, Versteeg, Danielle, de Ruiter, Hesther, Vermunt, Marit W., Creyghton, Menno P., Huibers, Manon M.H., de Jonge, Nicolaas, van Oudenaarden, Alexander, and van Rooij, Eva
- Published
- 2017
- Full Text
- View/download PDF
14. Abstract 16614: Single-Cell RNA Sequencing Reveals ZEB2 as a Cardioprotective Factor After Ischemic Injury.
- Author
-
Gladka, Monika M, Kohela, Arwa, Molenaar, Bas, Versteeg, Danielle, Kooijman, Lieneke, Groenewold, Vincent, Vos, Harmjan R, Huibers, Manon M, Huylebroeck, Danny, Giacca, Mauro, and van Rooij, Eva
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.