Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sebastian Schafer is active.

Publication


Featured researches published by Sebastian Schafer.


Nature Medicine | 2012

RBM20 , a gene for hereditary cardiomyopathy, regulates titin splicing

Wei Guo; Sebastian Schafer; Marion L. Greaser; Michael H. Radke; Martin Liss; Thirupugal Govindarajan; Henrike Maatz; Herbert Schulz; Shijun Li; Amanda M. Parrish; Vita Dauksaite; Padmanabhan Vakeel; Sabine Klaassen; Brenda Gerull; Ludwig Thierfelder; Vera Regitz-Zagrosek; Timothy A. Hacker; Kurt W. Saupe; G. William Dec; Patrick T. Ellinor; Calum A. MacRae; Bastian Spallek; Robert S. Fischer; Andreas Perrot; Cemil Özcelik; Kathrin Saar; Norbert Hubner; Michael Gotthardt

Alternative splicing has a major role in cardiac adaptive responses, as exemplified by the isoform switch of the sarcomeric protein titin, which adjusts ventricular filling. By positional cloning using a previously characterized rat strain with altered titin mRNA splicing, we identified a loss-of-function mutation in the gene encoding RNA binding motif protein 20 (Rbm20) as the underlying cause of pathological titin isoform expression. The phenotype of Rbm20-deficient rats resembled the pathology seen in individuals with dilated cardiomyopathy caused by RBM20 mutations. Deep sequencing of the human and rat cardiac transcriptome revealed an RBM20-dependent regulation of alternative splicing. In addition to titin (TTN), we identified a set of 30 genes with conserved splicing regulation between humans and rats. This network is enriched for genes that have previously been linked to cardiomyopathy, ion homeostasis and sarcomere biology. Our studies emphasize the key role of post-transcriptional regulation in cardiac function and provide mechanistic insights into the pathogenesis of human heart failure.


Science | 2015

Titin Mutations in iPS cells Define Sarcomere Insufficiency as a Cause of Dilated Cardiomyopathy

John T. Hinson; Anant Chopra; N. Nafissi; William J. Polacheck; Craig C. Benson; Sandra Swist; Joshua M. Gorham; Luhan Yang; Sebastian Schafer; Calvin C. Sheng; Alireza Haghighi; Jason Homsy; Norbert Hubner; George M. Church; Stuart A. Cook; Wolfgang A. Linke; Christopher S. Chen; Jonathan G. Seidman; Christine E. Seidman

A giant disruption of the heart Certain forms of heart failure originate from genetic mutations. Understanding how the culprit mutant proteins alter normal heart function could lead to more effective treatments. One candidate is the giant protein tintin, which is mutated in a subset of patients with dilated cardiomyopathy. Through a combination of patient-derived stem cells, tissue engineering, and gene editing, Hinson et al. found that disease-associated titin mutations disrupt the function of the contractile unit in heart muscle. As a result, the heart does not respond properly to mechanical and other forms of stress. Science, this issue p. 982 Mutations in titin cause heart disease by disrupting the sarcomere, which normally helps the heart adapt to stress. Human mutations that truncate the massive sarcomere protein titin [TTN-truncating variants (TTNtvs)] are the most common genetic cause for dilated cardiomyopathy (DCM), a major cause of heart failure and premature death. Here we show that cardiac microtissues engineered from human induced pluripotent stem (iPS) cells are a powerful system for evaluating the pathogenicity of titin gene variants. We found that certain missense mutations, like TTNtvs, diminish contractile performance and are pathogenic. By combining functional analyses with RNA sequencing, we explain why truncations in the A-band domain of TTN cause DCM, whereas truncations in the I band are better tolerated. Finally, we demonstrate that mutant titin protein in iPS cell–derived cardiomyocytes results in sarcomere insufficiency, impaired responses to mechanical and β-adrenergic stress, and attenuated growth factor and cell signaling activation. Our findings indicate that titin mutations cause DCM by disrupting critical linkages between sarcomerogenesis and adaptive remodeling.


Blood | 2011

Engineered cell homing

Debanjan Sarkar; Joel A. Spencer; Joseph A. Phillips; Weian Zhao; Sebastian Schafer; Dawn P. Spelke; Luke J. Mortensen; Juan P. Ruiz; Praveen Kumar Vemula; Rukmani Sridharan; Sriram Kumar; Rohit Karnik; Charles P. Lin; Jeffrey M. Karp

One of the greatest challenges in cell therapy is to minimally invasively deliver a large quantity of viable cells to a tissue of interest with high engraftment efficiency. Low and inefficient homing of systemically delivered mesenchymal stem cells (MSCs), for example, is thought to be a major limitation of existing MSC-based therapeutic approaches, caused predominantly by inadequate expression of cell surface adhesion receptors. Using a platform approach that preserves the MSC phenotype and does not require genetic manipulation, we modified the surface of MSCs with a nanometer-scale polymer construct containing sialyl Lewis(x) (sLe(x)) that is found on the surface of leukocytes and mediates cell rolling within inflamed tissue. The sLe(x) engineered MSCs exhibited a robust rolling response on inflamed endothelium in vivo and homed to inflamed tissue with higher efficiency compared with native MSCs. The modular approach described herein offers a simple method to potentially target any cell type to specific tissues via the circulation.


Science Translational Medicine | 2015

Integrated allelic, transcriptional, and phenomic dissection of the cardiac effects of titin truncations in health and disease

Angharad M. Roberts; James S. Ware; Daniel S. Herman; Sebastian Schafer; John Baksi; Alexander G. Bick; Rachel Buchan; Roddy Walsh; Shibu John; Samuel Wilkinson; Francesco Mazzarotto; Leanne E. Felkin; Sungsam Gong; Jacqueline A. L. MacArthur; Fiona Cunningham; Jason Flannick; Stacey B. Gabriel; David Altshuler; P. Macdonald; Matthias Heinig; Anne Keogh; Christopher S. Hayward; Nicholas R. Banner; Dudley J. Pennell; Declan P. O’Regan; Tan Ru San; Antonio de Marvao; Timothy Dawes; Ankur Gulati; Emma J. Birks

Truncating variants of the giant protein titin cause dilated cardiomyopathy when they occur toward the protein’s carboxyl terminus and in highly expressed exons. What Happens When Titins Are Trimmed? The most common form of inherited heart failure, dilated cardiomyopathy, can be caused by mutations in a mammoth heart protein, appropriately called titin. Now, Roberts et al. sort out which titin mutations cause disease and why some people can carry certain titin mutations but remain perfectly healthy. In an exhaustive survey of more than 5200 people, with and without cardiomyopathy, the authors sequenced the titin gene and measured its corresponding RNA and protein levels. The alterations in titin were truncating mutations, which cause short nonfunctional versions of the RNA or protein. These defects produced cardiomyopathy when they occurred closer to the protein’s carboxyl terminus and in exons that were abundantly transcribed. The titin-truncating mutations that occur in the general population tended not to have these characteristics and were usually benign. This new detailed understanding of the molecular basis of dilated cardiomyopathy penetrance will promote better disease management and accelerate rational patient stratification. The recent discovery of heterozygous human mutations that truncate full-length titin (TTN, an abundant structural, sensory, and signaling filament in muscle) as a common cause of end-stage dilated cardiomyopathy (DCM) promises new prospects for improving heart failure management. However, realization of this opportunity has been hindered by the burden of TTN-truncating variants (TTNtv) in the general population and uncertainty about their consequences in health or disease. To elucidate the effects of TTNtv, we coupled TTN gene sequencing with cardiac phenotyping in 5267 individuals across the spectrum of cardiac physiology and integrated these data with RNA and protein analyses of human heart tissues. We report diversity of TTN isoform expression in the heart, define the relative inclusion of TTN exons in different isoforms (using the TTN transcript annotations available at http://cardiodb.org/titin), and demonstrate that these data, coupled with the position of the TTNtv, provide a robust strategy to discriminate pathogenic from benign TTNtv. We show that TTNtv is the most common genetic cause of DCM in ambulant patients in the community, identify clinically important manifestations of TTNtv-positive DCM, and define the penetrance and outcomes of TTNtv in the general population. By integrating genetic, transcriptome, and protein analyses, we provide evidence for a length-dependent mechanism of disease. These data inform diagnostic criteria and management strategies for TTNtv-positive DCM patients and for TTNtv that are identified as incidental findings.


American Journal of Human Genetics | 2013

Fine Mapping of the 1p36 Deletion Syndrome Identifies Mutation of PRDM16 as a Cause of Cardiomyopathy

Anne-Karin Arndt; Sebastian Schafer; Jörg-Detlef Drenckhahn; M. Khaled Sabeh; Eva Plovie; Almuth Caliebe; Eva Klopocki; Gabriel Musso; Andreas A. Werdich; Hermann Kalwa; Matthias Heinig; Robert F. Padera; Katharina Wassilew; Julia Bluhm; Christine Harnack; Janine Martitz; Paul J.R. Barton; Matthias Greutmann; Felix Berger; Norbert Hubner; Reiner Siebert; Hans-Heiner Kramer; Stuart A. Cook; Calum A. MacRae; Sabine Klaassen

Deletion 1p36 syndrome is recognized as the most common terminal deletion syndrome. Here, we describe the loss of a gene within the deletion that is responsible for the cardiomyopathy associated with monosomy 1p36, and we confirm its role in nonsyndromic left ventricular noncompaction cardiomyopathy (LVNC) and dilated cardiomyopathy (DCM). With our own data and publically available data from array comparative genomic hybridization (aCGH), we identified a minimal deletion for the cardiomyopathy associated with 1p36del syndrome that included only the terminal 14 exons of the transcription factor PRDM16 (PR domain containing 16), a gene that had previously been shown to direct brown fat determination and differentiation. Resequencing of PRDM16 in a cohort of 75 nonsyndromic individuals with LVNC detected three mutations, including one truncation mutant, one frameshift null mutation, and a single missense mutant. In addition, in a series of cardiac biopsies from 131 individuals with DCM, we found 5 individuals with 4 previously unreported nonsynonymous variants in the coding region of PRDM16. None of the PRDM16 mutations identified were observed in more than 6,400 controls. PRDM16 has not previously been associated with cardiac disease but is localized in the nuclei of cardiomyocytes throughout murine and human development and in the adult heart. Modeling of PRDM16 haploinsufficiency and a human truncation mutant in zebrafish resulted in both contractile dysfunction and partial uncoupling of cardiomyocytes and also revealed evidence of impaired cardiomyocyte proliferative capacity. In conclusion, mutation of PRDM16 causes the cardiomyopathy in 1p36 deletion syndrome as well as a proportion of nonsyndromic LVNC and DCM.


Cell Reports | 2013

Quantitative and Qualitative Proteome Characteristics Extracted from In-Depth Integrated Genomics and Proteomics Analysis

Teck Yew Low; Sebastiaan van Heesch; Henk van den Toorn; Piero Giansanti; Alba Cristobal; Pim W. Toonen; Sebastian Schafer; Norbert Hubner; Bas van Breukelen; Shabaz Mohammed; Edwin Cuppen; Albert J. R. Heck; Victor Guryev

Quantitative and qualitative protein characteristics are regulated at genomic, transcriptomic, and posttranscriptional levels. Here, we integrated in-depth transcriptome and proteome analyses of liver tissues from two rat strains to unravel the interactions within and between these layers. We obtained peptide evidence for 26,463 rat liver proteins. We validated 1,195 gene predictions, 83 splice events, 126 proteins with nonsynonymous variants, and 20 isoforms with nonsynonymous RNA editing. Quantitative RNA sequencing and proteomics data correlate highly between strains but poorly among each other, indicating extensive nongenetic regulation. Our multilevel analysis identified a genomic variant in the promoter of the most differentially expressed gene Cyp17a1, a previously reported top hit in genome-wide association studies for human hypertension, as a potential contributor to the hypertension phenotype in SHR rats. These results demonstrate the power of and need for integrative analysis for understanding genetic control of molecular dynamics and phenotypic diversity in a system-wide manner.


Human Molecular Genetics | 2014

Recessive TTN truncating mutations define novel forms of core myopathy with heart disease

C. Chauveau; Carsten G. Bönnemann; C. Julien; Ay Lin Kho; H. Marks; Beril Talim; P. Maury; M.C. Arne-Bes; Emmanuelle Uro-Coste; Alexander Alexandrovich; Anna Vihola; Sebastian Schafer; B. Kaufmann; L. Medne; Norbert Hubner; A.R. Foley; Mariarita Santi; Bjarne Udd; Haluk Topaloglu; Steven A. Moore; Michael Gotthardt; M.E. Samuels; Mathias Gautel; Ana Ferreiro

Core myopathies (CM), the main non-dystrophic myopathies in childhood, remain genetically unexplained in many cases. Heart disease is not considered part of the typical CM spectrum. No congenital heart defect has been reported, and childhood-onset cardiomyopathy has been documented in only two CM families with homozygous mutations of the TTN gene. TTN encodes titin, a giant protein of striated muscles. Recently, heterozygous TTN truncating mutations have also been reported as a major cause of dominant dilated cardiomyopathy. However, relatively few TTN mutations and phenotypes are known, and titin pathophysiological role in cardiac and skeletal muscle conditions is incompletely understood. We analyzed a series of 23 families with congenital CM and primary heart disease using TTN M-line-targeted sequencing followed in selected patients by whole-exome sequencing and functional studies. We identified seven novel homozygous or compound heterozygous TTN mutations (five in the M-line, five truncating) in 17% patients. Heterozygous parents were healthy. Phenotype analysis identified four novel titinopathies, including cardiac septal defects, left ventricular non-compaction, Emery-Dreifuss muscular dystrophy or arthrogryposis. Additionally, in vitro studies documented the first-reported absence of a functional titin kinase domain in humans, leading to a severe antenatal phenotype. We establish that CM are associated with a large range of heart conditions of which TTN mutations are a major cause, thereby expanding the TTN mutational and phenotypic spectrum. Additionally, our results suggest titin kinase implication in cardiac morphogenesis and demonstrate that heterozygous TTN truncating mutations may not manifest unless associated with a second mutation, reassessing the paradigm of their dominant expression.


Nature Genetics | 2017

Titin-truncating variants affect heart function in disease cohorts and the general population

Sebastian Schafer; Antonio de Marvao; Eleonora Adami; Lorna R. Fiedler; Benjamin Ng; Ester Khin; Owen J. L. Rackham; Sebastiaan van Heesch; Chee Jian Pua; Miao Kui; Roddy Walsh; Upasana Tayal; Sanjay Prasad; Timothy Dawes; Nicole Shi Jie Ko; David Sim; Laura Lihua Chan; Calvin Chin; Francesco Mazzarotto; Paul J.R. Barton; Franziska Kreuchwig; Dominique P.V. de Kleijn; Teresa Totman; Carlo Biffi; Nicole Tee; Daniel Rueckert; Valentin Schneider; Allison Faber; Vera Regitz-Zagrosek; Jonathan G. Seidman

Titin-truncating variants (TTNtv) commonly cause dilated cardiomyopathy (DCM). TTNtv are also encountered in ∼1% of the general population, where they may be silent, perhaps reflecting allelic factors. To better understand TTNtv, we integrated TTN allelic series, cardiac imaging and genomic data in humans and studied rat models with disparate TTNtv. In patients with DCM, TTNtv throughout titin were significantly associated with DCM. Ribosomal profiling in rat showed the translational footprint of premature stop codons in Ttn, TTNtv-position-independent nonsense-mediated degradation of the mutant allele and a signature of perturbed cardiac metabolism. Heart physiology in rats with TTNtv was unremarkable at baseline but became impaired during cardiac stress. In healthy humans, machine-learning-based analysis of high-resolution cardiac imaging showed TTNtv to be associated with eccentric cardiac remodeling. These data show that TTNtv have molecular and physiological effects on the heart across species, with a continuum of expressivity in health and disease.


Cardiovascular Research | 2016

A mutation in the glutamate-rich region of RNA-binding motif protein 20 causes dilated cardiomyopathy through missplicing of titin and impaired Frank–Starling mechanism

Abdelaziz Beqqali; Ilse A. E. Bollen; T.B. Rasmussen; Maarten M.G. van den Hoogenhof; Hanneke W. M. van Deutekom; Sebastian Schafer; Jan Haas; Benjamin Meder; Keld E. Sørensen; Ralph J. van Oort; Jens Mogensen; Norbert Hubner; Esther E. Creemers; Jolanda van der Velden; Yigal M. Pinto

AIM Mutations in the RS-domain of RNA-binding motif protein 20 (RBM20) have recently been identified to segregate with aggressive forms of familial dilated cardiomyopathy (DCM). Loss of RBM20 in rats results in missplicing of the sarcomeric gene titin (TTN). The functional and physiological consequences of RBM20 mutations outside the mutational hotspot of RBM20 have not been explored to date. In this study, we investigated the pathomechanism of DCM caused by a novel RBM20 mutation in human cardiomyocytes. METHODS AND RESULTS We identified a family with DCM carrying a mutation (RBM20(E913K/+)) in a glutamate-rich region of RBM20. Western blot analysis of endogenous RBM20 protein revealed strongly reduced protein levels in the heart of an RBM20(E913K/+ )carrier. RNA deep-sequencing demonstrated massive inclusion of exons coding for the spring region of titin in the RBM20(E913K/+ )carrier. Titin isoform analysis revealed a dramatic shift from the less compliant N2B towards the highly compliant N2BA isoforms in RBM20(E913K/+ )heart. Moreover, an increased sarcomere resting-length was observed in single cardiomyocytes and isometric force measurements revealed an attenuated Frank-Starling mechanism (FSM), which was rescued by protein kinase A treatment. CONCLUSION A mutation outside the mutational hotspot of RBM20 results in haploinsufficiency of RBM20. This leads to disturbed alternative splicing of TTN, resulting in a dramatic shift to highly compliant titin isoforms and an impaired FSM. These effects may contribute to the early onset, and malignant course of DCM caused by RBM20 mutations. Altogether, our results demonstrate that heterozygous loss of RBM20 suffices to profoundly impair myocyte biomechanics by its disturbance of TTN splicing.


The FASEB Journal | 2011

Mimicking the inflammatory cell adhesion cascade by nucleic acid aptamer programmed cell-cell interactions

Weian Zhao; Weili Loh; Ilia A. Droujinine; Weisuong Teo; Namit Kumar; Sebastian Schafer; Cheryl H. Cui; Liang Zhang; Debanjan Sarkar; Rohit Karnik; Jeffrey M. Karp

Nature has evolved effective cell adhesion mechanisms to deliver inflammatory cells to inflamed tissue; however, many culture‐expanded therapeutic cells are incapable of targeting diseased tissues following systemic infusion, which represents a great challenge in cell therapy. Our aim was to develop simple approaches to program cell‐cell interactions that would otherwise not exist toward cell targeting and understanding the complex biology of cell‐cell interactions. We employed a chemistry approach to engineer P‐ or L‐selectin binding nucleic acid aptamers onto mesenchymal stem cells (MSCs) to enable them to engage inflamed endothelial cells and leukocytes, respectively. We show for the first time that engineered cells with a single artificial adhesion ligand can recapitulate 3 critical cell interactions in the inflammatory cell adhesion cascade under dynamic flow conditions. Aptamer‐engineered MSCs adhered on respective selectin surfaces under static conditions >10 times more efficiently than controls including scrambled‐DNA modified MSCs. Significantly, engineered MSCs can be directly captured from the flow stream by selectin surfaces or selectin‐expressing cells under flow conditions (≤2dyn/cm2). The simple chemistry approach and the versatility of aptamers permit the concept of engineered cell‐cell interactions to be generically applicable for targeting cells to diseased tissues and elucidating the biology of cell‐cell interactions.—Zhao, W., Loh, W., Droujinine, I. A., Teo, W., Kumar, N., Schafer, S., Cui, C. H., Zhang, L., Sarkar, D., Karnik, R., Karp, J. M. Mimicking the inflammatory cell adhesion cascade by nucleic acid aptamer programmed cell‐cell interactions. FASEB J. 25, 3045–3056 (2011). www.fasebj.org

Collaboration


Dive into the Sebastian Schafer's collaboration.

Top Co-Authors

Avatar

Norbert Hubner

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar

Stuart A. Cook

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eleonora Adami

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar

Carola Rintisch

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michal Pravenec

Academy of Sciences of the Czech Republic

View shared research outputs
Top Co-Authors

Avatar

Anissa Widjaja

National University of Singapore

View shared research outputs
Researchain Logo
Decentralizing Knowledge