Network


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

Hotspot


Dive into the research topics where Sven Schuierer is active.

Publication


Featured researches published by Sven Schuierer.


Nature Chemical Biology | 2015

SMN2 splice modulators enhance U1–pre-mRNA association and rescue SMA mice

James Palacino; Susanne E Swalley; Cheng Song; Atwood Cheung; Lei Shu; Xiaolu Zhang; Mailin Van Hoosear; Youngah Shin; Donovan Noel Chin; Caroline Gubser Keller; Martin Beibel; Nicole A. Renaud; Thomas M Smith; Michael Salcius; Xiaoying Shi; Marc Hild; Rebecca Servais; Monish Jain; Lin Deng; Caroline Bullock; Michael McLellan; Sven Schuierer; Leo Murphy; Marcel J J Blommers; Cecile Blaustein; Frada Berenshteyn; Arnaud Lacoste; Jason R Thomas; Guglielmo Roma; Gregory A. Michaud

Spinal muscular atrophy (SMA), which results from the loss of expression of the survival of motor neuron-1 (SMN1) gene, represents the most common genetic cause of pediatric mortality. A duplicate copy (SMN2) is inefficiently spliced, producing a truncated and unstable protein. We describe herein a potent, orally active, small-molecule enhancer of SMN2 splicing that elevates full-length SMN protein and extends survival in a severe SMA mouse model. We demonstrate that the molecular mechanism of action is via stabilization of the transient double-strand RNA structure formed by the SMN2 pre-mRNA and U1 small nuclear ribonucleic protein (snRNP) complex. The binding affinity of U1 snRNP to the 5 splice site is increased in a sequence-selective manner, discrete from constitutive recognition. This new mechanism demonstrates the feasibility of small molecule-mediated, sequence-selective splice modulation and the potential for leveraging this strategy in other splicing diseases.


Cell Host & Microbe | 2012

Selective and Specific Inhibition of the Plasmodium falciparum Lysyl-tRNA Synthetase by the Fungal Secondary Metabolite Cladosporin

Dominic Hoepfner; Case W. McNamara; Chek Shik Lim; Christian Studer; Ralph Riedl; Thomas Aust; Susan McCormack; David Plouffe; Stephan Meister; Sven Schuierer; Uwe Plikat; Nicole Hartmann; Frank Staedtler; Simona Cotesta; Esther K. Schmitt; Frank Petersen; Frantisek Supek; Richard Glynne; John A. Tallarico; Jeffrey A. Porter; Mark C. Fishman; Christophe Bodenreider; Thierry T. Diagana; N. Rao Movva; Elizabeth A. Winzeler

Summary With renewed calls for malaria eradication, next-generation antimalarials need be active against drug-resistant parasites and efficacious against both liver- and blood-stage infections. We screened a natural product library to identify inhibitors of Plasmodium falciparum blood- and liver-stage proliferation. Cladosporin, a fungal secondary metabolite whose target and mechanism of action are not known for any species, was identified as having potent, nanomolar, antiparasitic activity against both blood and liver stages. Using postgenomic methods, including a yeast deletion strains collection, we show that cladosporin specifically inhibits protein synthesis by directly targeting P. falciparum cytosolic lysyl-tRNA synthetase. Further, cladosporin is >100-fold more potent against parasite lysyl-tRNA synthetase relative to the human enzyme, which is conferred by the identity of two amino acids within the enzyme active site. Our data indicate that lysyl-tRNA synthetase is an attractive, druggable, antimalarial target that can be selectively inhibited.


Microbiological Research | 2014

High-resolution chemical dissection of a model eukaryote reveals targets, pathways and gene functions

Dominic Hoepfner; Stephen B. Helliwell; Heather Sadlish; Sven Schuierer; Ireos Filipuzzi; Sophie Brachat; Bhupinder Bhullar; Uwe Plikat; Yann Abraham; Marc Altorfer; Thomas Aust; Lukas Baeriswyl; Raffaele Cerino; Lena Chang; David Estoppey; Juerg Eichenberger; Mathias Frederiksen; Nicole Hartmann; Annika Hohendahl; Britta Knapp; Philipp Krastel; Nicolas Melin; Florian Nigsch; Virginie Petitjean; Frank Petersen; Ralph Riedl; Esther K. Schmitt; Frank Staedtler; Christian Studer; John A. Tallarico

Due to evolutionary conservation of biology, experimental knowledge captured from genetic studies in eukaryotic model organisms provides insight into human cellular pathways and ultimately physiology. Yeast chemogenomic profiling is a powerful approach for annotating cellular responses to small molecules. Using an optimized platform, we provide the relative sensitivities of the heterozygous and homozygous deletion collections for nearly 1800 biologically active compounds. The data quality enables unique insights into pathways that are sensitive and resistant to a given perturbation, as demonstrated with both known and novel compounds. We present examples of novel compounds that inhibit the therapeutically relevant fatty acid synthase and desaturase (Fas1p and Ole1p), and demonstrate how the individual profiles facilitate hypothesis-driven experiments to delineate compound mechanism of action. Importantly, the scale and diversity of tested compounds yields a dataset where the number of modulated pathways approaches saturation. This resource can be used to map novel biological connections, and also identify functions for unannotated genes. We validated hypotheses generated by global two-way hierarchical clustering of profiles for (i) novel compounds with a similar mechanism of action acting upon microtubules or vacuolar ATPases, and (ii) an un-annotated ORF, YIL060w, that plays a role in respiration in the mitochondria. Finally, we identify and characterize background mutations in the widely used yeast deletion collection which should improve the interpretation of past and future screens throughout the community. This comprehensive resource of cellular responses enables the expansion of our understanding of eukaryotic pathway biology.


ACS Chemical Biology | 2013

Evidence for a Functionally Relevant Rocaglamide Binding Site on the eIF4A–RNA Complex

Heather Sadlish; Gabriela Galicia-Vázquez; C. Gregory Paris; Thomas Aust; Bhupinder Bhullar; Lena Chang; Stephen B. Helliwell; Dominic Hoepfner; Britta Knapp; Ralph Riedl; Silvio Roggo; Sven Schuierer; Christian Studer; John A. Porco; Jerry Pelletier; N. Rao Movva

Translation initiation is an emerging target in oncology and neurobiology indications. Naturally derived and synthetic rocaglamide scaffolds have been used to interrogate this pathway; however, there is uncertainty regarding their precise mechanism(s) of action. We exploited the genetic tractability of yeast to define the primary effect of both a natural and a synthetic rocaglamide in a cellular context and characterized the molecular target using biochemical studies and in silico modeling. Chemogenomic profiling and mutagenesis in yeast identified the eIF (eukaryotic Initiation Factor) 4A helicase homologue as the primary molecular target of rocaglamides and defined a discrete set of residues near the RNA binding motif that confer resistance to both compounds. Three of the eIF4A mutations were characterized regarding their functional consequences on activity and response to rocaglamide inhibition. These data support a model whereby rocaglamides stabilize an eIF4A-RNA interaction to either alter the level and/or impair the activity of the eIF4F complex. Furthermore, in silico modeling supports the annotation of a binding pocket delineated by the RNA substrate and the residues identified from our mutagenesis screen. As expected from the high degree of conservation of the eukaryotic translation pathway, these observations are consistent with previous observations in mammalian model systems. Importantly, we demonstrate that the chemically distinct silvestrol and synthetic rocaglamides share a common mechanism of action, which will be critical for optimization of physiologically stable derivatives. Finally, these data confirm the value of the rocaglamide scaffold for exploring the impact of translational modulation on disease.


Journal of Cell Science | 2015

Decatransin, a new natural product inhibiting protein translocation at the Sec61/SecYEG translocon

Tina Junne; Joanne Wong; Christian Studer; Thomas Aust; Benedikt W. Bauer; Martin Beibel; Bhupinder Bhullar; Robert Bruccoleri; Jürg Eichenberger; David Estoppey; Nicole Hartmann; Britta Knapp; Philipp Krastel; Nicolas Melin; Lukas Oberer; Ralph Riedl; Guglielmo Roma; Sven Schuierer; Frank Petersen; John A. Tallarico; Martin Spiess; Dominic Hoepfner

ABSTRACT A new cyclic decadepsipeptide was isolated from Chaetosphaeria tulasneorum with potent bioactivity on mammalian and yeast cells. Chemogenomic profiling in S. cerevisiae indicated that the Sec61 translocon complex, the machinery for protein translocation and membrane insertion at the endoplasmic reticulum, is the target. The profiles were similar to those of cyclic heptadepsipeptides of a distinct chemotype (including HUN-7293 and cotransin) that had previously been shown to inhibit cotranslational translocation at the mammalian Sec61 translocon. Unbiased, genome-wide mutagenesis followed by full-genome sequencing in both fungal and mammalian cells identified dominant mutations in Sec61p (yeast) or Sec61&agr;1 (mammals) that conferred resistance. Most, but not all, of these mutations affected inhibition by both chemotypes, despite an absence of structural similarity. Biochemical analysis confirmed inhibition of protein translocation into the endoplasmic reticulum of both co- and post-translationally translocated substrates by both chemotypes, demonstrating a mechanism independent of a translating ribosome. Most interestingly, both chemotypes were found to also inhibit SecYEG, the bacterial Sec61 translocon homolog. We suggest ‘decatransin’ as the name for this new decadepsipeptide translocation inhibitor.


Antimicrobial Agents and Chemotherapy | 2013

Identification and Evaluation of Novel Acetolactate Synthase Inhibitors as Antifungal Agents

Daryl L. Richie; Katherine Thompson; Christian Studer; Vivian Prindle; Thomas Aust; Ralph Riedl; David Estoppey; Jianshi Tao; Jessica A. Sexton; Thomas Zabawa; Joseph Drumm; Simona Cotesta; Jürg Eichenberger; Sven Schuierer; Nicole Hartmann; N. Rao Movva; John A. Tallarico; Neil S. Ryder; Dominic Hoepfner

ABSTRACT High-throughput phenotypic screening against the yeast Saccharomyces cerevisiae revealed a series of triazolopyrimidine-sulfonamide compounds with broad-spectrum antifungal activity, no significant cytotoxicity, and low protein binding. To elucidate the target of this series, we have applied a chemogenomic profiling approach using the S. cerevisiae deletion collection. All compounds of the series yielded highly similar profiles that suggested acetolactate synthase (Ilv2p, which catalyzes the first common step in branched-chain amino acid biosynthesis) as a possible target. The high correlation with profiles of known Ilv2p inhibitors like chlorimuron-ethyl provided further evidence for a similar mechanism of action. Genome-wide mutagenesis in S. cerevisiae identified 13 resistant clones with 3 different mutations in the catalytic subunit of acetolactate synthase that also conferred cross-resistance to established Ilv2p inhibitors. Mapping of the mutations into the published Ilv2p crystal structure outlined the chlorimuron-ethyl binding cavity, and it was possible to dock the triazolopyrimidine-sulfonamide compound into this pocket in silico. However, fungal growth inhibition could be bypassed through supplementation with exogenous branched-chain amino acids or by the addition of serum to the medium in all of the fungal organisms tested except for Aspergillus fumigatus. Thus, these data support the identification of the triazolopyrimidine-sulfonamide compounds as inhibitors of acetolactate synthase but suggest that targeting may be compromised due to the possibility of nutrient bypass in vivo.


Lecture Notes in Computer Science | 2003

A lower bound for randomized searching on m rays

Sven Schuierer

We consider the problem of on-line searching on m rays. A point robot is assumed to stand at the origin of m concurrent rays one of which contains a goal g that the point robot has to find. Neither the ray containing g nor the distance to g are known to the robot. The only way the robot can detect g is by reaching its location. We use the competitive ratio as a measure of the performance of a search strategy, that is, the worst case ratio of the total distance DR traveled by the robot to find g to the distance D from the origin to g.We present a new proof of a tight lower bound of the competitive ratio for randomized strategies to search on m rays. Our proof allows us to obtain a lower bound on the optimal competitive ratio for a fixed m even if the distance of the goal to the origin is bounded from above.Finally, we show that the optimal competitive ratio converges to 1 +2(eα- 1)/α2 m ∼ 1 + 2.1.544 m, for large m where α minimizes the function(ex- 1)/x2.


BMC Genomics | 2016

Identification of oncogenic driver mutations by genome-wide CRISPR-Cas9 dropout screening

Michael K. Kiessling; Sven Schuierer; Silke Stertz; Martin Beibel; Sebastian Bergling; Judith Knehr; Walter Carbone; Cheryl de Valliere; Joelle Tchinda; Tewis Bouwmeester; Klaus Seuwen; Gerhard Rogler; Guglielmo Roma

BackgroundGenome-wide CRISPR-Cas9 dropout screens can identify genes whose knockout affects cell viability. Recent CRISPR screens detected thousands of essential genes required for cellular survival and key cellular processes; however discovering novel lineage-specific genetic dependencies from the many hits still remains a challenge.ResultsTo assess whether CRISPR-Cas9 dropout screens can help identify cancer dependencies, we screened two human cancer cell lines carrying known and distinct oncogenic mutations using a genome-wide sgRNA library. We found that the gRNA targeting the driver mutation EGFR was one of the highest-ranking candidates in the EGFR-mutant HCC-827 lung adenocarcinoma cell line. Likewise, sgRNAs for NRAS and MAP2K1 (MEK1), a downstream kinase of mutant NRAS, were identified among the top hits in the NRAS-mutant neuroblastoma cell line CHP-212. Depletion of these genes targeted by the sgRNAs strongly correlated with the sensitivity to specific kinase inhibitors of the EGFR or RAS pathway in cell viability assays. In addition, we describe other dependencies such as TBK1 in HCC-827 cells and TRIB2 in CHP-212 cells which merit further investigation.ConclusionsWe show that genome-wide CRISPR dropout screens are suitable for the identification of oncogenic drivers and other essential genes.


Nucleic Acids Research | 2016

The exon quantification pipeline (EQP): a comprehensive approach to the quantification of gene, exon and junction expression from RNA-seq data

Sven Schuierer; Guglielmo Roma

The quantification of transcriptomic features is the basis of the analysis of RNA-seq data. We present an integrated alignment workflow and a simple counting-based approach to derive estimates for gene, exon and exon–exon junction expression. In contrast to previous counting-based approaches, EQP takes into account only reads whose alignment pattern agrees with the splicing pattern of the features of interest. This leads to improved gene expression estimates as well as to the generation of exon counts that allow disambiguating reads between overlapping exons. Unlike other methods that quantify skipped introns, EQP offers a novel way to compute junction counts based on the agreement of the read alignments with the exons on both sides of the junction, thus providing a uniformly derived set of counts. We evaluated the performance of EQP on both simulated and real Illumina RNA-seq data and compared it with other quantification tools. Our results suggest that EQP provides superior gene expression estimates and we illustrate the advantages of EQPs exon and junction counts. The provision of uniformly derived high-quality counts makes EQP an ideal quantification tool for differential expression and differential splicing studies. EQP is freely available for download at https://github.com/Novartis/EQP-cluster.


Journal of Biomolecular Screening | 2016

Advantages and Challenges of Phenotypic Screens The Identification of Two Novel Antifungal Geranylgeranyltransferase I Inhibitors

Verena Pries; Simona Cotesta; Ralph Riedl; Thomas Aust; Sven Schuierer; Jianshi Tao; Ireos Filipuzzi; Dominic Hoepfner

Phenotypic screens are effective starting points to identify compounds with desirable activities. To find novel antifungals, we conducted a phenotypic screen in Saccharomyces cerevisiae and identified two discrete scaffolds with good growth inhibitory characteristics. Lack of broad-spectrum activity against pathogenic fungi called for directed chemical compound optimization requiring knowledge of the molecular target. Chemogenomic profiling identified effects on geranylgeranyltransferase I (GGTase I), an essential enzyme that prenylates proteins involved in cell signaling, such as Cdc42p and Rho1p. Selection of resistant mutants against both compounds confirmed the target hypothesis and enabled mapping of the compound binding site to the substrate binding pocket. Differential resistance-conferring mutations and selective substrate competition demonstrate distinct binding modes for the two chemotypes. Exchange of the S. cerevisiae GGTase I subunits with those of Candida albicans resulted in an absence of growth inhibition for both compounds, thus confirming the identified target as well as the narrow antifungal spectrum of activity. This prenylation pathway is reported to be nonessential in pathogenic species and challenges the therapeutic value of these leads while demonstrating the importance of an integrated target identification platform following a phenotypic screen.

Collaboration


Dive into the Sven Schuierer's collaboration.

Researchain Logo
Decentralizing Knowledge