Network


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

Hotspot


Dive into the research topics where Friedrich Rippmann is active.

Publication


Featured researches published by Friedrich Rippmann.


Journal of Molecular Graphics & Modelling | 1997

LIGSITE: automatic and efficient detection of potential small molecule-binding sites in proteins

Manfred Hendlich; Friedrich Rippmann; Gerhard Barnickel

LIGSITE is a new program for the automatic and time-efficient detection of pockets on the surface of proteins that may act as binding sites for small molecule ligands. Pockets are identified with a series of simple operations on a cubic grid. Using a set of receptor-ligand complexes we show that LIGSITE is able to identify the binding sites of small molecule ligands with high precision. The main advantage of LIGSITE is its speed. Typical search times are in the range of 5 to 20 s for medium-sized proteins. LIGSITE is therefore well suited for identification of pockets in large sets of proteins (e.g., protein families) for comparative studies. For graphical display LIGSITE produces VRML representations of the protein-ligand complex and the binding site for display with a VRML viewer such as WebSpace from SGI.


Journal of Molecular Recognition | 2009

Computational approaches to identifying and characterizing protein binding sites for ligand design

Stefan Henrich; Outi M. H. Salo-Ahen; Bingding Huang; Friedrich Rippmann; Gabriele Cruciani; Rebecca C. Wade

Given the three‐dimensional structure of a protein, how can one find the sites where other molecules might bind to it? Do these sites have the properties necessary for high affinity binding? Is this protein a suitable target for drug design? Here, we discuss recent developments in computational methods to address these and related questions. Geometric methods to identify pockets on protein surfaces have been developed over many years but, with new algorithms, their performance is still improving. Simulation methods show promise in accounting for protein conformational variability to identify transient pockets but lack the ease of use of many of the (rigid) shape‐based tools. Sequence and structure comparison approaches are benefiting from the constantly increasing size of sequence and structure databases. Energetic methods can aid identification and characterization of binding pockets, and have undergone recent improvements in the treatment of solvation and hydrophobicity. The “druggability” of a binding site is still difficult to predict with an automated procedure. The methodologies available for this purpose range from simple shape and hydrophobicity scores to computationally demanding free energy simulations. Copyright


Bioinformatics | 2012

DoGSiteScorer: a web server for automatic binding site prediction, analysis and druggability assessment

Andrea Volkamer; Daniel Kuhn; Friedrich Rippmann; Matthias Rarey

MOTIVATION Many drug discovery projects fail because the underlying target is finally found to be undruggable. Progress in structure elucidation of proteins now opens up a route to automatic structure-based target assessment. DoGSiteScorer is a newly developed automatic tool combining pocket prediction, characterization and druggability estimation and is now available through a web server. AVAILABILITY The DoGSiteScorer web server is freely available for academic use at http://dogsite.zbh.uni-hamburg.de CONTACT [email protected].


Journal of Chemical Information and Modeling | 2011

A normal mode-based geometric simulation approach for exploring biologically relevant conformational transitions in proteins.

Aqeel Ahmed; Friedrich Rippmann; Gerhard Barnickel; Holger Gohlke

A three-step approach for multiscale modeling of protein conformational changes is presented that incorporates information about preferred directions of protein motions into a geometric simulation algorithm. The first two steps are based on a rigid cluster normal-mode analysis (RCNMA). Low-frequency normal modes are used in the third step (NMSim) to extend the recently introduced idea of constrained geometric simulations of diffusive motions in proteins by biasing backbone motions of the protein, whereas side-chain motions are biased toward favorable rotamer states. The generated structures are iteratively corrected regarding steric clashes and stereochemical constraint violations. The approach allows performing three simulation types: unbiased exploration of conformational space; pathway generation by a targeted simulation; and radius of gyration-guided simulation. When applied to a data set of proteins with experimentally observed conformational changes, conformational variabilities are reproduced very well for 4 out of 5 proteins that show domain motions, with correlation coefficients r > 0.70 and as high as r = 0.92 in the case of adenylate kinase. In 7 out of 8 cases, NMSim simulations starting from unbound structures are able to sample conformations that are similar (root-mean-square deviation = 1.0-3.1 Å) to ligand bound conformations. An NMSim generated pathway of conformational change of adenylate kinase correctly describes the sequence of domain closing. The NMSim approach is a computationally efficient alternative to molecular dynamics simulations for conformational sampling of proteins. The generated conformations and pathways of conformational transitions can serve as input to docking approaches or as starting points for more sophisticated sampling techniques.


Journal of Chemical Information and Modeling | 2013

TRAPP: A Tool for Analysis of Transient Binding Pockets in Proteins

Daria B. Kokh; Stefan Richter; Stefan Henrich; Paul Czodrowski; Friedrich Rippmann; Rebecca C. Wade

We present TRAPP (TRAnsient Pockets in Proteins), a new automated software platform for tracking, analysis, and visualization of binding pocket variations along a protein motion trajectory or within an ensemble of protein structures that may encompass conformational changes ranging from local side chain fluctuations to global backbone motions. TRAPP performs accurate grid-based calculations of the shape and physicochemical characteristics of a binding pocket for each structure and detects the conserved and transient regions of the pocket in an ensemble of protein conformations. It also provides tools for tracing the opening of a particular subpocket and residues that contribute to the binding site. TRAPP thus enables an assessment of the druggability of a disease-related target protein taking its flexibility into account.


Journal of Chemical Information and Computer Sciences | 1997

BALI : AUTOMATIC ASSIGNMENT OF BOND AND ATOM TYPES FOR PROTEIN LIGANDS IN THE BROOKHAVEN PROTEIN DATABANK

Manfred Hendlich; Friedrich Rippmann; Gerhard Barnickel

A method for assigning hybridization states and bond orders to protein ligands from the Brookhaven Protein Databank1 is presented. The assignment procedure is based on the recognition of simple chemical groups and on an analysis of bond length and bond angle data. Special care was taken on the proper handling of aromatic heterocyclic ring systems which are frequently found in small molecule ligands. Hybridization states and bond orders are assigned with a success rate significantly higher than what has been reported for previously published algorithms. The main application of BALI is to identify protein ligands in the Brookhaven Protein Databank and to add information about bond and atom types with a minimum amount of manual intervention.


BMC Bioinformatics | 2017

KinMap: a web-based tool for interactive navigation through human kinome data

Sameh Eid; Samo Turk; Andrea Volkamer; Friedrich Rippmann; Simone Fulle

BackgroundAnnotations of the phylogenetic tree of the human kinome is an intuitive way to visualize compound profiling data, structural features of kinases or functional relationships within this important class of proteins. The increasing volume and complexity of kinase-related data underlines the need for a tool that enables complex queries pertaining to kinase disease involvement and potential therapeutic uses of kinase inhibitors.ResultsHere, we present KinMap, a user-friendly online tool that facilitates the interactive navigation through kinase knowledge by linking biochemical, structural, and disease association data to the human kinome tree. To this end, preprocessed data from freely-available sources, such as ChEMBL, the Protein Data Bank, and the Center for Therapeutic Target Validation platform are integrated into KinMap and can easily be complemented by proprietary data. The value of KinMap will be exemplarily demonstrated for uncovering new therapeutic indications of known kinase inhibitors and for prioritizing kinases for drug development efforts.ConclusionKinMap represents a new generation of kinome tree viewers which facilitates interactive exploration of the human kinome. KinMap enables generation of high-quality annotated images of the human kinome tree as well as exchange of kinome-related data in scientific communications. Furthermore, KinMap supports multiple input and output formats and recognizes alternative kinase names and links them to a unified naming scheme, which makes it a useful tool across different disciplines and applications. A web-service of KinMap is freely available at http://www.kinhub.org/kinmap/.


Journal of Chemical Information and Modeling | 2015

Pocketome of Human Kinases: Prioritizing the ATP Binding Sites of (Yet) Untapped Protein Kinases for Drug Discovery

Andrea Volkamer; Sameh Eid; Samo Turk; Sabrina Jaeger; Friedrich Rippmann; Simone Fulle

Protein kinases are involved in a variety of diseases including cancer, inflammation, and autoimmune disorders. Although the development of new kinase inhibitors is a major focus in pharmaceutical research, a large number of kinases remained so far unexplored in drug discovery projects. The selection and assessment of targets is an essential but challenging area. Today, a few thousands of experimentally determined kinase structures are available, covering about half of the human kinome. This large structural source allows guiding the target selection via structure-based druggability prediction approaches such as DoGSiteScorer. Here, a thorough analysis of the ATP pockets of the entire human kinome in the DFG-in state is presented in order to prioritize novel kinase structures for drug discovery projects. For this, all human kinase X-ray structures available in the PDB were collected, and homology models were generated for the missing part of the kinome. DoGSiteScorer was used to calculate geometrical and physicochemical properties of the ATP pockets and to predict the potential of each kinase to be druggable. The results indicate that about 75% of the kinome are in principle druggable. Top ranking structures comprise kinases that are primary targets of known approved drugs but additionally point to so far less explored kinases. The presented analysis provides new insights into the druggability of ATP binding pockets of the entire kinome. We anticipate this comprehensive druggability assessment of protein kinases to be helpful for the community to prioritize so far untapped kinases for drug discovery efforts.


Bioorganic & Medicinal Chemistry Letters | 1996

New antithrombotic RGD-mimetics with high bioavailability

Joachim Prof Dr Gante; Horst Juraszyk; Peter Raddatz; Hanns Wurziger; Sabine Bernotat-Danielowski; Guido Melzer; Friedrich Rippmann

Abstract A new class of antithrombotic RGD-mimetics with a novel axazolidinonemethyl scaffold was synthesized. High oral activity and bioavailability was found in this series of compounds.


Journal of Medicinal Chemistry | 2017

Profiling Prediction of Kinase Inhibitors: Toward the Virtual Assay

Benjamin Merget; Samo Turk; Sameh Eid; Friedrich Rippmann; Simone Fulle

Kinome-wide screening would have the advantage of providing structure-activity relationships against hundreds of targets simultaneously. Here, we report the generation of ligand-based activity prediction models for over 280 kinases by employing Machine Learning methods on an extensive data set of proprietary bioactivity data combined with open data. High quality (AUC > 0.7) was achieved for ∼200 kinases by (1) combining open with proprietary data, (2) choosing Random Forest over alternative tested Machine Learning methods, and (3) balancing the training data sets. Tests on left-out and external data indicate a high value for virtual screening projects. Importantly, the derived models are evenly distributed across the kinome tree, allowing reliable profiling prediction for all kinase branches. The prediction quality was further improved by employing experimental bioactivity fingerprints of a small kinase subset. Overall, the generated models can support various hit identification tasks, including virtual screening, compound repurposing, and the detection of potential off-targets.

Collaboration


Dive into the Friedrich Rippmann's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Simone Fulle

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Samo Turk

University of Ljubljana

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge