Network


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

Hotspot


Dive into the research topics where Martin Hintersteiner is active.

Publication


Featured researches published by Martin Hintersteiner.


Nature Biotechnology | 2005

In vivo detection of amyloid-β deposits by near-infrared imaging using an oxazine-derivative probe

Martin Hintersteiner; Albert Enz; Peter Frey; Anne-Lise Jaton; Willy Kinzy; Rainer Kneuer; Ulf Neumann; Markus Rudin; Matthias Staufenbiel; Markus Stoeckli; Karl-Heinz Wiederhold; Hans-Ulrich Gremlich

As Alzheimers disease pathogenesis is associated with the formation of insoluble aggregates of amyloid β-peptide, approaches allowing the direct, noninvasive visualization of plaque growth in vivo would be beneficial for biomedical research. Here we describe the synthesis and characterization of the near-infrared fluorescence oxazine dye AOI987, which readily penetrates the intact blood-brain barrier and binds to amyloid plaques. Using near-infrared fluorescence imaging, we demonstrated specific interaction of AOI987 with amyloid plaques in APP23 transgenic mice in vivo, as confirmed by postmortem analysis of brain slices. Quantitative analysis revealed increasing fluorescence signal intensity with increasing plaque load of the animals, and significant binding of AOI987 was observed for APP23 transgenic mice aged 9 months and older. Thus, AOI987 is an attractive probe to noninvasively monitor disease progression in animal models of Alzheimer disease and to evaluate effects of potential Alzheimer disease drugs on the plaque load.


The EMBO Journal | 2013

The rough endoplasmatic reticulum is a central nucleation site of siRNA-mediated RNA silencing

Lukas Stalder; Wolf Heusermann; Lena Sokol; Dominic Trojer; Joel Wirz; Justin Hean; Anja Fritzsche; Florian Aeschimann; Vera Pfanzagl; Pascal Basselet; Jan Weiler; Martin Hintersteiner; David V. Morrissey; Nicole Meisner-Kober

Despite progress in mechanistic understanding of the RNA interference (RNAi) pathways, the subcellular sites of RNA silencing remain under debate. Here we show that loading of lipid‐transfected siRNAs and endogenous microRNAs (miRNA) into RISC (RNA‐induced silencing complexes), encounter of the target mRNA, and Ago2‐mediated mRNA slicing in mammalian cells are nucleated at the rough endoplasmic reticulum (rER). Although the major RNAi pathway proteins are found in most subcellular compartments, the miRNA‐ and siRNA‐loaded Ago2 populations co‐sediment almost exclusively with the rER membranes, together with the RISC loading complex (RLC) factors Dicer, TAR RNA binding protein (TRBP) and protein activator of the interferon‐induced protein kinase (PACT). Fractionation and membrane co‐immune precipitations further confirm that siRNA‐loaded Ago2 physically associates with the cytosolic side of the rER membrane. Additionally, RLC‐associated double‐stranded siRNA, diagnostic of RISC loading, and RISC‐mediated mRNA cleavage products exclusively co‐sediment with rER. Finally, we identify TRBP and PACT as key factors anchoring RISC to ER membranes in an RNA‐independent manner. Together, our findings demonstrate that the outer rER membrane is a central nucleation site of siRNA‐mediated RNA silencing.


Chemistry & Biology | 2009

Single bead labeling method for combining confocal fluorescence on-bead screening and solution validation of tagged one-bead one-compound libraries.

Martin Hintersteiner; Thierry Kimmerlin; Frank Kalthoff; Markus Stoeckli; Geraldine Garavel; Jan-Marcus Seifert; Nicole-Claudia Meisner; Volker Uhl; Christof Buehler; Thomas Weidemann; Manfred Auer

Screening of one-bead one-compound libraries by incubating beads with fluorescently labeled target protein requires isolation and structure elucidation of a large number of primary hit beads. However, the potency of the identified ligands is only revealed after time consuming and expensive larger scale resynthesis and testing in solution. Often, many of the resynthesized compounds turn out to be weak target binders in solution due to large differences between surface and solution binding affinities. For an industry style high-throughput screening (HTS) process a high false positive rate is detrimental. We have therefore combined single bead and single molecule/single cell techniques into an integrated HTS process in which the picomole amount of substance contained on one isolated hit bead is sufficient for quality control, structure determination, and precise affinity determination to the target protein in solution.


ChemBioChem | 2008

Covalent fluorescence labeling of His-tagged proteins on the surface of living cells

Martin Hintersteiner; Thomas Weidemann; Thierry Kimmerlin; Nimet Filiz; Christof Buehler; Manfred Auer

Mechanistic studies in living cells require fluorescent labeling of the proteins of interest. The widespread application of GFP variants in combination with fluorescence microscopy has had an immense impact on our understanding of the dynamic processes in living cells. Due to the recent progress in detection technologies and microspectroscopy, the expansion of the available labeling strategies to more photostable, smaller, multicolor reagents has become an active field of research. The majority of these new fluorescent tags are small chemical entities that bind with various affinities to genetically fused peptide stretches in the protein of interest. Confocal images provide snapshots of dynamic molecular rearrangements. However, the observation of such processes by medium to highaffinity fluorescent ligands can be complicated because of a floating background inherent to the limited stability of the complex. Thus, covalent, site-selective protein-labeling techniques offer improvements in separating cellular from tag-specific biochemical events. Up to now much emphasis has been given to covalent-labeling strategies that rely on enzymatic couplings; only recently has a novel nonenzymatic protein-labeling concept been described. Herein we present an alternative strategy that combines the reversible binding of a medium affinity probe with a photoreactive moiety to generate a covalent linkage in the proximity of the tag. Substituted arylazides have been widely used in protein-interaction studies for photoaffinity labeling. Photoactivation is well suited for generating covalent bonds because of the simplicity of the activation procedure, good efficiencies, and short timescales of the reaction. The combination of a photocrosslinking moiety and an oligohistidine directing Ni-NTA group has previously been exploited for in vitro protein functionalization to build self-assembling protein arrays. Reasoning that this method could provide a simple means of covalently tagging proteins on living cells, we synthesized a trifunctional labeling tag that consists of a fluorophore, a Ni-NTA moiety, and a photoactivatable arylazide. Our labeling strategy comprises two steps: reversible binding at the His-tag of a target protein followed by irreversible photolinkage at the binding site (Figure 1). Irreversible crosslinking to a recombinant His-tagged GFP was demonstrated in vitro


Biophysical Journal | 2011

Single cell analysis of ligand binding and complex formation of interleukin-4 receptor subunits.

Thomas Weidemann; Remigiusz Worch; Kristina Kurgonaite; Martin Hintersteiner; Christian Bökel; Petra Schwille

Interleukin-4 (IL-4) is an important class I cytokine involved in adaptive immunity. IL-4 binds with high affinity to the single-pass transmembrane receptor IL-4Rα. Subsequently, IL-4Rα/IL-4 is believed to engage a second receptor chain, either IL-2Rγ or IL-13Rα1, to form type I or II receptor complexes, respectively. This ternary complex formation then triggers downstream signaling via intracellular Janus kinases bound to the cytoplasmic receptor tails. Here, we study the successive steps of complex formation at the single cell level with confocal fluorescence imaging and correlation spectroscopy. We characterize binding and signaling of fluorescently labeled IL-4 by flow cytometry of IL-4-dependent BaF3 cells. The affinity to ectopically expressed IL-4Rα was then measured by single-color fluorescence correlation spectroscopy in adherent HEK293T cells that express the components of the type II IL-4R but not type I. Finally, IL-4-induced complex formation was tested by dual-color fluorescence cross-correlation spectroscopy. The data provide evidence for codiffusion of IL-4-A647 bound IL-4Rα and the type II subunit IL-13Rα1 fused to enhanced green fluorescent protein, whereas type I complexes containing IL-2Rγ and JAK3 were not detected at the cell surface. This behavior may reflect hitherto undefined differences in the mode of receptor activation between type I (lymphoid) and type II (epithelial) receptor expressing cells.


Biophysical Journal | 2014

Dynamics and Interaction of Interleukin-4 Receptor Subunits in Living Cells

Hetvi Gandhi; Remigiusz Worch; Kristina Kurgonaite; Martin Hintersteiner; Petra Schwille; Christian Bökel; Thomas Weidemann

It has long been established that dimerization of Interleukin-4 receptor (IL-4R) subunits is a pivotal step for JAK/STAT signal transduction. However, ligand-induced complex formation at the surface of living cells has been challenging to observe. Here we report an experimental assay employing trisNTA dyes for orthogonal, external labeling of eGFP-tagged receptor constructs that allows the quantification of receptor heterodimerization by dual-color fluorescence cross-correlation spectroscopy. Fluorescence cross-correlation spectroscopy analysis at the plasma membrane shows that IL-4R subunit dimerization is indeed a strictly ligand-induced process. Under conditions of saturating cytokine occupancy, we determined intramembrane dissociation constants (K(d,2D)) of 180 and 480 receptors per μm(2) for the type-2 complexes IL-4:IL-4Rα/IL-13Rα1 and IL-13:IL-13Rα1/IL-4Rα, respectively. For the lower affinity type-1 complex IL-4:IL-4Rα/IL-2Rγ, we estimated a K(d,2D) of ∼1000 receptors per μm(2). The receptor densities required for effective dimerization thus exceed the typical, average expression levels by several orders of magnitude. In addition, we find that all three receptor subunits accumulate rapidly within a subpopulation of early sorting and recycling endosomes stably anchored just beneath the plasma membrane (cortical endosomes, CEs). The receptors, as well as labeled IL-4 and trisNTA ligands are specifically trafficked into CEs by a constitutive internalization mechanism. This may compensate for the inherent weak affinities that govern ligand-induced receptor dimerization at the plasma membrane. Consistently, activated receptors are also concentrated at the CEs. Our observations thus suggest that receptor trafficking may play an important role for the regulation of IL-4R-mediated JAK/STAT signaling.


ChemBioChem | 2009

A Highly Potent and Cellularly Active β-Peptidic Inhibitor of the p53/hDM2 Interaction

Martin Hintersteiner; Thierry Kimmerlin; Geraldine Garavel; Thorsten Schindler; Roman Bauer; Nicole-Claudia Meisner; Jan-Marcus Seifert; Volker Uhl; Manfred Auer

New and improved: The incorporation of a 6‐chlorotryptophan (6‐Cl‐Trp) into a β‐peptide (M)‐314 helix leads to a high‐affinity hDM2 inhibitor, as demonstrated by fluorescence fluctuation analysis at single molecule resolution. When conjugated to penetratin, the newly derived hDM2 binder specifically inhibits tumour cell growth in vitro.


Angewandte Chemie | 2014

Identification and X-ray co-crystal structure of a small-molecule activator of LFA-1-ICAM-1 binding.

Martin Hintersteiner; Joerg Kallen; Mario Schmied; Christine Graf; Thomas Jung; Gemma Mudd; Steven Shave; Hubert Gstach; Manfred Auer

Stabilization of protein–protein interactions by small molecules is a concept with few examples reported to date. Herein we describe the identification and X-ray co-crystal structure determination of IBE-667, an ICAM-1 binding enhancer for LFA-1. IBE-667 was designed based on the SAR information obtained from an on-bead screen of tagged one-bead one-compound combinatorial libraries by confocal nanoscanning and bead picking (CONA). Cellular assays demonstrate the activity of IBE-667 in promoting the binding of LFA-1 on activated immune cells to ICAM-1.


ACS Combinatorial Science | 2010

Analysis of protein-small molecule interactions by microscale equilibrium dialysis and its application as a secondary confirmation method for on-bead screening.

Thomas Weidemann; Jan-Marcus Seifert; Martin Hintersteiner; Manfred Auer

On-bead screening of one-bead one compound (OBOC) libraries is an ultra fast surface based primary high-throughput screening (HTS) method. Typically the binding of a tagged target protein to bead immobilized compounds or its altered enzymatic activity are detected. For an efficient and reliable ligand discovery process secondary assays to confirm on-bead compound activity in homogeneous solution are key to exclude artifacts and weak binders. Ideally they should allow to flag hit compounds with undesirable biophysical properties such as aggregation, unspecific binding, or insufficient solubility and the like. Here we demonstrate that miniaturized and parallelized equilibrium dialysis is an excellent and generic secondary confirmation method for hit compounds identified by on-bead screening. We further show that microscale dialysis can be reliably performed prior to decoding and resynthesis even with hit-compounds cleaved from the single beads. Down-scaling of the method takes advantage of the fluorescent tag, AIDA, which is integrated as permanent tracer in our library design. Our results suggest that microscale equilibrium dialysis followed by high performance liquid chromatography (HPLC) analysis is a generic, cheap, and meaningful confirmation method for identifying the most promising candidates within a series hit compounds derived from fluorescently tagged one-bead one-compound libraries.


Journal of Chemical Biology | 2012

Towards mimicking short linear peptide motifs: identification of new mixed α,β-peptidomimetic ligands for SLAM-Associated Protein (SAP) by confocal on-bead screening.

Martin Hintersteiner; Andrew J. S. Knox; Gemma Mudd; Manfred Auer

An array of chemical modifications have recently emerged, designed to improve the stability of natural peptides that inherently suffer from short in vivo half-lives, thereby preventing their use as therapeutics. The resultant peptidomimetics resemble native peptides; however, they contain synthetic elements (e.g. non-coded amino acids) which confer improved biophysical properties. An elegant approach towards the identification of peptidomimetics is through screening of large combinatorial chemical libraries incorporating both coded and non-coded amino acids (e.g. β amino acids). We apply here our recently developed Integrated Chemical Biophysics (ICB) platform, which combines microscale one-bead one-compound screening with fluorescence tagging of retrieved hit beads and subsequent affinity determination of hit compounds in homogenous solution, to the task of identifying novel mixed α, β peptidomimetic binders for the adaptor protein SLAM-associated protein (SAP), which acts as an intracellular adapter that transduces T and NK cell activation. An enhancement to the ICB process is introduced which enables ranking hit compounds from single-point measurements even if the library compound is <95% pure and without HPLC purification of single-bead-derived substance. Finally, a novel computational protocol enabling binding mode and SAR rationalisation of hit compounds is also described which we now utilise to inform future library design. Application of the full ICB process has allowed identification of a highly interesting motif, Ac-β3-Pro-α-pTyr, as a mimic for the −1 and −2 positions of the natural binding motif and provides a promising starting point for further optimization towards higher-affinity SAP inhibitors with enhanced metabolic stability.

Collaboration


Dive into the Martin Hintersteiner's collaboration.

Top Co-Authors

Avatar

Manfred Auer

Lawrence Berkeley National Laboratory

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gemma Mudd

University of Edinburgh

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge