Network


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

Hotspot


Dive into the research topics where Markus Tusche is active.

Publication


Featured researches published by Markus Tusche.


Science | 2017

Fibril structure of amyloid-beta (1-42) by cryo-electron microscopy.

Lothar Gremer; Daniel Schölzel; Carla Schenk; Elke Reinartz; Jörg Labahn; Raimond B. G. Ravelli; Markus Tusche; Wolfgang Hoyer; Henrike Heise; Dieter Willbold; Gunnar F. Schröder

Elucidating pathological fibril structure Amyloid-β (Aβ) is a key pathological contributor to Alzheimers disease. Gremer et al. used cryoelectron microscopy data to build a high-quality de novo atomic model of Aβ fibrils (see the Perspective by Pospich and Raunser). The complete structure reveals all 42 amino acids (including the entire N terminus) and provides a structural basis for understanding the effect of several disease-causing and disease-preventing mutations. The fibril consists of two intertwined protofilaments with an unexpected dimer interface that is different from those proposed previously. The structure has implications for the mechanism of fibril growth and will be an important stepping stone to rational drug design. Science, this issue p. 116; see also p. 45 Cryo–electron microscopy structure of an amyloid-β(1–42) fibril reveals a protofilament interface and the entire N-terminal region. Amyloids are implicated in neurodegenerative diseases. Fibrillar aggregates of the amyloid-β protein (Aβ) are the main component of the senile plaques found in brains of Alzheimer’s disease patients. We present the structure of an Aβ(1–42) fibril composed of two intertwined protofilaments determined by cryo–electron microscopy (cryo-EM) to 4.0-angstrom resolution, complemented by solid-state nuclear magnetic resonance experiments. The backbone of all 42 residues and nearly all side chains are well resolved in the EM density map, including the entire N terminus, which is part of the cross-β structure resulting in an overall “LS”-shaped topology of individual subunits. The dimer interface protects the hydrophobic C termini from the solvent. The characteristic staggering of the nonplanar subunits results in markedly different fibril ends, termed “groove” and “ridge,” leading to different binding pathways on both fibril ends, which has implications for fibril growth.


Scientific Reports | 2015

QIAD assay for quantitating a compound's efficacy in elimination of toxic Aβ oligomers.

Oleksandr Brener; Tina Dunkelmann; Lothar Gremer; Thomas van Groen; Ewa A. Mirecka; Inga Kadish; Antje Willuweit; Janine Kutzsche; Dagmar Jürgens; Stephan Rudolph; Markus Tusche; Patrick Bongen; Jörg Pietruszka; Filipp Oesterhelt; Karl-Josef Langen; Hans-Ulrich Demuth; Arnold Janssen; Wolfgang Hoyer; Susanne Aileen Funke; Luitgard Nagel-Steger; Dieter Willbold

Strong evidence exists for a central role of amyloid β-protein (Aβ) oligomers in the pathogenesis of Alzheimer’s disease. We have developed a fast, reliable and robust in vitro assay, termed QIAD, to quantify the effect of any compound on the Aβ aggregate size distribution. Applying QIAD, we studied the effect of homotaurine, scyllo-inositol, EGCG, the benzofuran derivative KMS88009, ZAβ3W, the D-enantiomeric peptide D3 and its tandem version D3D3 on Aβ aggregation. The predictive power of the assay for in vivo efficacy is demonstrated by comparing the oligomer elimination efficiency of D3 and D3D3 with their treatment effects in animal models of Alzheimer´s disease.


Scientific Reports | 2017

The Aβ oligomer eliminating D -enantiomeric peptide RD2 improves cognition without changing plaque pathology

Thomas van Groen; Sarah Schemmert; Oleksandr Brener; Lothar Gremer; Tamar Ziehm; Markus Tusche; Luitgard Nagel-Steger; Inga Kadish; Elena Schartmann; Anne Elfgen; Dagmar Jürgens; Antje Willuweit; Janine Kutzsche; Dieter Willbold

While amyloid-β protein (Aβ) aggregation into insoluble plaques is one of the pathological hallmarks of Alzheimer’s disease (AD), soluble oligomeric Aβ has been hypothesized to be responsible for synapse damage, neurodegeneration, learning, and memory deficits in AD. Here, we investigate the in vitro and in vivo efficacy of the d-enantiomeric peptide RD2, a rationally designed derivative of the previously described lead compound D3, which has been developed to efficiently eliminate toxic Aβ42 oligomers as a promising treatment strategy for AD. Besides the detailed in vitro characterization of RD2, we also report the results of a treatment study of APP/PS1 mice with RD2. After 28 days of treatment we observed enhancement of cognition and learning behaviour. Analysis on brain plaque load did not reveal significant changes, but a significant reduction of insoluble Aβ42. Our findings demonstrate that RD2 was significantly more efficient in Aβ oligomer elimination in vitro compared to D3. Enhanced cognition without reduction of plaque pathology in parallel suggests that synaptic malfunction due to Aβ oligomers rather than plaque pathology is decisive for disease development and progression. Thus, Aβ oligomer elimination by RD2 treatment may be also beneficial for AD patients.


ACS Chemical Neuroscience | 2016

Increase of Positive Net Charge and Conformational Rigidity Enhances the Efficacy of d-Enantiomeric Peptides Designed to Eliminate Cytotoxic Aβ Species

Tamar Ziehm; Oleksandr Brener; Thomas van Groen; Inga Kadish; Daniel Frenzel; Markus Tusche; Janine Kutzsche; Kerstin Reiß; Lothar Gremer; Luitgard Nagel-Steger; Dieter Willbold

Alzheimers disease (AD) is a neurodegenerative disorder and the most common type of dementia. Until now, there is no curative therapy available. Previously, we selected the amyloid-beta (Aβ) targeting peptide D3 consisting of 12 d-enantiomeric amino acid residues by mirror image phage display as a potential drug candidate for the treatment of AD. In the current approach, we investigated the optimization potential of linear D3 with free C-terminus (D3COOH) by chemical modifications. First, the impact of the net charge was investigated and second, cyclization was introduced which is a well-known tool for the optimization of peptides for enhanced target affinity. Following this strategy, three D3 derivatives in addition to D3COOH were designed: C-terminally amidated linear D3 (D3CONH2), cyclic D3 (cD3), and cyclic D3 with an additional arginine residue (cD3r) to maintain the net charge of linear D3CONH2. These four compounds were compared to each other according to their binding affinities to Aβ(1-42), their efficacy to eliminate cytotoxic oligomers, and consequently their potency to neutralize Aβ(1-42) oligomer induced neurotoxicity. D3CONH2 and cD3r versions with equally increased net charge showed superior properties over D3COOH and cD3, respectively. The cyclic versions showed superior properties compared to their linear version with equal net charge, suggesting cD3r to be the most efficient compound among these four. Indeed, treatment of the transgenic AD mouse model Tg-SwDI with cD3r significantly enhanced spatial memory and cognition of these animals as revealed by water maze performance. Therefore, charge increase and cyclization imply suitable modification steps for an optimization approach of the Aβ targeting compound D3.


Molecules | 2017

Large-Scale Oral Treatment Study with the Four Most Promising D3-Derivatives for the Treatment of Alzheimer’s Disease

Janine Kutzsche; Sarah Schemmert; Markus Tusche; Jörg Neddens; Roland Rabl; Dagmar Jürgens; Oleksandr Brener; Antje Willuweit; Birgit Hutter-Paier; Dieter Willbold

Alzheimer’s disease (AD) is a progressive neurodegenerative disorder that is associated with the aggregation of the amyloid β protein (Aβ). Aβ oligomers are currently thought to be the major neurotoxic agent responsible for disease development and progression. Thus, their elimination is highly desirable for therapy development. Our therapeutic approach aims at specific and direct elimination of toxic Aβ oligomers by stabilizing Aβ monomers in an aggregation-incompetent conformation. We have proven that our lead compound “D3”, an all d-enantiomeric-peptide, specifically eliminates Aβ oligomers in vitro. In vivo, D3 enhances cognition and reduces plaque load in several transgenic AD mouse models. Here, we performed a large-scale oral proof of concept efficacy study, in which we directly compared four of the most promising D3-derivatives in transgenic mice expressing human amyloid precursor protein with Swedish and London mutations (APPSL), transgenic mice, to identify the most effective compound. RD2 and D3D3, both derived from D3 by rational design, were discovered to be the most effective derivatives in improving cognition in the Morris water maze. The performance of RD2- and D3D3-treated mice within the Morris water maze was significantly better than placebo-treated mice and, importantly, nearly as good as those of non-transgenic littermates, suggesting a complete reversal of the cognitive deficit of APPSL mice.


ACS Chemical Neuroscience | 2017

Optimization of d-Peptides for Aβ Monomer Binding Specificity Enhances Their Potential to Eliminate Toxic Aβ Oligomers

Antonia Nicole Klein; Tamar Ziehm; Thomas van Groen; Inga Kadish; Anne Elfgen; Markus Tusche; Maren Thomaier; Kerstin Reiss; Oleksandr Brener; Lothar Gremer; Janine Kutzsche; Dieter Willbold

Amyloid-beta (Aβ) oligomers are thought to be causative for the development and progression of Alzheimers disease (AD). Starting from the Aβ oligomer eliminating d-enantiomeric peptide D3, we developed and applied a two-step procedure based on peptide microarrays to identify D3 derivatives with increased binding affinity and specificity for monomeric Aβ(1-42) to further enhance the Aβ oligomer elimination efficacy. Out of more than 1000 D3 derivatives, we selected seven novel d-peptides, named ANK1 to ANK7, and characterized them in more detail in vitro. All ANK peptides bound to monomeric Aβ(1-42), eliminated Aβ(1-42) oligomers, inhibited Aβ(1-42) fibril formation, and reduced Aβ(1-42)-induced cytotoxicity more efficiently than D3. Additionally, ANK6 completely inhibited the prion-like propagation of preformed Aβ(1-42) seeds and showed a nonsignificant tendency for improving memory performance of tg-APPSwDI mice after i.p. application for 4 weeks. This supports the hypothesis that stabilization of Aβ monomers and thereby induced elimination of Aβ oligomers is a suitable therapeutic strategy.


PLOS ONE | 2016

Optimization of the All-D Peptide D3 for Aβ Oligomer Elimination.

Antonia Nicole Klein; Tamar Ziehm; Markus Tusche; Johan Buitenhuis; Dirk Bartnik; Annett Boeddrich; Thomas Wiglenda; Erich E. Wanker; Susanne Aileen Funke; Oleksandr Brener; Lothar Gremer; Janine Kutzsche; Dieter Willbold

The aggregation of amyloid-β (Aβ) is postulated to be the crucial event in Alzheimer’s disease (AD). In particular, small neurotoxic Aβ oligomers are considered to be responsible for the development and progression of AD. Therefore, elimination of thesis oligomers represents a potential causal therapy of AD. Starting from the well-characterized d-enantiomeric peptide D3, we identified D3 derivatives that bind monomeric Aβ. The underlying hypothesis is that ligands bind monomeric Aβ and stabilize these species within the various equilibria with Aβ assemblies, leading ultimately to the elimination of Aβ oligomers. One of the hereby identified d-peptides, DB3, and a head-to-tail tandem of DB3, DB3DB3, were studied in detail. Both peptides were found to: (i) inhibit the formation of Thioflavin T-positive fibrils; (ii) bind to Aβ monomers with micromolar affinities; (iii) eliminate Aβ oligomers; (iv) reduce Aβ-induced cytotoxicity; and (v) disassemble preformed Aβ aggregates. The beneficial effects of DB3 were improved by DB3DB3, which showed highly enhanced efficacy. Our approach yielded Aβ monomer-stabilizing ligands that can be investigated as a suitable therapeutic strategy against AD.


European Journal of Pharmaceutical Sciences | 2018

Comparison of blood-brain barrier penetration efficiencies between linear and cyclic all-d-enantiomeric peptides developed for the treatment of Alzheimer's disease

Elena Schartmann; Sarah Schemmert; Tamar Ziehm; Leonie H. E. Leithold; Nan Jiang; Markus Tusche; N. Joni Shah; Karl-Josef Langen; Janine Kutzsche; Dieter Willbold; Antje Willuweit

Abstract Alzheimers disease (AD), until now, is an incurable progressive neurodegenerative disease. To target toxic amyloid &bgr; oligomers in AD patients’ brains and to convert them into non‐toxic aggregation‐incompetent species, we designed peptides consisting solely of d‐enantiomeric amino acid residues. The original lead compound was named D3 and several D3 derivatives were designed to enhance beneficial properties. Here, we compare four d‐peptides concerning their efficiencies to pass the blood‐brain barrier (BBB). We demonstrate that the d‐peptides’ concentrations in murine brain directly correlate with concentrations in cerebrospinal fluid. The cyclic d‐enantiomeric peptide cRD2D3 is characterized by the highest efficiency to pass the BBB. For in total three cyclic peptides we show that administration of cyclic peptides resulted in up to tenfold higher peak concentrations in brain as compared to their linear equivalents which have partially been characterized before (Jiang et al., 2015; Leithold et al., 2016a). These results suggest that cyclic peptides pass the murine BBB more efficiently than their linear equivalents. cRD2D3s proteolytic stability, oral bioavailability, long duration of action and its favorable brain/plasma ratio reveal that it may become a suitable drug for long‐term AD‐treatment from a pharmacokinetic point of view. Graphical abstract Figure. No caption available.


Scientific Reports | 2017

Direct binding to GABARAP family members is essential for HIV-1 Nef plasma membrane localization

Alexandra Boeske; Melanie Schwarten; Peixiang Ma; Markus Tusche; Jessica Mötter; Christina Möller; Philipp Neudecker; Silke Hoffmann; Dieter Willbold

HIV-1 Nef is an important pathogenic factor for HIV/AIDS pathogenesis. Studies have shown that the association of Nef with the inner leaflet of the plasma membrane and with endocytic and perinuclear vesicles is essential for most activities of Nef. Using purified recombinant proteins in pull-down assays and by co-immunoprecipitation assays we demonstrate that Nef binds directly and specifically to all GABARAP family members, but not to LC3 family members. Based on nuclear magnetic resonance (NMR) experiments we showed that Nef binds to GABARAP via two surface exposed hydrophobic pockets. S53 and F62 of GABARAP were identified as key residues for the interaction with Nef. During live-cell fluorescence microscopy an accumulation of Nef and all GABARAP family members in vesicular structures throughout the cytoplasm and at the plasma membrane was observed. This plasma membrane accumulation was significantly reduced after knocking down GABARAP, GABARAPL1 and GABARAPL2 with respective siRNAs. We identified GABARAPs as the first known direct interaction partners of Nef that are essential for its plasma membrane localization.


Neuropeptides | 2017

Aβ oligomer eliminating compounds interfere successfully with pEAβ(3-42) induced motor neurodegenerative phenotype in transgenic mice

Tina Dunkelmann; Sarah Schemmert; Antje Willuweit; Karl-Josef Langen; Nadim Jon Shah; Markus Tusche; Hans-Ulrich Demuth; Dagmar Jürgens; Tamar Ziehm; Christina Dammers; Kerstin Teichmann; Janine Kutzsche; Dieter Willbold; Daniel Frenzel

Currently, there are no causative or disease modifying treatments available for Alzheimers disease (AD). Previously, it has been shown that D3, a small, fully d-enantiomeric peptide is able to eliminate low molecular weight Aβ oligomers in vitro, enhance cognition and reduce plaque load in AD transgenic mice. To further characterise the therapeutic potential of D3 towards N-terminally truncated and pyroglutamated Aβ (pEAβ(3-42)) we tested D3 and its head-to-tail tandem derivative D3D3 both in vitro and in vivo in the new mouse model TBA2.1. These mice produce human pEAβ(3-42) leading to a strong, early onset motor neurodegenerative phenotype. In the present study, we were able to demonstrate 1) strong binding affinity of both D3 and D3D3 to pEAβ(3-42) in comparison to Aβ(1-42) and 2) increased affinity of the tandem derivative D3D3 in comparison to D3. Subsequently we tested the therapeutic potentials of both peptides in the TBA2.1 animal model. Truly therapeutic, non-preventive treatment with D3 and D3D3 clearly slowed the progression of the neurodegenerative TBA2.1 phenotype, indicating the strong therapeutic potential of both peptides against pEAβ(3-42) induced neurodegeneration.

Collaboration


Dive into the Markus Tusche's collaboration.

Top Co-Authors

Avatar

Dieter Willbold

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar

Janine Kutzsche

Forschungszentrum Jülich

View shared research outputs
Top Co-Authors

Avatar

Lothar Gremer

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tamar Ziehm

Forschungszentrum Jülich

View shared research outputs
Top Co-Authors

Avatar

Antje Willuweit

Forschungszentrum Jülich

View shared research outputs
Top Co-Authors

Avatar

Sarah Schemmert

Forschungszentrum Jülich

View shared research outputs
Top Co-Authors

Avatar

Anne Elfgen

Forschungszentrum Jülich

View shared research outputs
Top Co-Authors

Avatar

Dagmar Jürgens

Forschungszentrum Jülich

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge