Network


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

Hotspot


Dive into the research topics where Janine Kutzsche is active.

Publication


Featured researches published by Janine Kutzsche.


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.


PLOS ONE | 2015

Preclinical Pharmacokinetic Studies of the Tritium Labelled D-Enantiomeric Peptide D3 Developed for the Treatment of Alzheimer´s Disease

Nan Jiang; Leonie H. E. Leithold; Julia Post; Tamar Ziehm; Jörg Mauler; Lothar Gremer; Markus Cremer; Elena Schartmann; N. Jon Shah; Janine Kutzsche; Karl-Josef Langen; Jörg Breitkreutz; Dieter Willbold; Antje Willuweit

Targeting toxic amyloid beta (Aβ) oligomers is currently a very attractive drug development strategy for treatment of Alzheimer´s disease. Using mirror-image phage display against Aβ1-42, we have previously identified the fully D-enantiomeric peptide D3, which is able to eliminate Aβ oligomers and has proven therapeutic potential in transgenic Alzheimer´s disease animal models. However, there is little information on the pharmacokinetic behaviour of D-enantiomeric peptides in general. Therefore, we conducted experiments with the tritium labelled D-peptide D3 (3H-D3) in mice with different administration routes to study its distribution in liver, kidney, brain, plasma and gastrointestinal tract, as well as its bioavailability by i.p. and p.o. administration. In addition, we investigated the metabolic stability in liver microsomes, mouse plasma, brain, liver and kidney homogenates, and estimated the plasma protein binding. Based on its high stability and long biological half-life, our pharmacokinetic results support the therapeutic potential of D-peptides in general, with D3 being a new promising drug candidate for Alzheimer´s disease treatment.


European Journal of Pharmaceutical Sciences | 2016

Pharmacokinetic properties of tandem d-peptides designed for treatment of Alzheimer's disease

Leonie H. E. Leithold; Nan Jiang; Julia Post; Nicole Niemietz; Elena Schartmann; Tamar Ziehm; Janine Kutzsche; N. Jon Shah; Jörg Breitkreutz; Karl-Josef Langen; Antje Willuweit; Dieter Willbold

Peptides are more and more considered for the development of drug candidates. However, they frequently exhibit severe disadvantages such as instability and unfavourable pharmacokinetic properties. Many peptides are rapidly cleared from the organism and oral bioavailabilities as well as in vivo half-lives often remain low. In contrast, some peptides consisting solely of d-enantiomeric amino acid residues were shown to combine promising therapeutic properties with high proteolytic stability and enhanced pharmacokinetic parameters. Recently, we have shown that D3 and RD2 have highly advantageous pharmacokinetic properties. Especially D3 has already proven promising properties suitable for treatment of Alzheimers disease. Here, we analyse the pharmacokinetic profiles of D3D3 and RD2D3, which are head-to-tail tandem d-peptides built of D3 and its derivative RD2. Both D3D3 and RD2D3 show proteolytic stability in mouse plasma and organ homogenates for at least 24h and in murine and human liver microsomes for 4h. Notwithstanding their high affinity to plasma proteins, both peptides are taken up into the brain following i.v. as well as i.p. administration. Although both peptides contain identical d-amino acid residues, they are arranged in a different sequence order and the peptides show differences in pharmacokinetic properties. After i.p. administration RD2D3 exhibits lower plasma clearance and higher bioavailability than D3D3. We therefore concluded that the amino acid sequence of RD2 leads to more favourable pharmacokinetic properties within the tandem peptide, which underlines the importance of particular sequence motifs, even in short peptides, for the design of further therapeutic d-peptides.


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.


European Journal of Pharmaceutical Sciences | 2017

Surprisingly high stability of the Aβ oligomer eliminating all-d-enantiomeric peptide D3 in media simulating the route of orally administered drugs

Anne Elfgen; Beatrix Santiago-Schübel; Lothar Gremer; Janine Kutzsche; Dieter Willbold

Abstract The aggregation of the amyloid &bgr; protein (A&bgr;) plays an important role in the pathology of Alzheimers disease. Previously, we have developed the all‐d‐enantiomeric peptide D3, which is able to eliminate neurotoxic A&bgr; oligomers in vitro and improve cognition in a transgenic Alzheimers disease mouse model in vivo even after oral administration. d‐Peptides are expected to be more resistant against enzymatic proteolysis compared to their l‐enantiomeric equivalents, and indeed, a pharmacokinetic study with tritiated D3 revealed the oral bioavailability to be about 58%. To further investigate the underlying properties, we examined the stability of D3 in comparison to its corresponding all‐l‐enantiomeric mirror image l‐D3 in media simulating the gastrointestinal tract, blood and liver. Potential metabolization was followed by reversed‐phase high‐performance liquid chromatography. In simulated gastric fluid, D3 remained almost completely stable (89%) within 24 h, while 70% of l‐D3 was degraded within the same time period. Notably, in simulated intestinal fluid, D3 also remained stable (96%) for 24 h, whereas l‐D3 was completely metabolized within seconds. In human plasma and human liver microsomes, l‐D3 was metabolized several hundred times faster than D3. The remarkably high stability may explain the high oral bioavailability seen in previous studies allowing oral administration of the drug candidate. Thus, all‐d‐enantiomeric peptides may represent a promising new compound class for drug development. Graphical abstract Figure. No Caption available.


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.


Analytical and Bioanalytical Chemistry | 2015

Structure characterization of unexpected covalent O-sulfonation and ion-pairing on an extremely hydrophilic peptide with CE-MS and FT-ICR-MS.

Martin Pattky; Simone Nicolardi; Beatrix Santiago-Schübel; Daniel Sydes; Yuri E. M. van der Burgt; Antonia Nicole Klein; Nan Jiang; Jeannine Mohrlüder; Karen Hänel; Janine Kutzsche; Susanne Aileen Funke; Dieter Willbold; Sabine Willbold; Carolin Huhn

In this study, we characterized unexpected side-products in a commercially synthesized peptide with the sequence RPRTRLHTHRNR. This so-called peptide D3 was selected by mirror phage display against low molecular weight amyloid-β-peptide (Aβ) associated with Alzheimer’s disease. Capillary electrophoresis (CE) was the method of choice for structure analysis because the extreme hydrophilicity of the peptide did not allow reversed-phase liquid chromatography (RPLC) and hydrophilic interaction stationary phases (HILIC). CE-MS analysis, applying a strongly acidic background electrolyte and different statically adsorbed capillary coatings, provided fast and efficient analysis and revealed that D3 unexpectedly showed strong ion-pairing with sulfuric acid. Moreover, covalent O-sulfonation at one or two threonine residues was identified as a result of a side reaction during peptide synthesis, and deamidation was found at either the asparagine residue or at the C-terminus. In total, more than 10 different species with different m/z values were observed. Tandem-MS analysis with collision induced dissociation (CID) using a CE-quadrupole-time-of-flight (QTOF) setup predominantly resulted in sulfate losses and did not yield any further characteristic fragment ions at high collision energies. Therefore, direct infusion Fourier transform ion cyclotron resonance (FT-ICR) MS was employed to identify the covalent modification and discriminate O-sulfonation from possible O-phosphorylation by using an accurate mass analysis. Electron transfer dissociation (ETD) was used for the identification of the threonine O-sulfation sites. In this work, it is shown that the combination of CE-MS and FT-ICR-MS with ETD fragmentation was essential for the full characterization of this extremely basic peptide with labile modifications.


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.

Collaboration


Dive into the Janine Kutzsche's collaboration.

Top Co-Authors

Avatar

Dieter Willbold

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar

Antje Willuweit

Forschungszentrum Jülich

View shared research outputs
Top Co-Authors

Avatar

Tamar Ziehm

Forschungszentrum Jülich

View shared research outputs
Top Co-Authors

Avatar

Sarah Schemmert

Forschungszentrum Jülich

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lothar Gremer

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar

Markus Tusche

Forschungszentrum Jülich

View shared research outputs
Top Co-Authors

Avatar

Nan Jiang

Forschungszentrum Jülich

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge