Network


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

Hotspot


Dive into the research topics where Dirk Bartnik is active.

Publication


Featured researches published by Dirk Bartnik.


ACS Chemical Neuroscience | 2010

Oral treatment with the d-enantiomeric peptide D3 improves the pathology and behavior of Alzheimer's Disease transgenic mice.

Susanne Aileen Funke; Thomas van Groen; Inga Kadish; Dirk Bartnik; Luitgard Nagel-Steger; Oleksandr Brener; Torsten Sehl; Renu Batra-Safferling; Christine Moriscot; Guy Schoehn; Anselm H. C. Horn; Andreas Müller-Schiffmann; Carsten Korth; Heinrich Sticht; Dieter Willbold

Several lines of evidence suggest that the amyloid-β-peptide (Aβ) plays a central role in the pathogenesis of Alzheimers disease (AD). Not only Aβ fibrils but also small soluble Aβ oligomers in particular are suspected to be the major toxic species responsible for disease development and progression. The present study reports on in vitro and in vivo properties of the Aβ targeting d-enantiomeric amino acid peptide D3. We show that next to plaque load and inflammation reduction, oral application of the peptide improved the cognitive performance of AD transgenic mice. In addition, we provide in vitro data elucidating the potential mechanism underlying the observed in vivo activity of D3. These data suggest that D3 precipitates toxic Aβ species and converts them into nonamyloidogenic, nonfibrillar, and nontoxic aggregates without increasing the concentration of monomeric Aβ. Thus, D3 exerts an interesting and novel mechanism of action that abolishes toxic Aβ oligomers and thereby supports their decisive role in AD development and progression.


Angewandte Chemie | 2010

Combining Independent Drug Classes into Superior, Synergistically Acting Hybrid Molecules

Andreas Müller-Schiffmann; Julia März-Berberich; Aksana Andreyeva; Raik Rönicke; Dirk Bartnik; Oleksandr Brener; Janine Kutzsche; Anselm H. C. Horn; Marco Hellmert; Jolanta Polkowska; Kurt Gottmann; Klaus G. Reymann; S. Aileen Funke; Luitgart Nagel‐Steger; Christine Moriscot; Guy Schoehn; Heinrich Sticht; Dieter Willbold; Thomas Schrader; Carsten Korth

Increasing the potency of synthesized drugs has been a stepwise process accomplished by progressively modifying the chemical scaffold of a single parent lead compound. To date, there has been no basis for thinking that the combination of pharmacological effects of independently acting drugs could be achieved beyond mere simultaneous administration. We reasoned that if the target molecule of two independent classes of drugs was the same, chemical synthesis of a hybrid compound where these drugs presented moieties within one molecule might yield synergistic effects; that is, a new quality might emerge that would be more than the sum of the singlemoiety compounds. Such multifunctional hybrid compounds that assign different functions to its different moieties to achieve a synergistic pharmacodynamic effect have successful predecessors in nature: for example, bleomycin is a natural compound with three different moieties acting in concert to cleave DNA.


Rejuvenation Research | 2010

Differently Selected d-Enantiomeric Peptides Act on Different Aβ Species

Dirk Bartnik; Susanne Aileen Funke; Luminita-Cornelia Andrei-Selmer; Michael Bacher; Richard Dodel; Dieter Willbold

Aging is the most significant risk factor for Alzheimer disease (AD). The pathological hallmark of AD is the accumulation of aggregated amyloid-beta (Abeta) forms and insoluble plaques, mainly composed of Abeta, in the brain of the patient. Recently, we reported on the selection of D-enantiomeric, Abeta-binding peptides D1 and D3. D1 was selected against aggregated Abeta species to address diagnosis by in vivo imaging of amyloid plaques, whereas D3 was selected using low-molecular-weight Abeta species, therefore addressing therapeutical studies. Here, we use a surface plasmon resonance method to confirm that both peptides show the desired binding specificities.


Journal of Alzheimer's Disease | 2013

Treatment with D3 removes amyloid deposits, reduces inflammation, and improves cognition in aged AβPP/PS1 double transgenic mice.

Thomas van Groen; Inga Kadish; Susanne Aileen Funke; Dirk Bartnik; Dieter Willbold

One of the characteristic pathological hallmarks of Alzheimers disease (AD) is neuritic plaques. The sequence of events leading to deposition of amyloid-β (Aβ) peptides in plaques is not clear. Here we investigate the effects of D3, an Aβ oligomer directed D-enantiomeric peptide that was obtained from a mirror image phage display selection against monomeric or small oligomeric forms of Aβ42, on Aβ deposition in aged AβPP/PS1 double transgenic AD-model mice. Using Alzet minipumps, we infused the brains of these AD model mice for 8 weeks with FITC-labeled D3, and examined the subsequent changes in pathology and cognitive deficits. Initial cognitive deficits are similar comparing control and D3-FITC-treated mice, but the treated mice show a significant improvement on the last day of testing. Further, we show that there is a substantial reduction in the amount of amyloid deposits in the animals treated with D3-FITC, compared to the control mice. Finally, the amount of activated microglia and astrocytes surrounding Aβ deposits is dramatically reduced in the D3-FITC-treated mice. Our findings demonstrate that treatments with the high affinity Aβ42 oligomer binding D-enantiomeric peptide D3 significantly decrease Aβ deposits and the associated inflammatory response, and improve cognition even when applied only at late stages and high age. Together, this suggests that the treatment reduces the level of Aβ peptide in the brains of AβPP/PS1 mice, possibly by increasing Aβ outflow from the brain. In conclusion, treatments with this D-peptide have great potential to be successful in AD patients.


Advances in Protein Chemistry | 2012

Treatment with Aβ42 binding D-amino acid peptides reduce amyloid deposition and inflammation in APP/PS1 double transgenic mice.

Thomas van Groen; Inga Kadish; Aileen Funke; Dirk Bartnik; Dieter Willbold

One of the two characteristic pathological hallmarks of Alzheimers disease (AD) are neuritic plaques. The sequence of events leading to the extracellular deposition of amyloid β (Aβ) peptides in plaques or in diffuse deposits is not clear. Here we investigate the relationship between aggregation and deposition of Aβ by using peptides that bind to Aβ as antifibrillization treatments in APP/PS1 double transgenic AD-model mice. Using Alzet minipumps, we infused the brain of these AD-model mice for 4 weeks with one of the three small D-amino acid peptides (i.e., D1, D3, or D3-FITC) that were designed to bind specifically to Aβ42, and examined the subsequent improvement in cognitive deficits after 3 weeks and analyzed amyloid deposition in the brain following the behavioral analysis. Cognitive deficits are similar comparing control and D3-treated mice, but D1-treated mice are slightly, but significantly, impaired. In contrast, there is a substantial improvement in the cognitive deficits in the animals treated with D3-FITC, compared to the other mice. In contrast, we show that there is a substantial reduction in the amount of amyloid deposits in the animals treated with D3, compared to the other groups of mice. Furthermore, the amount of activated microglia and astrocytes surrounding Aβ deposits is dramatically reduced in both the D3- and D3-FITC-treated mice. Our findings demonstrate that treatments with a high-affinity Aβ-42-binding D-amino acid peptide significantly decrease Aβ deposits and the associated inflammatory response. Together, this suggests that aggregation likely plays an important role in the deposition of Aβ protein in APP/PS1 transgenic mice and that antiaggregation treatments with D-peptides may be successful in AD patients.


Current Medicinal Chemistry | 2014

Amyloid Aggregation Inhibitory Mechanism of Arginine-rich D-peptides

Olujide O. Olubiyi; Daniel Frenzel; Dirk Bartnik; Julian Glück; Oleksandr Brener; Luitgart Nagel‐Steger; Susanne Aileen Funke; Dieter Willbold; Birgit Strodel

It is widely believed that Alzheimers disease pathogenesis is driven by the production and deposition of the amyloid-β peptide (Aβ) in the brain. In this study, we employ a combination of in silico and in vitro approaches to investigate the inhibitory properties of selected arginine-rich D-enantiomeric peptides (D-peptides) against amyloid aggregation. The D-peptides include D3, a 12-residue peptide with anti-amyloid potencies demonstrated in vitro and in vivo, RD2, a scrambled sequence of D3, as well as truncated RD2 variants. Using a global optimization method together with binding free energy calculations followed by molecular dynamics simulations, we perform a detailed analysis of D-peptide binding to Aβ monomer and a fibrillar Aβ structure. Results obtained from both molecular simulations and surface plasmon resonance experiments reveal a strong binding of D3 and RD2 to Aβ, leading to a significant reduction in the amount of β structures in both monomer and fibril, which was also demonstrated in Thioflavin T assays. The binding of the D-peptides to Aβ is driven by electrostatic interactions, mostly involving the D-arginine residues and Glu11, Glu22 and Asp23 of Aβ. Furthermore, we show that the anti-amyloid activities of the D-peptides depend on the length and sequence of the Dpeptide, its ability to form multiple weak hydrophobic interactions with Aβ, as well as the Aβ oligomer size.


PLOS ONE | 2012

Development of a Small D-Enantiomeric Alzheimer’s Amyloid-β Binding Peptide Ligand for Future In Vivo Imaging Applications

Susanne Aileen Funke; Dirk Bartnik; Julian Glück; Kasia Piorkowska; Katja Wiesehan; Urs Weber; Balázs Gulyás; Christer Halldin; Andrea Pfeifer; Christian Spenger; Andreas Muhs; Dieter Willbold

Alzheimer’s disease (AD) is a devastating disease affecting predominantly the aging population. One of the characteristic pathological hallmarks of AD are neuritic plaques, consisting of amyloid-β peptide (Aβ). While there has been some advancement in diagnostic classification of AD patients according to their clinical severity, no fully reliable method for pre-symptomatic diagnosis of AD is available. To enable such early diagnosis, which will allow the initiation of treatments early in the disease progress, neuroimaging tools are under development, making use of Aβ-binding ligands that can visualize amyloid plaques in the living brain. Here we investigate the properties of a newly designed series of D-enantiomeric peptides which are derivatives of ACI-80, formerly called D1, which was developed to specifically bind aggregated Aβ1–42. We describe ACI-80 derivatives with increased stability and Aβ binding properties, which were characterized using surface plasmon resonance and enzyme-linked immunosorbent assays. The specific interactions of the lead compounds with amyloid plaques were validated by ex vivo immunochemistry in transgenic mouse models of AD. The novel compounds showed increased binding affinity and are promising candidates for further development into in vivo imaging compounds.


Aging Cell | 2012

A synthetic amino acid substitution of Tyr10 in Aβ peptide sequence yields a dominant negative variant in amyloidogenesis

Honoree Mazargui; Christian Lévêque; Dirk Bartnik; Jacques Fantini; Tiphany Gouget; Mariarosa A. B. Melone; Susanne Aileen Funke; Dieter Willbold; Lorena Perrone

Alzheimer’s disease (AD) is the most common cause of dementia in elderly people, and age is the major nongenetic risk factor for sporadic AD. A hallmark of AD is the accumulation of amyloid in the brain, which is composed mainly of the amyloid beta‐peptide (Aβ) in the form of oligomers and fibrils. However, how aging induces Aβ aggregation is not yet fully determined. Some residues in the Aβ sequence seem to promote Aβ‐induced toxicity in association with age‐dependent risk factors for AD, such as (i) increased GM1 brain membrane content, (ii) altered lipid domain in brain membrane, (iii) oxidative stress. However, the role of Aβ sequence in promoting aggregation following interaction with the plasma membrane is not yet demonstrated. As Tyr10 is implicated in the induction of oxidative stress and stabilization of Aβ aggregation, we substituted Tyr 10 with a synthetic amino acid that abolishes Aβ‐induced oxidative stress and shows an accelerated interaction with GM1. This variant peptide shows impaired aggregation properties and increased affinity for GM1. It has a dominant negative effect on amyloidogenesis in vitro, in cellulo, and in isolated synaptosomes. The present study shed new light in the understanding of Aβ‐membrane interactions in Aβ‐induced neurotoxicity. It demonstrates the relevance of Aβ sequence in (i) Aβ‐membrane interaction, underlining the role of age‐dependent enhanced GM1 content in promoting Aβ aggregation, (ii) Aβ aggregation, and (iii) Aβ‐induced oxidative stress. Our results open the way for the design of peptides aimed to inhibit Aβ aggregation and neurotoxicity.


Rejuvenation Research | 2012

Identification and Characterization of an Aβ Oligomer Precipitating Peptide That May Be Useful to Explore Gene Therapeutic Approaches to Alzheimer Disease

Susanne Aileen Funke; Hongmei Liu; Torsten Sehl; Dirk Bartnik; Oleksandr Brener; Luitgard Nagel-Steger; Katja Wiesehan; Dieter Willbold

A key feature of Alzheimer disease (AD) is the pathologic self-association of the amyloid-β (Aβ) peptide, leading to the formation of diffusible toxic Aβ oligomers and extracellular amyloid plaques. Next to extracellular Aβ, intraneuronal Aβ has important pathological functions in AD. Agents that specifically interfere with the oligomerization processes either outside or inside of neurons are highly desired for the elucidation of the pathologic mechanisms of AD and might even pave the way for new AD gene therapeutic approaches. Here, we characterize the Aβ binding peptide L3 and its influence on Aβ oligomerization in vitro. Preliminary studies in cell culture demonstrate that stably expressed L3 reduces cell toxicity of externally added Aβ in neuroblastoma cells.


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 Dirk Bartnik's collaboration.

Top Co-Authors

Avatar

Dieter Willbold

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carsten Korth

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anselm H. C. Horn

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Heinrich Sticht

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Thomas Schrader

University of Duisburg-Essen

View shared research outputs
Top Co-Authors

Avatar

Inga Kadish

University of Alabama at Birmingham

View shared research outputs
Researchain Logo
Decentralizing Knowledge