Network


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

Hotspot


Dive into the research topics where Wolfgang Hoyer is active.

Publication


Featured researches published by Wolfgang Hoyer.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Stabilization of a beta-hairpin in monomeric Alzheimer's amyloid-beta peptide inhibits amyloid formation.

Wolfgang Hoyer; Caroline Grönwall; Andreas Jonsson; Stefan Ståhl; Torleif Härd

According to the amyloid hypothesis, the pathogenesis of Alzheimers disease is triggered by the oligomerization and aggregation of the amyloid-β (Aβ) peptide into protein plaques. Formation of the potentially toxic oligomeric and fibrillar Aβ assemblies is accompanied by a conformational change toward a high content of β-structure. Here, we report the solution structure of Aβ(1–40) in complex with the phage-display selected affibody protein ZAβ3, a binding protein of nanomolar affinity. Bound Aβ(1–40) features a β-hairpin comprising residues 17–36, providing the first high-resolution structure of Aβ in β conformation. The positions of the secondary structure elements strongly resemble those observed for fibrillar Aβ. ZAβ3 stabilizes the β-sheet by extending it intermolecularly and by burying both of the mostly nonpolar faces of the Aβ hairpin within a large hydrophobic tunnel-like cavity. Consequently, ZAβ3 acts as a stoichiometric inhibitor of Aβ fibrillation. The selected Aβ conformation allows us to suggest a structural mechanism for amyloid formation based on soluble oligomeric hairpin intermediates.


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.


PLOS Biology | 2010

Sequestration of the Aβ Peptide Prevents Toxicity and Promotes Degradation In Vivo

Leila M. Luheshi; Wolfgang Hoyer; Teresa P. Barros; Iris van Dijk Härd; Ann-Christin Brorsson; Bertil Macao; Cecilia Persson; Damian C. Crowther; David A. Lomas; Stefan Ståhl; Christopher M. Dobson; Torleif Härd

An engineered protein prevents aggregation of the Aβ peptide and facilitates clearance of Aβ from the brain in a fruit fly model of Alzheimers disease.


Angewandte Chemie | 2014

Sequestration of a β-hairpin for control of α-synuclein aggregation.

Ewa A. Mirecka; Hamed Shaykhalishahi; Aziz Gauhar; Şerife Akgül; Justin Lecher; Dieter Willbold; Matthias Stoldt; Wolfgang Hoyer

The misfolding and aggregation of the protein α-synuclein (α-syn), which results in the formation of amyloid fibrils, is involved in the pathogenesis of Parkinsons disease and other synucleinopathies. The emergence of amyloid toxicity is associated with the formation of partially folded aggregation intermediates. Here, we engineered a class of binding proteins termed β-wrapins (β-wrap proteins) with affinity for α-synuclein (α-syn). The NMR structure of an α-syn:β-wrapin complex reveals a β-hairpin of α-syn comprising the sequence region α-syn(37-54). The β-wrapin inhibits α-syn aggregation and toxicity at substoichiometric concentrations, demonstrating that it interferes with the nucleation of aggregation.


BMC Biotechnology | 2008

Recombinant amyloid beta-peptide production by coexpression with an affibody ligand.

Bertil Macao; Wolfgang Hoyer; Anders Sandberg; Ann-Christin Brorsson; Christopher M. Dobson; Torleif Härd

BackgroundOligomeric and fibrillar aggregates of the amyloid β-peptide (Aβ) have been implicated in the pathogenesis of Alzheimers disease (AD). The characterization of Aβ assemblies is essential for the elucidation of the mechanisms of Aβ neurotoxicity, but requires large quantities of pure peptide. Here we describe a novel approach to the recombinant production of Aβ. The method is based on the coexpression of the affibody protein ZAβ3, a selected affinity ligand derived from the Z domain three-helix bundle scaffold. ZAβ3 binds to the amyloidogenic central and C-terminal part of Aβ with nanomolar affinity and consequently inhibits aggregation.ResultsCoexpression of ZAβ3 affords the overexpression of both major Aβ isoforms, Aβ(1–40) and Aβ(1–42), yielding 4 or 3 mg, respectively, of pure 15N-labeled peptide per liter of culture. The method does not rely on a protein-fusion or -tag and thus does not require a cleavage reaction. The purified peptides were characterized by NMR, circular dichroism, SDS-PAGE and size exclusion chromatography, and their aggregation propensities were assessed by thioflavin T fluorescence and electron microscopy. The data coincide with those reported previously for monomeric, largely unstructured Aβ. ZAβ3 coexpression moreover permits the recombinant production of Aβ(1–42) carrying the Arctic (E22G) mutation, which causes early onset familial AD. Aβ(1–42)E22G is obtained in predominantly monomeric form and suitable, e.g., for NMR studies.ConclusionThe coexpression of an engineered aggregation-inhibiting binding protein offers a novel route to the recombinant production of amyloidogenic Aβ peptides that can be advantageously employed to study the molecular basis of AD. The presented expression system is the first for which expression and purification of the aggregation-prone Arctic variant (E22G) of Aβ(1–42) is reported.


Journal of Molecular Biology | 2008

Interaction of Alzheimer’s Aβ Peptide with an Engineered Binding Protein—Thermodynamics and Kinetics of Coupled Folding–Binding

Wolfgang Hoyer; Torleif Härd

The oligomerization and aggregation of the amyloid-beta (A beta) peptide, a cleavage product of the amyloid precursor protein predominantly 40 or 42 amino acids in length, has been implicated in the pathogenesis of Alzheimers disease. The identification of A beta-binding agents, e.g., antibodies or peptides, constitutes a promising therapeutic approach. However, the amount of structural and biophysical data on the underlying A beta interactions is currently very limited. We have earlier determined the structure of A beta (1-40) in complex with the affibody protein Z(A beta 3), a selected binding protein based on a three-helix bundle scaffold (Z domain). Z(A beta 3) is a dimer of affibody subunits linked via a disulfide bridge involving a selected cysteine mutation at position 28. Z(A beta 3) binds to the central and C-terminal part of A beta (residues 17-36), which adopts a beta-hairpin conformation in the complex. Here we present a detailed biophysical analysis of the Z(A beta 3):A beta (1-40) interaction, employing NMR, circular dichroism spectroscopy, 8-anilino-1-naphthalenesulfonic acid and tyrosine fluorescence, size-exclusion chromatography, thermal denaturation profiles and isothermal titration calorimetry. We conclude that (i) free Z(A beta 3) is characterized by conformational exchange and the loss of helix 1 of the three-helix bundle scaffold; (ii) a high-energy barrier is associated with the conversion of an initial Z(A beta 3):A beta (1-40) recognition complex into the native complex structure, entailing slow binding kinetics; (iii) both A beta and Z(A beta 3) fold upon binding, which, e.g., becomes manifest in the binding thermodynamics that feature a large negative change in heat capacity; (iv) the C28-disulfide does not merely afford dimerization, but its impact on the binding interfaces of the affibody subunits and A beta is a prerequisite for tight binding. The extensive folding coupled to binding observed here likely constitutes an obligate feature of biomolecular interactions involving the central and C-terminal part of A beta. Options for improvement of Z(A beta) binding proteins are discussed.


Biophysical Chemistry | 2002

A kinetic trap is an intrinsic feature in the folding pathway of single-chain Fv fragments.

Wolfgang Hoyer; Kathrin Ramm; Andreas Plückthun

We have studied the equilibrium unfolding and the kinetics of folding and unfolding of an antibody scFv fragment devoid of cis-prolines. An anti-GCN4 scFv fragment carrying a VL lambda domain, obtained by ribosome display, served as the model system together with an engineered destabilized mutant in VH carrying the R66K exchange. Kinetic and equilibrium unfolding experiments indicate that the VH mutation also affects VL unfolding, possibly by partially destabilizing the interface provided by VH, even though the mutation is distant from the interface. Upon folding of the scFv fragment, a kinetic trap is populated whose escape rate is much faster with the more stable VH domain. The formation of the trap can be avoided if refolding is carried out stepwise, with VH folding first. These results show that antibody scFv fragments do not fold by the much faster independent domain folding, but instead form a kinetically trapped off-pathway intermediate, which slows down folding under native conditions. This intermediate is characterized by premature interaction of the unfolded domains, and particularly involving unfolded VH, independent of proline cis-trans isomerization in VL. This work also implies that VH should be a prime target in engineering well behaving antibody fragments.


Journal of Molecular Biology | 2015

Single Fibril Growth Kinetics of α-Synuclein

Michael M. Wördehoff; Oliver Bannach; Hamed Shaykhalishahi; Andreas Kulawik; Stephanie Schiefer; Dieter Willbold; Wolfgang Hoyer; Eva Birkmann

Neurodegenerative disorders associated with protein misfolding are fatal diseases that are caused by fibrillation of endogenous proteins such as α-synuclein (α-syn) in Parkinsons disease (PD) or amyloid-β in Alzheimers disease. Fibrils of α-syn are a major pathological hallmark of PD and certain aggregation intermediates are postulated to cause synaptic failure and cell death of dopaminergic neurons in the substantia nigra. For the development of therapeutic approaches, the mechanistic understanding of the fibrillation process is essential. Here we report real-time observation of α-syn fibril elongation on a glass surface, imaged by total internal reflection fluorescence microscopy using thioflavin T fluorescence. Fibrillation on the glass surface occurred in the same time frame and yielded fibrils of similar length as fibrillation in solution. Time-resolved imaging of fibrillation on a single fibril level indicated that α-syn fibril elongation follows a stop-and-go mechanism; that is, fibrils either extend at a homogenous growth rate or stop to grow for variable time intervals. The fibril growth kinetics were compatible with a model featuring two states, a growth state and a stop state, which were approximately isoenergetic and interconverted with rate constants of ~1.5×10(-4) s(-1). In the growth state, α-syn monomers were incorporated into the fibril with a rate constant of 8.6×10(3) M(-1) s(-1). Fibril elongation of α-syn is slow compared to other amyloidogenic proteins.


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.


Journal of Biological Chemistry | 2013

The Off-rate of Monomers Dissociating from Amyloid-β Protofibrils

Clara S. R. Grüning; Stefan Klinker; Martin Wolff; Mario Schneider; Küpra Toksöz; Antonia Nicole Klein; Luitgard Nagel-Steger; Dieter Willbold; Wolfgang Hoyer

Background: Protofibrils of the amyloid-β peptide (Aβ) are neurotoxic oligomers implicated in development and progression of Alzheimer disease. Results: The dissociation of Aβ protofibrils into their monomeric subunits is a slow process, occurring on the time scale of hours. Conclusion: Aβ protofibrils possess a high kinetic stability toward dissociation into monomers. Significance: The longevity of Aβ protofibrils permits sustained toxic effects. The interconversion of monomers, oligomers, and amyloid fibrils of the amyloid-β peptide (Aβ) has been implicated in the pathogenesis of Alzheimer disease. The determination of the kinetics of the individual association and dissociation reactions is hampered by the fact that forward and reverse reactions to/from different aggregation states occur simultaneously. Here, we report the kinetics of dissociation of Aβ monomers from protofibrils, prefibrillar high molecular weight oligomers previously shown to possess pronounced neurotoxicity. An engineered binding protein sequestering specifically monomeric Aβ was employed to follow protofibril dissociation by tryptophan fluorescence, precluding confounding effects of reverse or competing reactions. Aβ protofibril dissociation into monomers follows exponential decay kinetics with a time constant of ∼2 h at 25 °C and an activation energy of 80 kJ/mol, values typical for high affinity biomolecular interactions. This study demonstrates the high kinetic stability of Aβ protofibrils toward dissociation into monomers and supports the delineation of the Aβ folding and assembly energy landscape.

Collaboration


Dive into the Wolfgang Hoyer's collaboration.

Top Co-Authors

Avatar

Dieter Willbold

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ewa A. Mirecka

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar

Matthias Stoldt

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar

Lothar Gremer

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar

Henrike Heise

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aziz Gauhar

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar

Torleif Härd

Swedish University of Agricultural Sciences

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge