59 results on '"Andreas, Ebneth"'
Search Results
2. Pharmacological characterization of mutant huntingtin aggregate-directed PET imaging tracer candidates
- Author
-
Frank Herrmann, Manuela Hessmann, Sabine Schaertl, Karola Berg-Rosseburg, Christopher J Brown, Galina Bursow, Anass Chiki, Andreas Ebneth, Miriam Gehrmann, Nicole Hoeschen, Madlen Hotze, Stefanie Jahn, Peter D Johnson, Vinod Khetarpal, Alex Kiselyov, Karsten Kottig, Stefanie Ladewig, Hilal Lashuel, Sven Letschert, Matthew R Mills, Kathrin Petersen, Michael E Prime, Christoph Scheich, Gerhard Schmiedel, John Wityak, Longbin Liu, Celia Dominguez, Ignacio Muñoz-Sanjuán, and Jonathan A Bard
- Subjects
Medicine ,Science - Abstract
Abstract Huntington’s disease (HD) is caused by a CAG trinucleotide repeat expansion in the first exon of the huntingtin (HTT) gene coding for the huntingtin (HTT) protein. The misfolding and consequential aggregation of CAG-expanded mutant HTT (mHTT) underpin HD pathology. Our interest in the life cycle of HTT led us to consider the development of high-affinity small-molecule binders of HTT oligomerized/amyloid-containing species that could serve as either cellular and in vivo imaging tools or potential therapeutic agents. We recently reported the development of PET tracers CHDI-180 and CHDI-626 as suitable for imaging mHTT aggregates, and here we present an in-depth pharmacological investigation of their binding characteristics. We have implemented an array of in vitro and ex vivo radiometric binding assays using recombinant HTT, brain homogenate-derived HTT aggregates, and brain sections from mouse HD models and humans post-mortem to investigate binding affinities and selectivity against other pathological proteins from indications such as Alzheimer’s disease and spinocerebellar ataxia 1. Radioligand binding assays and autoradiography studies using brain homogenates and tissue sections from HD mouse models showed that CHDI-180 and CHDI-626 specifically bind mHTT aggregates that accumulate with age and disease progression. Finally, we characterized CHDI-180 and CHDI-626 regarding their off-target selectivity and binding affinity to beta amyloid plaques in brain sections and homogenates from Alzheimer’s disease patients.
- Published
- 2021
- Full Text
- View/download PDF
3. Development of a fully human assay combining NGN2-inducible neurons co-cultured with iPSC-derived astrocytes amenable for electrophysiological studies
- Author
-
Pei-Yu Shih, Mohamed Kreir, Devesh Kumar, Frederik Seibt, Francisco Pestana, Benjamin Schmid, Bjørn Holst, Christian Clausen, Rachel Steeg, Benjamin Fischer, Juan Pita-Almenar, Andreas Ebneth, and Alfredo Cabrera-Socorro
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Neurogenin 2 encodes a neural-specific transcription factor (NGN2) able to drive neuronal fate on somatic and stem cells. NGN2 is expressed in neural progenitors within the developing central and peripheral nervous systems. Overexpression of NGN2 in human induced pluripotent stem cells (hiPSCs) or human embryonic stem cells has been shown to efficiently trigger conversion to neurons. Here we describe two gene-edited hiPSC lines harbouring a doxycycline (DOX)-inducible cassette in the AAVS1 locus driving expression of NGN2 (BIONi010-C-13) or NGN2-T2A-GFP (BIONi010-C-15). By introducing NGN2-expressing cassette, we reduce variability associated with conventional over-expression methods such as viral transduction, making these lines amenable for scale-up production and screening processes. DOX-treated hiPSCs convert to neural phenotype within one week and display the expression of structural neuronal markers such as Beta-III tubulin and tau. We performed functional characterization of NGN2-neurons co-cultured with hiPSC-derived astrocytes in a “fully-humanized” set up. Passive properties of NGN2-neurons were indistinguishable from mouse primary cells while displaying variable activity in extracellular recordings performed in multi-electrode arrays (MEAs). We demonstrate that hiPSC-derived astrocytes and neurons can be co-cultured and display functional properties comparable to the gold standard used in electrophysiology. Both lines are globally available via EBiSC repository at https://cells.ebisc.org/.
- Published
- 2021
- Full Text
- View/download PDF
4. Small Molecule Binding to Alzheimer Risk Factor CD33 Promotes Aβ Phagocytosis
- Author
-
Luke A. Miles, Stefan J. Hermans, Gabriela A.N. Crespi, Jonathan H. Gooi, Larissa Doughty, Tracy L. Nero, Jasmina Markulić, Andreas Ebneth, Berthold Wroblowski, Daniel Oehlrich, Andrés A. Trabanco, Marie-Laure Rives, Ines Royaux, Nancy C. Hancock, and Michael W. Parker
- Subjects
Science - Abstract
Summary: Polymorphism in the microglial receptor CD33 gene has been linked to late-onset Alzheimer disease (AD), and reduced expression of the CD33 sialic acid-binding domain confers protection. Thus, CD33 inhibition might be an effective therapy against disease progression. Progress toward discovery of selective CD33 inhibitors has been hampered by the absence of an atomic resolution structure. We report here the crystal structures of CD33 alone and bound to a subtype-selective sialic acid mimetic called P22 and use them to identify key binding residues by site-directed mutagenesis and binding assays to reveal the molecular basis for its selectivity toward sialylated glycoproteins and glycolipids. We show that P22, when presented on microparticles, increases uptake of the toxic AD peptide, amyloid-β (Aβ), into microglial cells. Thus, the sialic acid-binding site on CD33 is a promising pharmacophore for developing therapeutics that promote clearance of the Aβ peptide that is thought to cause AD. : Molecular Structure; Neuroscience; Molecular Neuroscience; Components of the Immune System; Protein Structure Aspects Subject Areas: Molecular Structure, Neuroscience, Molecular Neuroscience, Components of the Immune System, Protein Structure Aspects
- Published
- 2019
- Full Text
- View/download PDF
5. Generation of a set of isogenic iPSC lines carrying all APOE genetic variants (Ɛ2/Ɛ3/Ɛ4) and knock-out for the study of APOE biology in health and disease
- Author
-
Benjamin Schmid, Bjørn Holst, Christian Clausen, Lamiaa Bahnassawy, Peter Reinhardt, Margot H.M. Bakker, Eva Díaz-Guerra, Carlos Vicario, Pamela V. Martino-Adami, Michaela Thoenes, Alfredo Ramirez, Klaus Flissbach, Clara Grezella, Oliver Brüstle, Michael Peitz, Andreas Ebneth, and Alfredo Cabrera-Socorro
- Subjects
Biology (General) ,QH301-705.5 - Abstract
APOE genotype is the strongest genetic risk factor for Alzheimer’s Disease (AD). The low degree of homology between mouse and human APOE is a concerning issue in preclinical models currently used to study the role of this gene in AD pathophysiology. A key objective of ADAPTED (Alzheimer's Disease Apolipoprotein Pathology for Treatment Elucidation and Development) project was to generate in vitro models that better recapitulate human APOE biology. We describe a new set of induced pluripotent stem cells (iPSC) lines carrying common APOE variants (Ɛ2, Ɛ3, and Ɛ3/Ɛ4) and a knock-out isogenic to the parental APOE Ɛ4/Ɛ4 line (UKBi011-A).
- Published
- 2021
- Full Text
- View/download PDF
6. IP-LC-MSMS Enables Identification of Three Tau O-GlcNAcylation Sites as O-GlcNAcase Inhibition Pharmacodynamic Readout in Transgenic Mice Overexpressing Human Tau
- Author
-
Sebastiaan Bijttebier, Dina Rodrigues Martins, Liesbeth Mertens, Karolien Grauwen, Wouter Bruinzeel, Roland Willems, José Manuel Bartolomé-Nebreda, Clara Theunis, Alexis Bretteville, Andreas Ebneth, and Lieve Dillen
- Subjects
General Chemistry ,Biochemistry - Published
- 2023
- Full Text
- View/download PDF
7. The EBiSC iPSC bank for disease studies
- Author
-
Rachel Steeg, Julia C. Neubauer, Sabine C. Müller, Andreas Ebneth, and Heiko Zimmermann
- Subjects
iPSC ,EBiSC ,Induced ,Pluripotent ,Stem ,Cell ,Biology (General) ,QH301-705.5 - Abstract
The European Bank for induced Pluripotent Stem Cells (EBiSC), a non-profit repository for storage, banking, Quality Control (QC) and subsequent distribution of research-grade human induced Pluripotent Stem Cell (iPSC) lines, has centralised iPSC lines generated internationally across >35 disease areas and made them available to users via the EBiSC Catalogue, for research use (cells.ebisc.org/). Comprehensive datasets are accessible prior to purchase detailing the disease background of the original tissue sample, background of iPSC reprogramming and cell line characterisation data. EBiSC also performs robust QC screening to ensure supply of reliable, well-characterised iPSC lines, compliant with ISO9001:2015 principles. Whole Genome Sequencing data for specific iPSC lines can be downloaded from the European Genome Archive, subject to application to the EBiSC Data Access Committee. The EBiSC Access and Use Agreement, required to be completed prior to shipping, can be downloaded from the website along with specific Cell Line Information Packs; together these documents clarify how EBiSC lines can be used for research and detail any specific Third Party Obligations and/or restrictions for use which may apply. A protocol for how to culture and monitor iPSC lines including implementation of routine cell line screening is also available. A second project phase will continue collecting iPSC lines generated internationally, provide iPSC derived differentiated products using improved automation strategies for upscaling and develop the current services provided by EBiSC, including iPSC reprogramming, gene-editing and characterisation.
- Published
- 2020
- Full Text
- View/download PDF
8. Corrigendum to 'Generation of a set of isogenic, gene-edited iPSC lines homozygous for all main APOE variants and an APOE knock-out line' [Stem Cell Res. 34/1873–5061 (2019) 101349–55]
- Author
-
Benjamin Schmid, Kennie R. Prehn, Natakarn Nimsanor, Blanca Irene Aldana Garcia, Ulla Poulsen, Ida Jørring, Mikkel A. Rasmussen, Christian Clausen, Ulrike A. Mau-Holzmann, Sarayu Ramakrishna, Ravi Muddashetty, Rachel Steeg, Kevin Bruce, Peter Mackintosh, Andreas Ebneth, Bjørn Holst, and Alfredo Cabrera-Socorro
- Subjects
Biology (General) ,QH301-705.5 - Published
- 2020
- Full Text
- View/download PDF
9. Genetically Engineered iPSC-Derived FTDP-17 MAPT Neurons Display Mutation-Specific Neurodegenerative and Neurodevelopmental Phenotypes
- Author
-
An Verheyen, Annick Diels, Joke Reumers, Kirsten Van Hoorde, Ilse Van den Wyngaert, Constantin van Outryve d’Ydewalle, An De Bondt, Jacobine Kuijlaars, Louis De Muynck, Ronald De Hoogt, Alexis Bretteville, Steffen Jaensch, Arjan Buist, Alfredo Cabrera-Socorro, Selina Wray, Andreas Ebneth, Peter Roevens, Ines Royaux, and Pieter J. Peeters
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Summary: Tauopathies such as frontotemporal dementia (FTD) remain incurable to date, partially due to the lack of translational in vitro disease models. The MAPT gene, encoding the microtubule-associated protein tau, has been shown to play an important role in FTD pathogenesis. Therefore, we used zinc finger nucleases to introduce two MAPT mutations into healthy donor induced pluripotent stem cells (iPSCs). The IVS10+16 mutation increases the expression of 4R tau, while the P301S mutation is pro-aggregant. Whole-transcriptome analysis of MAPT IVS10+16 neurons reveals neuronal subtype differences, reduced neural progenitor proliferation potential, and aberrant WNT/SHH signaling. Notably, these neurodevelopmental phenotypes could be recapitulated in neurons from patients carrying the MAPT IVS10+16 mutation. Moreover, the additional pro-aggregant P301S mutation revealed additional phenotypes, such as an increased calcium burst frequency, reduced lysosomal acidity, tau oligomerization, and neurodegeneration. This series of iPSCs could serve as a platform to unravel a potential link between pathogenic 4R tau and FTD. : In this article, Verheyen and colleagues introduced 2 MAPT mutations (IVS10+16/P301S) into healthy donor iPSCs. Characterization of MAPT IVS10+16 neurons revealed neurodevelopmental neuron subtype differences, reduced neural progenitor proliferation, and aberrant WNT/SHH signaling, while the additional pro-aggregant P301S mutation revealed phenotypes more related to neurodegeneration. These iPSCs could help to unravel a potential link between pathogenic 4R tau and FTD. Keywords: FTDP-17, MAPT, iPSC, ZFN, disease modelling
- Published
- 2018
- Full Text
- View/download PDF
10. SliceMap: an algorithm for automated brain region annotation.
- Author
-
Michaël Barbier, Astrid Bottelbergs, Rony Nuydens, Andreas Ebneth, and Winnok De Vos
- Published
- 2018
- Full Text
- View/download PDF
11. Rapid establishment of the European Bank for induced Pluripotent Stem Cells (EBiSC) - the Hot Start experience
- Author
-
Paul A. De Sousa, Rachel Steeg, Elisabeth Wachter, Kevin Bruce, Jason King, Marieke Hoeve, Shalinee Khadun, George McConnachie, Julie Holder, Andreas Kurtz, Stefanie Seltmann, Johannes Dewender, Sascha Reimann, Glyn Stacey, Orla O'Shea, Charlotte Chapman, Lyn Healy, Heiko Zimmermann, Bryan Bolton, Trisha Rawat, Isobel Atkin, Anna Veiga, Bernd Kuebler, Blanca Miranda Serano, Tomo Saric, Jürgen Hescheler, Oliver Brüstle, Michael Peitz, Cornelia Thiele, Niels Geijsen, Bjørn Holst, Christian Clausen, Majlinda Lako, Lyle Armstrong, Shailesh K. Gupta, Alexander J. Kvist, Ryan Hicks, Anna Jonebring, Gabriella Brolén, Andreas Ebneth, Alfredo Cabrera-Socorro, Patrik Foerch, Martine Geraerts, Tina C. Stummann, Shawn Harmon, Carol George, Ian Streeter, Laura Clarke, Helen Parkinson, Peter W. Harrison, Adam Faulconbridge, Luca Cherubin, Tony Burdett, Cesar Trigueros, Minal J Patel, Christa Lucas, Barry Hardy, Rok Predan, Joh Dokler, Maja Brajnik, Oliver Keminer, Ole Pless, Philip Gribbon, Carsten Claussen, Annette Ringwald, Beate Kreisel, Aidan Courtney, and Timothy E. Allsopp
- Subjects
Biology (General) ,QH301-705.5 - Abstract
A fast track “Hot Start” process was implemented to launch the European Bank for Induced Pluripotent Stem Cells (EBiSC) to provide early release of a range of established control and disease linked human induced pluripotent stem cell (hiPSC) lines. Established practice amongst consortium members was surveyed to arrive at harmonised and publically accessible Standard Operations Procedures (SOPs) for tissue procurement, bio-sample tracking, iPSC expansion, cryopreservation, qualification and distribution to the research community. These were implemented to create a quality managed foundational collection of lines and associated data made available for distribution. Here we report on the successful outcome of this experience and work flow for banking and facilitating access to an otherwise disparate European resource, with lessons to benefit the international research community. eTOC: The report focuses on the EBiSC experience of rapidly establishing an operational capacity to procure, bank and distribute a foundational collection of established hiPSC lines. It validates the feasibility and defines the challenges of harnessing and integrating the capability and productivity of centres across Europe using commonly available resources currently in the field.
- Published
- 2017
- Full Text
- View/download PDF
12. Deploying human pluripotent stem cells to treat central nervous system disorders: facts, challenges and realising the potential
- Author
-
Timothy E Allsopp, Andreas Ebneth, and Alfredo Cabrera-Socorro
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Human pluripotent stem cells (hPSC) represent a unique opportunity to study fundamental biological processes in a human- and cell-specific setting. Its translational potential and the impact on human health makes this technology revolutionary. The possibility to generate stem cells from almost any somatic cell, and their capacity to be differentiated in virtually all cells of the body has been demonstrated extensively during the last decade of research. Target-centric as well as phenotypic screenings using differentiated cells have become a reality, while the use of these cells for “disease modelling” is still challenging due to the paucity of relevant and reproducible phenotypes. The combination of hPSCs with gene editing technologies aiming to e.g. reduce immunogenic response has enabled promising clinical trials that will eventually demonstrate their therapeutic potential in tissue regeneration and cancer treatment. Maximizing the therapeutic applications of hPSCs requires systematic data comparison, consensus between scientists and health care professionals, as well as a close collaboration between research labs, clinics, and regulators. The goal of this review is to provide a comprehensive outlook of the current use of hPSCs in drug development and regenerative medicine for the treatment of central nervous system (CNS) disorders. In the first part, we analyse how hPSCs are currently used in drug development and discuss their use in challenging paradigms such as neurodegeneration. In the second part we review the status of hPSCs in regenerative medicine. Finally, key challenges and pitfalls of the technology will be discussed, and strategies proposed to improve hPSC research and to benefit patients across different therapeutic areas.
- Published
- 2019
- Full Text
- View/download PDF
13. Generation of a set of isogenic, gene-edited iPSC lines homozygous for all main APOE variants and an APOE knock-out line
- Author
-
Benjamin Schmid, Kennie R. Prehn, Natakarn Nimsanor, Blanca Irene Aldana Garcia, Ulla Poulsen, Ida Jørring, Mikkel A. Rasmussen, Christian Clausen, Ulrike A. Mau-Holzmann, Sarayu Ramakrishna, Ravi Muddashetty, Rachel Steeg, Kevin Bruce, Peter Mackintosh, Andreas Ebneth, Bjørn Holst, and Alfredo Cabrera-Socorro
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Alzheimer's disease (AD) is the most frequent neurodegenerative disease amongst the elderly. The SNPs rs429358 and rs7412 in the APOE gene are the most common risk factor for sporadic AD, and there are three different alleles commonly referred to as APOE-ε2, APOE-ε3 and APOE-ε4. Induced pluripotent stem cells (iPSCs) hold great promise to model AD as such cells can be differentiated in vitro to the required cell type. Here we report the use of CRISPR/Cas9 technology employed on iPSCs from a healthy individual with an APOE-ε3/ε4 genotype to obtain isogenic APOE-ε2/ε2, APOE-ε3/ε3, APOE-ε4/ε4 lines as well as an APOE-knock-out line.
- Published
- 2019
- Full Text
- View/download PDF
14. Pharmacological characterization of mutant huntingtin aggregate-directed PET imaging tracer candidates
- Author
-
Madlen Hotze, Miriam Gehrmann, Manuela Hessmann, Longbin Liu, Christoph Scheich, Kathrin Petersen, Nicole Hoeschen, Peter Johnson, Sabine Schaertl, Stefanie Jahn, Matthew R. Mills, Ignacio Munoz-Sanjuan, Alex S. Kiselyov, John Wityak, Anass Chiki, Vinod Khetarpal, Karsten Kottig, Gerhard Schmiedel, Galina Bursow, Stefanie Ladewig, Christopher J. Brown, Karola Berg-Rosseburg, Michael Prime, Sven Letschert, Andreas Ebneth, Jonathan Bard, Celia Dominguez, Frank Herrmann, and Hilal A. Lashuel
- Subjects
exon-1 ,Huntingtin ,Mutant ,law.invention ,Mice ,Radioligand Assay ,amyloid fibrils ,Exon ,law ,alzheimers-disease ,Huntingtin Protein ,dysfunction ,Multidisciplinary ,Chemistry ,Immunohistochemistry ,Recombinant Proteins ,Cell biology ,small molecules ,Huntington Disease ,Recombinant DNA ,Medicine ,Preclinical imaging ,congenital, hereditary, and neonatal diseases and abnormalities ,brain ,Science ,Mice, Transgenic ,Protein Aggregation, Pathological ,Article ,Protein Aggregates ,Alzheimer Disease ,mental disorders ,Animals ,Humans ,Radioactive Tracers ,beta-sheet structure ,Pharmacology ,Nitrogen Radioisotopes ,toxicity ,In vitro ,nervous system diseases ,Disease Models, Animal ,polyglutamine aggregation ,Positron-Emission Tomography ,Diseases of the nervous system ,Autoradiography ,identification ,Radiopharmaceuticals ,Trinucleotide repeat expansion ,Biomarkers ,Ex vivo - Abstract
Huntington’s disease (HD) is caused by a CAG trinucleotide repeat expansion in the first exon of the huntingtin (HTT) gene coding for the huntingtin (HTT) protein. The misfolding and consequential aggregation of CAG-expanded mutant HTT (mHTT) underpin HD pathology. Our interest in the life cycle of HTT led us to consider the development of high-affinity small-molecule binders of HTT oligomerized/amyloid-containing species that could serve as either cellular and in vivo imaging tools or potential therapeutic agents. We recently reported the development of PET tracers CHDI-180 and CHDI-626 as suitable for imaging mHTT aggregates, and here we present an in-depth pharmacological investigation of their binding characteristics. We have implemented an array of in vitro and ex vivo radiometric binding assays using recombinant HTT, brain homogenate-derived HTT aggregates, and brain sections from mouse HD models and humans post-mortem to investigate binding affinities and selectivity against other pathological proteins from indications such as Alzheimer’s disease and spinocerebellar ataxia 1. Radioligand binding assays and autoradiography studies using brain homogenates and tissue sections from HD mouse models showed that CHDI-180 and CHDI-626 specifically bind mHTT aggregates that accumulate with age and disease progression. Finally, we characterized CHDI-180 and CHDI-626 regarding their off-target selectivity and binding affinity to beta amyloid plaques in brain sections and homogenates from Alzheimer’s disease patients.
- Published
- 2021
15. Scalable expansion of iPSC and their derivatives across multiple lineages
- Author
-
Chee Keong Kwok, Isabelle Sébastien, Krithika Hariharan, Ina Meiser, Jeanette Wihan, Saskia Altmaier, Isabell Karnatz, Dominic Bauer, Benjamin Fischer, Alexander Feile, Alfredo Cabrera-Socorro, Mikkel Rasmussen, Bjørn Holst, Julia C. Neubauer, Christian Clausen, Catherine Verfaillie, Andreas Ebneth, Mattias Hansson, Rachel Steeg, Heiko Zimmermann, and Publica
- Subjects
Pluripotent Stem Cells ,upscaling ,induced pluripotent stem cells ,bioprocessing ,Induced Pluripotent Stem Cells ,suspension-based bioreactor ,Cell Culture Techniques ,Cell Differentiation ,EBiSC ,differentiation ,cell processing ,Toxicology ,Bioreactors ,cell banking - Abstract
Induced pluripotent stem cell (iPSC) technology enabled the production of pluripotent stem cell lines from somatic cells from a range of known genetic backgrounds. Their ability to differentiate and generate a wide variety of cell types has resulted in their use for various biomedical applications, including toxicity testing. Many of these iPSC lines are now registered in databases and stored in biobanks such as the European Bank for induced pluripotent Stem Cells (EBiSC), which can streamline the quality control and distribution of these individual lines. To generate the quantities of cells for banking and applications like high-throughput toxicity screening, scalable and robust methods need to be developed to enable the large-scale production of iPSCs. 3D suspension culture platforms are increasingly being used by stem cell researchers, owing to a higher cell output in a smaller footprint, as well as simpler scaling by increasing culture volume. Here we describe our strategies for successful scalable production of iPSCs using a benchtop bioreactor and incubator for 3D suspension cultures, while maintaining quality attributes expected of high-quality iPSC lines. Additionally, to meet the increasing demand for "ready-to-use" cell types, we report recent work to establish robust, scalable differentiation protocols to cardiac, neural, and hepatic fate to enable EBiSC to increase available research tools. ispartof: REPRODUCTIVE TOXICOLOGY vol:112 pages:23-35 ispartof: location:United States status: published
- Published
- 2022
16. Diazaspirononane Nonsaccharide Inhibitors of O-GlcNAcase (OGA) for the Treatment of Neurodegenerative Disorders
- Author
-
Francisca Delgado, Jordi Royes, Alcira del Cerro, Fulgencio Tovar, Juan Antonio Vega, Paul Shaffer, Andrés A. Trabanco, Elena Fernández, Alexis Bretteville, Gary Tresadern, Andreas Ebneth, José Manuel Bartolomé-Nebreda, Carlos M Martínez-Viturro, Liesbeth Mertens, Marijke Somers, Aránzazu García Molina, and José Manuel Alonso
- Subjects
Male ,Models, Molecular ,Tau protein ,Context (language use) ,O-GlcNAcase ,01 natural sciences ,Catalysis ,Mice ,Structure-Activity Relationship ,03 medical and health sciences ,Drug Discovery ,Animals ,Humans ,Structure–activity relationship ,Enzyme Inhibitors ,030304 developmental biology ,chemistry.chemical_classification ,Aza Compounds ,0303 health sciences ,Molecular Structure ,biology ,Chemistry ,Mutagenesis ,Neurodegenerative Diseases ,beta-N-Acetylhexosaminidases ,0104 chemical sciences ,Mice, Inbred C57BL ,010404 medicinal & biomolecular chemistry ,Enzyme ,Biochemistry ,biology.protein ,Molecular Medicine - Abstract
O-GlcNAcylation is a post-translational modification of tau understood to lower the speed and yield of its aggregation, a pathological hallmark of Alzheimer's disease (AD). O-GlcNAcase (OGA) is the only enzyme that removes O-linked N-acetyl-d-glucosamine (O-GlcNAc) from target proteins. Therefore, inhibition of OGA represents a potential approach for the treatment of AD by preserving the O-GlcNAcylated tau protein. Herein, we report the multifactorial optimization of high-throughput screening hit 8 to a potent, metabolically stable, and orally bioavailable diazaspirononane OGA inhibitor (+)-56. The human OGA X-ray crystal structure has been recently solved, but bacterial hydrolases are still widely used as structural homologues. For the first time, we reveal how a nonsaccharide series of inhibitors binds bacterial OGA and discuss the suitability of two different bacterial orthologues as surrogates for human OGA. These breakthroughs enabled structure-activity relationships to be understood and provided context and boundaries for the optimization of druglike properties.
- Published
- 2020
- Full Text
- View/download PDF
17. Characterization of HTT inclusion size, location, and timing in the zQ175 mouse model of Huntington's disease: an in vivo high-content imaging study.
- Author
-
Nikisha Carty, Nadège Berson, Karsten Tillack, Christina Thiede, Diana Scholz, Karsten Kottig, Yalda Sedaghat, Christina Gabrysiak, George Yohrling, Heinz von der Kammer, Andreas Ebneth, Volker Mack, Ignacio Munoz-Sanjuan, and Seung Kwak
- Subjects
Medicine ,Science - Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by a CAG trinucleotide repeat expansion in the huntingtin gene. Major pathological hallmarks of HD include inclusions of mutant huntingtin (mHTT) protein, loss of neurons predominantly in the caudate nucleus, and atrophy of multiple brain regions. However, the early sequence of histological events that manifest in region- and cell-specific manner has not been well characterized. Here we use a high-content histological approach to precisely monitor changes in HTT expression and characterize deposition dynamics of mHTT protein inclusion bodies in the recently characterized zQ175 knock-in mouse line. We carried out an automated multi-parameter quantitative analysis of individual cortical and striatal cells in tissue slices from mice aged 2-12 months and confirmed biochemical reports of an age-associated increase in mHTT inclusions in this model. We also found distinct regional and subregional dynamics for inclusion number, size and distribution with subcellular resolution. We used viral-mediated suppression of total HTT in the striatum of zQ175 mice as an example of a therapeutically-relevant but heterogeneously transducing strategy to demonstrate successful application of this platform to quantitatively assess target engagement and outcome on a cellular basis.
- Published
- 2015
- Full Text
- View/download PDF
18. Ready-to-use adherent hiPSC-derived neural co-cultures by vitrification
- Author
-
Ina Meiser, Bianca Stephan, Julia Majer, Saskia Altmaier, Dieter Van De Sande, Mohamed Kreir, Alfredo Cabrera, Juan D. Pita Almenar, Julia C. Neubauer, Andreas Ebneth, and Heiko Zimmermann
- Subjects
General Medicine ,General Agricultural and Biological Sciences ,General Biochemistry, Genetics and Molecular Biology - Published
- 2022
- Full Text
- View/download PDF
19. Small Molecule Binding to Alzheimer Risk Factor CD33 Promotes Aβ Phagocytosis
- Author
-
Andreas Ebneth, Luke A. Miles, S.J. Hermans, Marie-Laure Rives, Daniel Oehlrich, Berthold Wroblowski, Jasmina Markulić, Michael W. Parker, Ines Royaux, Nancy C. Hancock, Gabriela A. N. Crespi, Andrés A. Trabanco, Jonathan H. Gooi, Larissa Doughty, and Tracy L. Nero
- Subjects
0301 basic medicine ,Peptide ,02 engineering and technology ,Molecular neuroscience ,Alzheimer's Disease ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,medicine ,Receptor ,lcsh:Science ,Components of the Immune System ,chemistry.chemical_classification ,Multidisciplinary ,Molecular Structure ,Microglia ,021001 nanoscience & nanotechnology ,Protein Structure Aspects ,3. Good health ,Sialic acid ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,lcsh:Q ,CD33 ,Molecular Neuroscience ,Small molecule binding ,Pharmacophore ,0210 nano-technology ,Glycoprotein ,Neuroscience - Abstract
Summary Polymorphism in the microglial receptor CD33 gene has been linked to late-onset Alzheimer disease (AD), and reduced expression of the CD33 sialic acid-binding domain confers protection. Thus, CD33 inhibition might be an effective therapy against disease progression. Progress toward discovery of selective CD33 inhibitors has been hampered by the absence of an atomic resolution structure. We report here the crystal structures of CD33 alone and bound to a subtype-selective sialic acid mimetic called P22 and use them to identify key binding residues by site-directed mutagenesis and binding assays to reveal the molecular basis for its selectivity toward sialylated glycoproteins and glycolipids. We show that P22, when presented on microparticles, increases uptake of the toxic AD peptide, amyloid-β (Aβ), into microglial cells. Thus, the sialic acid-binding site on CD33 is a promising pharmacophore for developing therapeutics that promote clearance of the Aβ peptide that is thought to cause AD., Graphical Abstract, Highlights • A sialic acid mimetic increases the uptake of Alzheimer peptide into microglia • Crystal structure of cell surface receptor CD33 bound to the sialic acid mimetic • Crystal structure of unliganded CD33 and mutagenesis studies revealed key residues • CD33 carbohydrate-binding site is a pharmacophore for Alzheimer drug development, Molecular Structure; Neuroscience; Molecular Neuroscience; Components of the Immune System; Protein Structure Aspects
- Published
- 2019
20. SOX9-induced Generation of Functional Astrocytes Supporting Neuronal Maturation in an All-human System
- Author
-
Alfredo Cabrera-Socorro, Pei-Yu Shih, Juan Diego Pita Almenar, Jonathan De Smedt, Johanna Van Daele, Devesh Kumar, Tim Vervliet, Katrien Neyrinck, Astrid D'hondt, Melissa Nijs, Catherine M. Verfaillie, Mohamed Kreir, Geert Bultynck, Keimpe D. Wierda, Mélanie Planque, Tom Vanbokhoven, Andreas Ebneth, Vania Broccoli, Frederik Seibt, and Sarah-Maria Fendt
- Subjects
Genome engineering ,Cell type ,medicine.medical_treatment ,Neurogenesis ,Induced Pluripotent Stem Cells ,SOX9 ,Biology ,Article ,All-human co-culture system ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Neural Stem Cells ,Pluripotent stem cells ,medicine ,Animals ,Humans ,Secretion ,Induced pluripotent stem cell ,030304 developmental biology ,Neurons ,0303 health sciences ,Growth factor ,SOX9 Transcription Factor ,General Medicine ,Differentiation protocol ,Neural stem cell ,Cytokine ,Astrocytes ,Stem cell ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Astrocytes, the main supportive cell type of the brain, show functional impairments upon ageing and in a broad spectrum of neurological disorders. Limited access to human astroglia for pre-clinical studies has been a major bottleneck delaying our understanding of their role in brain health and disease. We demonstrate here that functionally mature human astrocytes can be generated by SOX9 overexpression for 6 days in pluripotent stem cell (PSC)-derived neural progenitor cells. Inducible (i)SOX9-astrocytes display functional properties comparable to primary human astrocytes comprising glutamate uptake, induced calcium responses and cytokine/growth factor secretion. Importantly, electrophysiological properties of iNGN2-neurons co-cultured with iSOX9-astrocytes are indistinguishable from gold-standard murine primary cultures. The high yield, fast timing and the possibility to cryopreserve iSOX9-astrocytes without losing functional properties makes them suitable for scaled-up production for high-throughput analyses. Our findings represent a step forward to an all-human iPSC-derived neural model for drug development in neuroscience and towards the reduction of animal use in biomedical research. Graphical Abstract
- Published
- 2021
21. Generation of a set of isogenic iPSC lines carrying all APOE genetic variants (Ɛ2/Ɛ3/Ɛ4) and knock-out for the study of APOE biology in health and disease
- Author
-
Christian Clausen, Alfredo Ramirez, Andreas Ebneth, Pamela V. Martino-Adami, Alfredo Cabrera-Socorro, Eva Díaz-Guerra, Oliver Brüstle, Michaela Thoenes, Benjamin Schmid, Peter Reinhardt, Clara Grezella, Lamiaa Bahnassawy, Carlos Vicario, Bjørn Holst, Margot H.M. Bakker, Michael Peitz, Klaus Fliessbach, Innovative Medicines Initiative, and European Commission
- Subjects
0301 basic medicine ,Apolipoprotein E ,Apolipoprotein B ,Genotype ,Induced Pluripotent Stem Cells ,genetics [Alzheimer Disease] ,Disease ,Homology (biology) ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Apolipoproteins E ,Alzheimer Disease ,ddc:570 ,Animals ,Induced pluripotent stem cell ,Gene ,lcsh:QH301-705.5 ,Biology ,Genetics ,biology ,Cell Biology ,General Medicine ,In vitro ,030104 developmental biology ,lcsh:Biology (General) ,biology.protein ,genetics [Apolipoproteins E] ,lipids (amino acids, peptides, and proteins) ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
APOE genotype is the strongest genetic risk factor for Alzheimer’s Disease (AD). The low degree of homology between mouse and human APOE is a concerning issue in preclinical models currently used to study the role of this gene in AD pathophysiology. A key objective of ADAPTED (Alzheimer’s Disease Apolipoprotein Pathology for Treatment Elucidation and Development) project was to generate in vitro models that better recapitulate human APOE biology. We describe a new set of induced pluripotent stem cells (iPSC) lines carrying common APOE variants (Ɛ2, Ɛ3, and Ɛ3/Ɛ4) and a knock-out isogenic to the parental APOE Ɛ4/Ɛ4 line (UKBi011-A)., This study was funded by the ADAPTED (Alzheimer’s Disease Apolipoprotein Pathology for Treatment Elucidation and Development) consortium which has received funding from the Innovative Medicines Initiative 2 Joint Undertaking under Grant Agreement No 115975. This Joint Undertaking receives support from the European Union’s Horizon 2020 research and innovation programme and the European Federation of Pharmaceutical Industries and Associations.
- Published
- 2021
- Full Text
- View/download PDF
22. Generation of two gene edited iPSC-lines carrying a DOX-inducible NGN2 expression cassette with and without GFP in the AAVS1 locus
- Author
-
Alfredo Cabrera-Socorro, Benjamin Schmid, Ida Jørring, Andreas Ebneth, Bjørn Holst, Ulrike A. Mau-Holzmann, Ulla B. Poulsen, Rachel Steeg, Mikkel A. Rasmussen, Christian Clausen, Hugh Nuthall, and Publica
- Subjects
0301 basic medicine ,Gene Editing ,Transgene ,Cellular differentiation ,Induced Pluripotent Stem Cells ,Cell Differentiation ,Cell Biology ,General Medicine ,Biology ,Green fluorescent protein ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,lcsh:Biology (General) ,Genes, Reporter ,Expression cassette ,Transgenes ,Stem cell ,Induced pluripotent stem cell ,Transcription factor ,Gene ,lcsh:QH301-705.5 ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Neurog2 is the gene encoding the neuronal transcription factor NGN2, which can convert stem cells into functional neurons in a fast and efficient way. Here we report the generation of two iPS cell lines, where DOX inducible constructs of neurog2 either without or with T2A-eGFP were inserted into the safe-site locus AAVS1. These iPS cell lines, BIONi010-C-13 and BIONi010-C-15, respectively, stay pluripotent without DOX but differentiate to (GFP positive) neurons when DOX is added without the need of differentiation factors.
- Published
- 2020
23. Imaging Mutant Huntingtin Aggregates: Development of a Potential PET Ligand
- Author
-
Michael Prime, Samantha Green, Celia Dominguez, Edith Monteagudo, Malcolm Taylor, Akihiro Takano, Longbin Liu, Adrian Kotey, Wayne Thomas, Zhisheng Jia, Anthony P Dickie, Catherine Greenaway, Samuel Coe, Miklós Tóth, John Wityak, Vinod Khetarpal, Sergio Menta, Simone Esposito, Katarina Vanräs, Andreas Ebneth, John E. Mangette, Ian Wigginton, Todd Herbst, Peter Johnson, Sabine Schaertl, Vladimir Stepanov, Jonathan Bard, Sebastien Galan, Elise Gadouleau, Randall Davis, Christopher John Brown, Frank Herrmann, Richard W Marston, Darshan Gunvant Vaidya, Laura Orsatti, Xuemei Chen, Martina Nibbio, Manuela Heßmann, Joanne Sproston, Matthew R Mills, Ignacio Munoz-Sanjuan, Daniel Clark-Frew, Derek Alexander Weddell, Ladislav Mrzljak, Christoph Scheich, Xinjie Gai, Christer Halldin, Sangram Nag, Lee Matthew, Patricia Miranda-Azpiazu, Paul Giles, Thomas Krulle, Alexander Kiselyov, Marie Svedberg, and Sarah Hayes
- Subjects
Male ,Huntingtin ,Imaging biomarker ,Mutant ,Protein aggregation ,medicine.disease_cause ,Ligands ,01 natural sciences ,Protein Aggregation, Pathological ,Madin Darby Canine Kidney Cells ,Rats, Sprague-Dawley ,03 medical and health sciences ,Mice ,Dogs ,Drug Discovery ,medicine ,Animals ,Humans ,030304 developmental biology ,0303 health sciences ,Mutation ,Huntingtin Protein ,medicine.diagnostic_test ,Chemistry ,Ligand (biochemistry) ,0104 chemical sciences ,Cell biology ,Mice, Inbred C57BL ,010404 medicinal & biomolecular chemistry ,Disease Models, Animal ,Huntington Disease ,Positron emission tomography ,Positron-Emission Tomography ,Molecular Medicine ,Female ,Efflux ,Radiopharmaceuticals ,Peptides - Abstract
Mutant huntingtin (mHTT) protein carrying the elongated N-terminal polyglutamine (polyQ) tract misfolds and forms protein aggregates characteristic of Huntington's disease (HD) pathology. A high-affinity ligand specific for mHTT aggregates could serve as a positron emission tomography (PET) imaging biomarker for HD therapeutic development and disease progression. To identify such compounds with binding affinity for polyQ aggregates, we embarked on systematic structural activity studies; lead optimization of aggregate-binding affinity, unbound fractions in brain, permeability, and low efflux culminated in the discovery of compound 1, which exhibited target engagement in autoradiography (ARG) studies in brain slices from HD mouse models and postmortem human HD samples. PET imaging studies with 11C-labeled 1 in both HD mice and WT nonhuman primates (NHPs) demonstrated that the right-hand-side labeled ligand [11C]-1R (CHDI-180R) is a suitable PET tracer for imaging of mHTT aggregates. [11C]-1R is now being advanced to human trials as a first-in-class HD PET radiotracer.
- Published
- 2020
24. Generation of a human induced pluripotent stem cell–based model for tauopathies combining three microtubule‐associated protein TAU mutations which displays several phenotypes linked to neurodegeneration
- Author
-
Joke Terryn, Keimpe D. Wierda, Bart De Strooper, Alfredo Cabrera-Socorro, Frederic Lluis, Francisco Pestana, Andreas Ebneth, Laura Ordovás, Juan Antonio García-León, Ana Quiles, Catherine M. Verfaillie, Ann Swijsen, Fatemeharefeh Nami, Lutgarde Serneels, Kristel Eggermont, Raheem Fazal, Mohamed Kreir, Lieven Thorrez, Annerieke Sierksma, and Philip Van Damme
- Subjects
0301 basic medicine ,Epidemiology ,Membrane Potentials ,CRISPR ,CRISPR-Cas ,Induced pluripotent stem cell ,PIGGYBAC TRANSPOSON ,Neurons ,Health Policy ,MISSENSE ,Neurodegeneration ,CORTICAL-NEURONS ,MOUSE MODEL ,Alzheimer's disease ,Phenotype ,NEOCORTEX ,Cell biology ,ALZHEIMERS-DISEASE ,Psychiatry and Mental health ,Disease modeling ,Drug screening ,Tauopathies ,Tauopathy ,Life Sciences & Biomedicine ,Frontotemporal dementia ,Neurite ,Neurogenesis ,Induced Pluripotent Stem Cells ,Neuronal Outgrowth ,Clinical Neurology ,INHIBITION ,Parkinsonism linked to chromosome 17 ,tau Proteins ,Biology ,Cell Line ,Progressive supranuclear palsy ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Downregulation and upregulation ,medicine ,Humans ,Science & Technology ,medicine.disease ,PATHOLOGY ,030104 developmental biology ,Mutation ,Nerve Degeneration ,HIPPOCAMPUS ,Neurosciences & Neurology ,Neurology (clinical) ,CRISPR-Cas Systems ,Geriatrics and Gerontology ,Transcriptome - Abstract
INTRODUCTION: Tauopathies are neurodegenerative diseases characterized by TAU protein-related pathology, including frontotemporal dementia and Alzheimer's disease among others. Mutant TAU animal models are available, but none of them faithfully recapitulates human pathology and are not suitable for drug screening. METHODS: To create a new in vitro tauopathy model, we generated a footprint-free triple MAPT-mutant human induced pluripotent stem cell line (N279K, P301L, and E10+16 mutations) using clustered regularly interspaced short palindromic repeats-FokI and piggyBac transposase technology. RESULTS: Mutant neurons expressed pathogenic 4R and phosphorylated TAU, endogenously triggered TAU aggregation, and had increased electrophysiological activity. TAU-mutant cells presented deficiencies in neurite outgrowth, aberrant sequence of differentiation to cortical neurons, and a significant activation of stress response pathways. RNA sequencing confirmed stress activation, demonstrated a shift toward GABAergic identity, and an upregulation of neurodegenerative pathways. DISCUSSION: In summary, we generated a novel in vitro human induced pluripotent stem cell TAU-mutant model displaying neurodegenerative disease phenotypes that could be used for disease modeling and drug screening. ispartof: ALZHEIMERS & DEMENTIA vol:14 issue:10 pages:1261-1280 ispartof: location:United States status: published
- Published
- 2018
- Full Text
- View/download PDF
25. Development of a fully human assay combining NGN2-inducible neurons co-cultured with iPSC-derived astrocytes amenable for electrophysiological studies
- Author
-
Benjamin Schmid, Pei-Yu Shih, Devesh Kumar, Alfredo Cabrera-Socorro, Christian Clausen, Andreas Ebneth, Francisco Pestana, Rachel Steeg, Bjørn Holst, Frederik Seibt, Juan D. Pita-Almenar, Mohamed Kreir, Benjamin Fischer, and Publica
- Subjects
QH301-705.5 ,Somatic cell ,Cellular differentiation ,Induced Pluripotent Stem Cells ,Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell & Tissue Engineering ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Humans ,Neurogenin-2 ,Biology (General) ,Progenitor cell ,Induced pluripotent stem cell ,Cells, Cultured ,030304 developmental biology ,Neurons ,0303 health sciences ,Science & Technology ,Cell Differentiation ,Cell Biology ,General Medicine ,Phenotype ,Embryonic stem cell ,Coculture Techniques ,Cell biology ,SYNAPTIC-TRANSMISSION ,DIFFERENTIATION ,Biotechnology & Applied Microbiology ,Astrocytes ,EXCITATORY NEURONS ,Stem cell ,Life Sciences & Biomedicine ,PLURIPOTENT STEM-CELLS ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Neurogenin 2 encodes a neural-specific transcription factor (NGN2) able to drive neuronal fate on somatic and stem cells. NGN2 is expressed in neural progenitors within the developing central and peripheral nervous systems. Overexpression of NGN2 in human induced pluripotent stem cells (hiPSCs) or human embryonic stem cells has been shown to efficiently trigger conversion to neurons. Here we describe two gene-edited hiPSC lines harbouring a doxycycline (DOX)-inducible cassette in the AAVS1 locus driving expression of NGN2 (BIONi010-C-13) or NGN2-T2A-GFP (BIONi010-C-15). By introducing NGN2-expressing cassette, we reduce variability associated with conventional over-expression methods such as viral transduction, making these lines amenable for scale-up production and screening processes. DOX-treated hiPSCs convert to neural phenotype within one week and display the expression of structural neuronal markers such as Beta-III tubulin and tau. We performed functional characterization of NGN2-neurons co-cultured with hiPSC-derived astrocytes in a "fully-humanized" set up. Passive properties of NGN2-neurons were indistinguishable from mouse primary cells while displaying variable activity in extracellular recordings performed in multi-electrode arrays (MEAs). We demonstrate that hiPSC-derived astrocytes and neurons can be co-cultured and display functional properties comparable to the gold standard used in electrophysiology. Both lines are globally available via EBiSC repository at https://cells.ebisc.org/. ispartof: STEM CELL RESEARCH vol:54 ispartof: location:England status: published
- Published
- 2021
- Full Text
- View/download PDF
26. Systemic immune-checkpoint blockade with anti-PD1 antibodies does not alter cerebral amyloid-β burden in several amyloid transgenic mouse models
- Author
-
Andreas Ebneth, Astrid Bottelbergs, Cindy Wintmolders, Philippe Goniot, Nathalie Schussler, Emmanuel Brault, Gary J. Nabel, David R. Riddell, Yaming Wang, Alexis Bretteville, Margaret M. Racke, Véronique Blanc, Bart Roucourt, Zhi Yong Yang, Laurent Pradier, Tyler McGathey, Bradford Elmer, Jirong Lu, Michael J. O'Neill, Martine Latta-Mahieu, Liesbeth Mertens, Caroline Hersley, Paul Ferrari, Mati Lopez-Grancha, Pascal Barneoud, Nicolas Moindrot, Philippe Bertrand, Véronique Blanchard, Valerie Roudieres, Ling Liu, Sofie Carmans, and Peter Larsen
- Subjects
Male ,0301 basic medicine ,Genetically modified mouse ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Mice, Transgenic ,Inflammation ,Antibodies ,Immune tolerance ,Pathogenesis ,Interferon-gamma ,Random Allocation ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Immune system ,Alzheimer Disease ,Presenilin-1 ,medicine ,Animals ,Humans ,RNA, Messenger ,Amyloid beta-Peptides ,biology ,Brain ,Immunotherapy ,Immunohistochemistry ,Immune checkpoint ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Neurology ,Immunology ,biology.protein ,Female ,medicine.symptom ,Antibody ,Spleen ,030217 neurology & neurosurgery - Abstract
Chronic inflammation represents a central component in the pathogenesis of Alzheimer's disease (AD). Recent work suggests that breaking immune tolerance by Programmed cell Death-1 (PD1) checkpoint inhibition produces an IFN-γ-dependent systemic immune response, with infiltration of the brain by peripheral myeloid cells and neuropathological as well as functional improvements even in mice with advanced amyloid pathology (Baruch et al., (2016): Nature Medicine, 22:135–137). Immune checkpoint inhibition was therefore suggested as potential treatment for neurodegenerative disorders when activation of the immune system is appropriate. Because a xenogeneic rat antibody (mAb) was used in the study, whether the effect was specific to PD1 target engagement was uncertain. In the present study we examined whether PD1 immunotherapy can lower amyloid-β pathology in a range of different amyloid transgenic models performed at three pharmaceutical companies with the exact same anti-PD1 isotype and two mouse chimeric variants. Although PD1 immunotherapy stimulated systemic activation of the peripheral immune system, monocyte-derived macrophage infiltration into the brain was not detected, and progression of brain amyloid pathology was not altered. Similar negative results of the effect of PD1 immunotherapy on amyloid brain pathology were obtained in two additional models in two separate institutions. These results show that inhibition of PD1 checkpoint signaling by itself is not sufficient to reduce amyloid pathology and that additional factors might have contributed to previously published results (Baruch et al., (2016): Nature Medicine, 22:135–137). Until such factors are elucidated, animal model data do not support further evaluation of PD1 checkpoint inhibition as a therapeutic modality for Alzheimer's disease.
- Published
- 2017
- Full Text
- View/download PDF
27. Rapid establishment of the European Bank for induced Pluripotent Stem Cells (EBiSC) - the Hot Start experience
- Author
-
Oliver Brüstle, Annette Ringwald, Christian Clausen, Charlotte Chapman, Glyn Stacey, Alexander J. Kvist, Marieke A. Hoeve, Shailesh Kumar Gupta, Blanca Miranda Serano, Minal Patel, Gabriella Brolén, Tomo Saric, George McConnachie, Cornelia Thiele, Julie Holder, Trisha Rawat, Tina C. Stummann, Joh Dokler, Maja Brajnik, Bryan Bolton, Lyle Armstrong, Bernd Kuebler, Alfredo Cabrera-Socorro, Isobel Atkin, Adam Faulconbridge, Christa Lucas, Jason A. King, Johannes Dewender, Ian Streeter, Rok Predan, Majlinda Lako, Lyn Healy, Bjørn Holst, Martine Geraerts, Luca Cherubin, Carsten Claussen, Heiko Zimmermann, Timothy E Allsopp, Orla O'Shea, Peter W. Harrison, K. Bruce, Niels Geijsen, Michael Peitz, Tony Burdett, Paul A. De Sousa, Ole Pless, Elisabeth Wachter, Anna Veiga, A. Courtney, Laura Clarke, Helen Parkinson, Cesar Trigueros, Anna Jonebring, Shalinee Khadun, Rachel Steeg, Patrik Foerch, Sascha Reimann, Ryan Hicks, Carol George, Andreas Ebneth, Oliver Keminer, Philip Gribbon, Barry Hardy, Stefanie Seltmann, Andreas Kurtz, Shawn Harmon, Jürgen Hescheler, Beate Kreisel, Hubrecht Institute for Developmental Biology and Stem Cell Research, and Publica
- Subjects
0301 basic medicine ,QH301-705.5 ,Process (engineering) ,media_common.quotation_subject ,Induced Pluripotent Stem Cells ,Biology ,Tissue procurement ,Cell Line ,03 medical and health sciences ,Resource (project management) ,Journal Article ,Humans ,Quality (business) ,Biology (General) ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Biological Specimen Banks ,media_common ,Cryopreservation ,International research ,Medicine(all) ,Hot start ,General Medicine ,Cell Biology ,Europe ,Engineering management ,030104 developmental biology ,lcsh:Biology (General) ,Work flow ,Developmental Biology - Abstract
A fast track “Hot Start” process was implemented to launch the European Bank for Induced Pluripotent Stem Cells (EBiSC) to provide early release of a range of established control and disease linked human induced pluripotent stem cell (hiPSC) lines. Established practice amongst consortium members was surveyed to arrive at harmonised and publically accessible Standard Operations Procedures (SOPs) for tissue procurement, bio-sample tracking, iPSC expansion, cryopreservation, qualification and distribution to the research community. These were implemented to create a quality managed foundational collection of lines and associated data made available for distribution. Here we report on the successful outcome of this experience and work flow for banking and facilitating access to an otherwise disparate European resource, with lessons to benefit the international research community.eTOCThe report focuses on the EBiSC experience of rapidly establishing an operational capacity to procure, bank and distribute a foundational collection of established hiPSC lines. It validates the feasibility and defines the challenges of harnessing and integrating the capability and productivity of centres across Europe using commonly available resources currently in the field.
- Published
- 2017
- Full Text
- View/download PDF
28. Development of a Scalable, High-Throughput-Compatible Assay to Detect Tau Aggregates Using iPSC-Derived Cortical Neurons Maintained in a Three-Dimensional Culture Format
- Author
-
Liesbeth Mertens, Alfredo Cabrera-Socorro, Alexis Bretteville, Ines Royaux, X. Medda, Sofie Versweyveld, L. Barnham, An Verheyen, Arjan Buist, Andreas Ebneth, and Annick Diels
- Subjects
0301 basic medicine ,Induced Pluripotent Stem Cells ,Cell ,Cell Culture Techniques ,Drug Evaluation, Preclinical ,tau Proteins ,Nanotechnology ,Protein aggregation ,Biology ,Biochemistry ,Analytical Chemistry ,Protein Aggregates ,03 medical and health sciences ,0302 clinical medicine ,High-Throughput Screening Assays ,medicine ,Humans ,Phosphorylation ,Cognitive decline ,Induced pluripotent stem cell ,Neurons ,Matrigel ,Neurodegeneration ,Brain ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Tauopathies ,Cell culture ,Molecular Medicine ,Neuroscience ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Tau aggregation is the pathological hallmark that best correlates with the progression of Alzheimer's disease (AD). The presence of neurofibrillary tangles (NFTs), formed of hyperphosphorylated tau, leads to neuronal dysfunction and loss, and is directly associated with the cognitive decline observed in AD patients. The limited success in targeting β-amyloid pathologies has reinforced the hypothesis of blocking tau phosphorylation, aggregation, and/or spreading as alternative therapeutic entry points to treat AD. Identification of novel therapies requires disease-relevant and scalable assays capable of reproducing key features of the pathology in an in vitro setting. Here we use induced pluripotent stem cells (iPSCs) as a virtually unlimited source of human cortical neurons to develop a robust and scalable tau aggregation model compatible with high-throughput screening (HTS). We downscaled cell culture conditions to 384-well plate format and used Matrigel to introduce an extra physical protection against cell detachment that reduces shearing stress and better recapitulates pathological conditions. We complemented the assay with AlphaLISA technology for the detection of tau aggregates in a high-throughput-compatible format. The assay is reproducible across users and works with different commercially available iPSC lines, representing a highly translational tool for the identification of novel treatments against tauopathies, including AD.
- Published
- 2016
- Full Text
- View/download PDF
29. Genetically Engineered iPSC-Derived FTDP-17 MAPT Neurons Display Mutation-Specific Neurodegenerative and Neurodevelopmental Phenotypes
- Author
-
P. Roevens, Constantin d’Ydewalle, Andreas Ebneth, Jacobine Kuijlaars, Selina Wray, Annick Diels, Alexis Bretteville, Arjan Buist, An Verheyen, Joke Reumers, Alfredo Cabrera-Socorro, Ronald de Hoogt, Louis De Muynck, Ines Royaux, Kirsten Van Hoorde, An De Bondt, Ilse Van den Wyngaert, Pieter J. Peeters, and Steffen Jaensch
- Subjects
0301 basic medicine ,Tau protein ,medicine.disease_cause ,Biochemistry ,Article ,FTDP-17 ,03 medical and health sciences ,0302 clinical medicine ,mental disorders ,medicine ,Genetics ,MAPT ,Induced pluripotent stem cell ,ZFN ,lcsh:QH301-705.5 ,030304 developmental biology ,lcsh:R5-920 ,Mutation ,0303 health sciences ,iPSC ,biology ,Neurodegeneration ,Wnt signaling pathway ,Correction ,Cell Biology ,medicine.disease ,Zinc finger nuclease ,Phenotype ,Cell biology ,disease modelling ,030104 developmental biology ,lcsh:Biology (General) ,biology.protein ,lcsh:Medicine (General) ,030217 neurology & neurosurgery ,Frontotemporal dementia ,Developmental Biology - Abstract
Summary Tauopathies such as frontotemporal dementia (FTD) remain incurable to date, partially due to the lack of translational in vitro disease models. The MAPT gene, encoding the microtubule-associated protein tau, has been shown to play an important role in FTD pathogenesis. Therefore, we used zinc finger nucleases to introduce two MAPT mutations into healthy donor induced pluripotent stem cells (iPSCs). The IVS10+16 mutation increases the expression of 4R tau, while the P301S mutation is pro-aggregant. Whole-transcriptome analysis of MAPT IVS10+16 neurons reveals neuronal subtype differences, reduced neural progenitor proliferation potential, and aberrant WNT/SHH signaling. Notably, these neurodevelopmental phenotypes could be recapitulated in neurons from patients carrying the MAPT IVS10+16 mutation. Moreover, the additional pro-aggregant P301S mutation revealed additional phenotypes, such as an increased calcium burst frequency, reduced lysosomal acidity, tau oligomerization, and neurodegeneration. This series of iPSCs could serve as a platform to unravel a potential link between pathogenic 4R tau and FTD., Highlights • Analysis of ZFN-engineered MAPT IVS10+16 with or without additional P301S mutation • Neurodevelopmental phenotypes in ZFN and patient-derived MAPT IVS10+16 neurons • Neurodegenerative phenotypes in MAPT IVS10+16/P301S double-mutant neurons, In this article, Verheyen and colleagues introduced 2 MAPT mutations (IVS10+16/P301S) into healthy donor iPSCs. Characterization of MAPT IVS10+16 neurons revealed neurodevelopmental neuron subtype differences, reduced neural progenitor proliferation, and aberrant WNT/SHH signaling, while the additional pro-aggregant P301S mutation revealed phenotypes more related to neurodegeneration. These iPSCs could help to unravel a potential link between pathogenic 4R tau and FTD.
- Published
- 2017
30. [P3–080]: AN ACADEMIC‐PRIVATE PARTNERSHIP FOR THE VALIDATION OF NEW MODELS TO UNDERSTAND TAU‐RELATED HYPEREXCITABILITY AND AGGREGATION USING HUMAN‐INDUCED PLURIPOTENT STEM CELLS
- Author
-
Ana Quiles, Hugh Nuthall, Pei-Yu Shih, Catherine M. Verfaillie, An Verheyen, Kristel Eggermont, Francisco Pestana, Ines Royaux, Joke Terryn, Kevin Bruce, Raheem Fazal, Patrik Foerch, Juan Diego Pita Almenar, Martine Geraerts, Alfredo Cabrera Socorro, Juan Antonio Garcia Leon, Peter J. Craig, Laura Ordovás, Mohamed Kreir, Andreas Ebneth, and Tina Charlotte Stumman
- Subjects
Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Developmental Neuroscience ,Epidemiology ,Health Policy ,General partnership ,Neurology (clinical) ,Geriatrics and Gerontology ,Human Induced Pluripotent Stem Cells ,Biology ,Neuroscience - Published
- 2017
- Full Text
- View/download PDF
31. [P4–047]: ANTI‐TAU ANTIBODY PROFILING FOR PASSIVE IMMUNIZATION THERAPY: COMPARING SEEDS DERIVED FROM TRANSGENIC ANIMALS AND HUMAN BRAIN TISSUE
- Author
-
Kristof Van Kolen, Inez Van de Weyer, Greet Vanhoof, Sofie Versweyveld, Katja De Waepenaert, Koen Dockx, Marc Mercken, Marc Vandermeeren, M. Borgers, and Andreas Ebneth
- Subjects
0301 basic medicine ,Epidemiology ,Health Policy ,Transgene ,Human brain ,Biology ,03 medical and health sciences ,Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Developmental Neuroscience ,Immunology ,medicine ,biology.protein ,Neurology (clinical) ,Geriatrics and Gerontology ,Antibody ,030217 neurology & neurosurgery - Published
- 2017
- Full Text
- View/download PDF
32. Generation of a set of isogenic, gene-edited iPSC lines homozygous for all main APOE variants and an APOE knock-out line
- Author
-
Blanca Irene Aldana Garcia, Mikkel A. Rasmussen, Kennie R. Prehn, K. Bruce, Alfredo Cabrera-Socorro, Ulla B. Poulsen, Benjamin Schmid, Rachel Steeg, Peter Mackintosh, Bjørn Holst, Sarayu Ramakrishna, Ida Jørring, Ulrike A. Mau-Holzmann, Natakarn Nimsanor, Christian Clausen, Andreas Ebneth, and Ravi S. Muddashetty
- Subjects
Male ,0301 basic medicine ,Apolipoprotein E ,Adolescent ,Induced Pluripotent Stem Cells ,Cell Culture Techniques ,Single-nucleotide polymorphism ,Biology ,Cell Line ,Gene Knockout Techniques ,03 medical and health sciences ,Apolipoproteins E ,0302 clinical medicine ,Genotype ,Humans ,CRISPR ,Allele ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Gene ,Gene Editing ,Genetics ,Homozygote ,Cell Biology ,General Medicine ,3. Good health ,030104 developmental biology ,lcsh:Biology (General) ,Mutation ,lipids (amino acids, peptides, and proteins) ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Alzheimer's disease (AD) is the most frequent neurodegenerative disease amongst the elderly. The SNPs rs429358 and rs7412 in the APOE gene are the most common risk factor for sporadic AD, and there are three different alleles commonly referred to as APOE-epsilon 2, APOE-epsilon 3 and APOE-epsilon 4. Induced pluripotent stem cells (iPSCs) hold great promise to model AD as such cells can be differentiated in vitro to the required cell type. Here we report the use of CRISPR/Cas9 technology employed on iPSCs from a healthy individual with an APOE-epsilon 3/epsilon 4 genotype to obtain isogenic APOE-epsilon 2/epsilon 2, APOE-epsilon 3/epsilon 3, APOE-epsilon 4/epsilon 4 lines as well as an APOE-knock-out line.
- Published
- 2019
- Full Text
- View/download PDF
33. Discovery and Structure–Activity Relationship of Potent and Selective Covalent Inhibitors of Transglutaminase 2 for Huntington’s Disease
- Author
-
Robert K. Y. Cheng, Celia Dominguez, Osamu Ichihara, Douglas Macdonald, Ignacio Munoz-Sanjuan, Mark A. Brooks, Andreas Ebneth, Leticia Toledo-Sherman, Michael Prime, Ian Toogood-Johnson, Sayeh Erfan, John Wityak, Richard W Marston, Frederick Arthur Brookfield, Darshan Gunvant Vaidya, Stephen Martin Courtney, Peter Johnson, Anna Pedret-Dunn, Andreas Scheel, Dirk Winkler, Christopher John Yarnold, Sabine Schaertl, Ole A. Andersen, Shilpa Palan, Maria Beconi, Brunella Felicetti, Siw F. Johnsen, John J. Barker, Jordan J. Palfrey, and Ina Sternberger
- Subjects
Male ,Models, Molecular ,Cell Membrane Permeability ,Pyridines ,Tissue transglutaminase ,Lysine ,In Vitro Techniques ,Piperazines ,Mice ,Structure-Activity Relationship ,Huntington's disease ,GTP-Binding Proteins ,Drug Discovery ,medicine ,Animals ,Humans ,Structure–activity relationship ,Protein Glutamine gamma Glutamyltransferase 2 ,chemistry.chemical_classification ,Acrylamides ,Sulfonamides ,Isopeptide bond ,Transglutaminases ,biology ,medicine.disease ,Molecular biology ,In vitro ,Rats ,Glutamine ,HEK293 Cells ,Huntington Disease ,Pyrimidines ,Enzyme ,Biochemistry ,chemistry ,Microsomes, Liver ,biology.protein ,Molecular Medicine ,Caco-2 Cells - Abstract
Tissue transglutaminase 2 (TG2) is a multifunctional protein primarily known for its calcium-dependent enzymatic protein cross-linking activity via isopeptide bond formation between glutamine and lysine residues. TG2 overexpression and activity have been found to be associated with Huntington's disease (HD); specifically, TG2 is up-regulated in the brains of HD patients and in animal models of the disease. Interestingly, genetic deletion of TG2 in two different HD mouse models, R6/1 and R6/2, results in improved phenotypes including a reduction in neuronal death and prolonged survival. Starting with phenylacrylamide screening hit 7d, we describe the SAR of this series leading to potent and selective TG2 inhibitors. The suitability of the compounds as in vitro tools to elucidate the biology of TG2 was demonstrated through mode of inhibition studies, characterization of druglike properties, and inhibition profiles in a cell lysate assay.
- Published
- 2012
- Full Text
- View/download PDF
34. Using Electrophysiology and In Silico Three-Dimensional Modeling to Reduce Human Ether-à-go-go Related Gene K+ Channel Inhibition in a Histamine H3 Receptor Antagonist Program
- Author
-
Alexander Heifetz, Mark J. Gemkow, Michael P. Mazanetz, Richard J. Law, Clemens Möller, Andreas Ebneth, and Adam James Davenport
- Subjects
Models, Molecular ,Patch-Clamp Techniques ,In silico ,hERG ,Biology ,Pharmacology ,Structure-Activity Relationship ,chemistry.chemical_compound ,Cricetulus ,Cricetinae ,Drug Discovery ,medicine ,Animals ,Humans ,Receptors, Histamine H3 ,Computer Simulation ,Channel blocker ,Antagonist ,medicine.disease ,Ether-A-Go-Go Potassium Channels ,High-Throughput Screening Assays ,chemistry ,Drug Design ,biology.protein ,Molecular Medicine ,Histamine H3 receptor ,Pharmacophore ,Histamine ,Histamine H3 Antagonists ,Narcolepsy - Abstract
The histamine H3 receptor (H3R) plays a regulatory role in the presynaptic release of histamine and several other neurotransmitters, and thus, it is an attractive target for central nervous system indications including cognitive disorders, narcolepsy, attention-deficit hyperactivity disorder, and pain. The development of H3R antagonists was complicated by the similarities between the pharmacophores of H3R and human Ether-a-go-go related gene (hERG) channel blockers, a fact that probably prevented promising compounds from being progressed into the clinic. Using a three-dimensional in silico modeling approach complemented with automated and manual patch clamping, we were able to separate these two pharmacophores and to develop highly potent H3R antagonists with reduced risk of hERG liabilities from initial hit series with low selectivity identified in a high-throughput screening campaign.
- Published
- 2010
- Full Text
- View/download PDF
35. The Structure of Mammalian Serine Racemase
- Author
-
Brunella Felicetti, Michael W. Wood, O.C. Mather, John M. Barker, Cesura Andrea, Myron A. Smith, Volker Mack, Dorian Leo Schönfeld, Isabel Moraes, and Andreas Ebneth
- Subjects
Drug design ,Cell Biology ,Isomerase ,Biology ,Biochemistry ,Enzyme structure ,Serine ,chemistry.chemical_compound ,Protein structure ,chemistry ,Serine racemase ,Pyridoxal phosphate ,Molecular Biology ,Pyridoxal - Abstract
Serine racemase is responsible for the synthesis of D-serine, an endogenous co-agonist for N-methyl-D-aspartate receptor-type glutamate receptors (NMDARs). This pyridoxal 5'-phosphate-dependent enzyme is involved both in the reversible conversion of L- to D-serine and serine catabolism by alpha,beta-elimination of water, thereby regulating D-serine levels. Because D-serine affects NMDAR signaling throughout the brain, serine racemase is a promising target for the treatment of disorders related to NMDAR dysfunction. To provide a molecular basis for rational drug design the x-ray crystal structures of human and rat serine racemase were determined at 1.5- and 2.1-A resolution, respectively, and in the presence and absence of the orthosteric inhibitor malonate. The structures revealed a fold typical of beta-family pyridoxal 5'-phosphate enzymes, with both a large domain and a flexible small domain associated into a symmetric dimer, and indicated a ligand-induced rearrangement of the small domain that organizes the active site for specific turnover of the substrate.
- Published
- 2010
- Full Text
- View/download PDF
36. Morpholine containing CB2 selective agonists
- Author
-
Angela Berry, David S. Thomson, Doris Riether, Sabine Löbbe, Todd Bosanac, Andreas Ebneth, Diane Thome, Ernest L. Raymond, Daw-Tsun Shih, Renee Zindell, and Mark J. Gemkow
- Subjects
Agonist ,Cannabinoid receptor ,medicine.drug_class ,Chemistry, Pharmaceutical ,Morpholines ,Clinical Biochemistry ,Anti-Inflammatory Agents ,Pharmaceutical Science ,Biochemistry ,Anti-inflammatory ,Cell Line ,Receptor, Cannabinoid, CB2 ,Mice ,chemistry.chemical_compound ,In vivo ,Oral administration ,Morpholine ,Drug Discovery ,medicine ,Animals ,Humans ,Molecular Biology ,Inflammation ,Analgesics ,Molecular Structure ,Chemistry ,Organic Chemistry ,Stereoisomerism ,In vitro ,Models, Chemical ,Drug Design ,Prednisolone ,Molecular Medicine ,lipids (amino acids, peptides, and proteins) ,medicine.drug - Abstract
Identification and optimization of two classes of CB2 selective agonists are described. A representative from each class is profiled in a murine model of inflammation and each shows similar efficacy to prednisolone upon oral dosing.
- Published
- 2009
- Full Text
- View/download PDF
37. The novel KMO inhibitor CHDI-340246 leads to a restoration of electrophysiological alterations in mouse models of Huntington's disease
- Author
-
Amyaouch Bradaia, Heike Deisemann, Larry Park, Simon Gelman, Esther Steidl, Melanie Gleyzes, Ulrike Dijkman, Celia Dominguez, Leticia Toledo-Sherman, Vinod Khetarpal, Outi Kontkanen, Dirk Winkler, Ioana Neagoe, Robert Freije, Mariette Heins, Taneli Heikkinen, Jukka Puoliväli, Robyn M. Javier, Ladislav Mrzljak, Vahri Beaumont, Geoffrey Tombaugh, Kimmo Lehtimäki, Arash Rassoulpour, Ignacio Munoz-Sanjuan, and Andreas Ebneth
- Subjects
0301 basic medicine ,Genetically modified mouse ,Male ,alpha7 Nicotinic Acetylcholine Receptor ,Microdialysis ,Central nervous system ,Green Fluorescent Proteins ,Mice, Transgenic ,Pharmacology ,Biology ,In Vitro Techniques ,Kynurenic Acid ,Transfection ,Hippocampus ,Receptors, N-Methyl-D-Aspartate ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Kynurenic acid ,Kynurenine 3-Monooxygenase ,Developmental Neuroscience ,Huntington's disease ,Trinucleotide Repeats ,medicine ,Animals ,Humans ,Enzyme Inhibitors ,Analysis of Variance ,Huntingtin Protein ,Dose-Response Relationship, Drug ,Neurodegeneration ,Brain ,Excitatory Postsynaptic Potentials ,Quinolinic Acid ,medicine.disease ,Electric Stimulation ,Electrophysiological Phenomena ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,HEK293 Cells ,Huntington Disease ,Pyrimidines ,Neurology ,chemistry ,Kynurenine ,Quinolinic acid ,Signal Transduction - Abstract
Dysregulation of the kynurenine (Kyn) pathway has been associated with the progression of Huntington's disease (HD). In particular, elevated levels of the kynurenine metabolites 3-hydroxy kynurenine (3-OH-Kyn) and quinolinic acid (Quin), have been reported in the brains of HD patients as well as in rodent models of HD. The production of these metabolites is controlled by the activity of kynurenine mono-oxygenase (KMO), an enzyme which catalyzes the synthesis of 3-OH-Kyn from Kyn. In order to determine the role of KMO in the phenotype of mouse models of HD, we have developed a potent and selective KMO inhibitor termed CHDI-340246. We show that this compound, when administered orally to transgenic mouse models of HD, potently and dose-dependently modulates the Kyn pathway in peripheral tissues and in the central nervous system. The administration of CHDI-340246 leads to an inhibition of the formation of 3-OH-Kyn and Quin, and to an elevation of Kyn and Kynurenic acid (KynA) levels in brain tissues. We show that administration of CHDI-340246 or of Kyn and of KynA can restore several electrophysiological alterations in mouse models of HD, both acutely and after chronic administration. However, using a comprehensive panel of behavioral tests, we demonstrate that the chronic dosing of a selective KMO inhibitor does not significantly modify behavioral phenotypes or natural progression in mouse models of HD.
- Published
- 2015
38. Characterization of HTT inclusion size, location, and timing in the zQ175 mouse model of Huntington's disease: an in vivo high-content imaging study
- Author
-
Volker Mack, Andreas Ebneth, Karsten Kottig, Diana Scholz, Karsten Tillack, Seung Kwak, George J. Yohrling, Ignacio Munoz-Sanjuan, Heinz Von Der Kammer, Nadege Berson, Christina Gabrysiak, Yalda Sedaghat, Nikisha Carty, and Christina Thiede
- Subjects
Pathology ,medicine.medical_specialty ,Heterozygote ,Huntingtin ,Cytoplasmic inclusion ,Intranuclear Inclusion Bodies ,Caudate nucleus ,Fluorescent Antibody Technique ,lcsh:Medicine ,Mice, Transgenic ,Nerve Tissue Proteins ,Striatum ,Biology ,Protein Aggregation, Pathological ,Inclusion bodies ,Mice ,Huntington's disease ,medicine ,Animals ,Nuclear protein ,lcsh:Science ,Cerebral Cortex ,Inclusion Bodies ,Huntingtin Protein ,Multidisciplinary ,lcsh:R ,Nuclear Proteins ,medicine.disease ,Corpus Striatum ,Cell biology ,Disease Models, Animal ,Protein Transport ,Huntington Disease ,Phenotype ,Gene Knockdown Techniques ,lcsh:Q ,Trinucleotide repeat expansion ,Research Article - Abstract
Huntington’s disease (HD) is an autosomal dominant neurodegenerative disorder caused by a CAG trinucleotide repeat expansion in the huntingtin gene. Major pathological hallmarks of HD include inclusions of mutant huntingtin (mHTT) protein, loss of neurons predominantly in the caudate nucleus, and atrophy of multiple brain regions. However, the early sequence of histological events that manifest in region- and cell-specific manner has not been well characterized. Here we use a high-content histological approach to precisely monitor changes in HTT expression and characterize deposition dynamics of mHTT protein inclusion bodies in the recently characterized zQ175 knock-in mouse line. We carried out an automated multi-parameter quantitative analysis of individual cortical and striatal cells in tissue slices from mice aged 2–12 months and confirmed biochemical reports of an age-associated increase in mHTT inclusions in this model. We also found distinct regional and subregional dynamics for inclusion number, size and distribution with subcellular resolution. We used viral-mediated suppression of total HTT in the striatum of zQ175 mice as an example of a therapeutically-relevant but heterogeneously transducing strategy to demonstrate successful application of this platform to quantitatively assess target engagement and outcome on a cellular basis.
- Published
- 2015
39. Hallmarks of Alzheimer’s Disease in Stem-Cell-Derived Human Neurons Transplanted into Mouse Brain
- Author
-
John Hardy, Karelle Leroy, Elke Vanden Eynden, Ira Espuny-Camacho, Sebastian Munck, Enrico Radaelli, Amaia M. Arranz, Ina Tesseur, Laurie Lambot, Bart De Strooper, Andreas Ebneth, Pierre Vanderhaeghen, Mark Fiers, Lorna Omodho, Selina Wray, Jean Pierre Brion, Kunie Ando, Jérôme Bonnefont, An Snellinx, and Nikky Corthout
- Subjects
Pluripotent Stem Cells ,0301 basic medicine ,Neurite ,Cellular differentiation ,tau Proteins ,Biology ,Mice ,03 medical and health sciences ,Downregulation and upregulation ,Alzheimer Disease ,Neurites ,medicine ,Animals ,Humans ,Phosphorylation ,Induced pluripotent stem cell ,Neurons ,Cell Death ,General Neuroscience ,Neurodegeneration ,Neuron projection ,Brain ,Cell Differentiation ,medicine.disease ,030104 developmental biology ,Stem cell ,Alzheimer's disease ,Neuroscience - Abstract
Human pluripotent stem cells (PSCs) provide a unique entry to study species-specific aspects of human disorders such as Alzheimer's disease (AD). However, in vitro culture of neurons deprives them of their natural environment. Here we transplanted human PSC-derived cortical neuronal precursors into the brain of a murine AD model. Human neurons differentiate and integrate into the brain, express 3R/4R Tau splice forms, show abnormal phosphorylation and conformational Tau changes, and undergo neurodegeneration. Remarkably, cell death was dissociated from tangle formation in this natural 3D model of AD. Using genome-wide expression analysis, we observed upregulation of genes involved in myelination and downregulation of genes related to memory and cognition, synaptic transmission, and neuron projection. This novel chimeric model for AD displays human-specific pathological features and allows the analysis of different genetic backgrounds and mutations during the course of the disease.
- Published
- 2017
- Full Text
- View/download PDF
40. Screening lead compounds for QT interval prolongation
- Author
-
Ulrike Bischoff, Rainer Netzer, Olaf Pongs, and Andreas Ebneth
- Subjects
Pharmacology ,biology ,Heart disease ,Drug discovery ,business.industry ,hERG ,KCNE2 ,medicine.disease ,QT interval ,Drug development ,In vivo ,Drug Discovery ,biology.protein ,medicine ,Pharmaceutical sciences ,business - Abstract
The late detection of cardiotoxic side effects, such as QT prolongation, induced by compounds of pharmacological interest can dramatically impede drug discovery and development projects, and consequently increase their cost. The launch of new drugs with undetected cardiotoxic side effects could have hazardous consequences and could trigger lethal cardiac dysrhythmias in patients. It is desirable, therefore, to test for the potential cardiotoxic side effects of compounds at an early stage of drug development. Electrophysiological test systems and cellular-based fluorometric high-throughput assays are now available for cloned human cardiac ion channels. These test systems are important tools in the preclinical safety evaluation of drugs and newly developed compounds.
- Published
- 2001
- Full Text
- View/download PDF
41. Automated brain region recognition based on elastic registration and atlas mapping
- Author
-
Michaël, Barbier, primary, Astrid, Bottelbergs, additional, Rosanne, Verboven, additional, Rony, Nuydens, additional, Andreas, Ebneth, additional, and Winnok, De Vos, additional
- Published
- 2016
- Full Text
- View/download PDF
42. MARK, a Novel Family of Protein Kinases That Phosphorylate Microtubule-Associated Proteins and Trigger Microtubule Disruption
- Author
-
Eva-Maria Mandelkow, Eckhard Mandelkow, Gerard Drewes, Andreas Ebneth, and Ute Preuss
- Subjects
DNA, Complementary ,Microtubule-associated protein ,Swine ,Recombinant Fusion Proteins ,Molecular Sequence Data ,Hyperphosphorylation ,Gene Expression ,tau Proteins ,CHO Cells ,Biology ,Protein Serine-Threonine Kinases ,Microtubules ,General Biochemistry, Genetics and Molecular Biology ,Substrate Specificity ,Microtubule ,Cricetinae ,Cell polarity ,Animals ,Humans ,Amino Acid Sequence ,RNA, Messenger ,Cloning, Molecular ,Phosphorylation ,Peptide sequence ,Phylogeny ,Protein-Serine-Threonine Kinases ,Sequence Homology, Amino Acid ,Kinase ,Biochemistry, Genetics and Molecular Biology(all) ,Sequence Analysis, DNA ,Peptide Fragments ,Cell biology ,Rats ,Molecular Weight ,Organ Specificity - Abstract
MARK phosphorylates the microtubule-associated proteins tau, MAP2, and MAP4 on their microtubule-binding domain, causing their dissociation from microtubules and increased microtubule dynamics. We describe the molecular cloning, distribution, activation mechanism, and overexpression of two MARK proteins from rat that arise from distinct genes. They encode Ser/Thr kinases of 88 and 81 kDa, respectively, and show similarity to the yeast kin1+ and C. elegans par-1 genes that are involved in the establishment of cell polarity. Expression of both isoforms is ubiquitous, and homologous genes are present in humans. Catalytic activity depends on phosphorylation of two residues in subdomain VIII. Overexpression of MARK in cells leads to hyperphosphorylation of MAPs on KXGS motifs and to disruption of the microtubule array, resulting in morphological changes and cell death.
- Published
- 1997
- Full Text
- View/download PDF
43. Development of LC/MS/MS, high-throughput enzymatic and cellular assays for the characterization of compounds that inhibit kynurenine monooxygenase (KMO)
- Author
-
Michael Prime, Celia Dominguez, Ignacio Munoz-Sanjuan, Andreas Ebneth, Maria Beconi, Leticia Toledo-Sherman, and Dirk Winkler
- Subjects
CHO Cells ,Biology ,Tandem mass spectrometry ,Biochemistry ,Peripheral blood mononuclear cell ,Analytical Chemistry ,Cell Line ,chemistry.chemical_compound ,Mice ,Cricetulus ,Dogs ,Kynurenine 3-Monooxygenase ,In vivo ,Tandem Mass Spectrometry ,Drug Discovery ,Animals ,Humans ,Rats, Wistar ,Kynurenine ,Enzyme Assays ,chemistry.chemical_classification ,Monooxygenase ,Rats ,Mice, Inbred C57BL ,Macaca fascicularis ,Enzyme ,HEK293 Cells ,Huntington Disease ,chemistry ,Cell culture ,Leukocytes, Mononuclear ,Molecular Medicine ,Biotechnology ,Chromatography, Liquid - Abstract
Kynurenine monooxygenase (KMO) catalyzes the conversion of kynurenine to 3-hydroxykynurenine. Modulation of KMO activity has been implicated in several neurodegenerative diseases, including Huntington disease. Our goal is to develop potent and selective small-molecule KMO inhibitors with suitable pharmacokinetic characteristics for in vivo proof-of-concept studies and subsequent clinical development. We developed a comprehensive panel of biochemical and cell-based assays that use liquid chromatography/tandem mass spectrometry to quantify unlabeled kynurenine and 3-hydroxykynurenine. We describe assays to measure KMO inhibition in cell and tissue extracts, as well as cellular assays including heterologous cell lines and primary rat microglia and human peripheral blood mononuclear cells.
- Published
- 2013
44. The structure of mammalian serine racemase: evidence for conformational changes upon inhibitor binding
- Author
-
Myron A, Smith, Volker, Mack, Andreas, Ebneth, Isabel, Moraes, Brunella, Felicetti, Michael, Wood, Dorian, Schonfeld, Owen, Mather, Andrea, Cesura, and John, Barker
- Subjects
Racemases and Epimerases ,Crystallography, X-Ray ,Receptors, N-Methyl-D-Aspartate ,Malonates ,Protein Structure, Tertiary ,Rats ,Catalytic Domain ,Pyridoxal Phosphate ,Protein Structure and Folding ,Serine ,Animals ,Humans ,Enzyme Inhibitors ,Protein Multimerization ,Protein Structure, Quaternary ,Protein Binding - Abstract
Serine racemase is responsible for the synthesis of D-serine, an endogenous co-agonist for N-methyl-D-aspartate receptor-type glutamate receptors (NMDARs). This pyridoxal 5'-phosphate-dependent enzyme is involved both in the reversible conversion of L- to D-serine and serine catabolism by alpha,beta-elimination of water, thereby regulating D-serine levels. Because D-serine affects NMDAR signaling throughout the brain, serine racemase is a promising target for the treatment of disorders related to NMDAR dysfunction. To provide a molecular basis for rational drug design the x-ray crystal structures of human and rat serine racemase were determined at 1.5- and 2.1-A resolution, respectively, and in the presence and absence of the orthosteric inhibitor malonate. The structures revealed a fold typical of beta-family pyridoxal 5'-phosphate enzymes, with both a large domain and a flexible small domain associated into a symmetric dimer, and indicated a ligand-induced rearrangement of the small domain that organizes the active site for specific turnover of the substrate.
- Published
- 2010
45. Arylsulfonamide CB2 receptor agonists: SAR and optimization of CB2 selectivity
- Author
-
Jenkins James Edward, Andreas Ebneth, Doris Riether, Andreas F. Kahrs, Sabine Löbbe, Innocent Mushi, Taylor Malcolm, Kathy O’Shea, Daw-Tsun Shih, Edward Walker, Brian W. Dymock, Monika Ermann, Mark L. Brewer, David S. Thomson, Stephen P. East, Beatriz Noya-Marino, Mark J. Gemkow, and Patricia Amouzegh
- Subjects
Agonist ,Cannabinoid receptor ,Molecular Structure ,Stereochemistry ,Chemistry ,medicine.drug_class ,medicine.medical_treatment ,High-throughput screening ,Organic Chemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Carboxamide ,Biochemistry ,Receptor, Cannabinoid, CB2 ,Structure-Activity Relationship ,Receptor, Cannabinoid, CB1 ,Drug Discovery ,medicine ,Cannabinoid receptor type 2 ,Molecular Medicine ,Structure–activity relationship ,lipids (amino acids, peptides, and proteins) ,Cannabinoid ,Receptor ,Molecular Biology - Abstract
A high-throughput screening campaign resulted in the discovery of a highly potent dual cannabinoid receptor 1 (CB1) and 2 (CB2) agonist. Following a thorough SAR exploration, a series of selective CB2 full agonists were identified.
- Published
- 2007
46. Evaluation of a high-throughput fluorescence assay method for HERG potassium channel inhibition
- Author
-
Christian M. Apfel, Hendrick Bothmann, Arnulf Dorn, Andreas Ebneth, Francis Hermann, Gerhard Trube, and Klaus Christensen
- Subjects
0301 basic medicine ,ERG1 Potassium Channel ,Patch-Clamp Techniques ,hERG ,CHO Cells ,Pharmacology ,01 natural sciences ,Biochemistry ,QT interval ,Analytical Chemistry ,03 medical and health sciences ,Cricetinae ,Animals ,Patch clamp ,Fluorescent Dyes ,biology ,Drug discovery ,Chemistry ,Chinese hamster ovary cell ,Reproducibility of Results ,Potassium channel ,Ether-A-Go-Go Potassium Channels ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,030104 developmental biology ,Spectrometry, Fluorescence ,Drug development ,Potassium Channels, Voltage-Gated ,biology.protein ,Biophysics ,Molecular Medicine ,Biotechnology - Abstract
The number of projects in drug development that fail in late phases because of cardiac side effects such as QT prolongation can impede drug discovery and development of projects. The molecular target responsible for QT prolongation by a wide range of pharmaceutical agents is the myocardial hERG potassium channel. It is therefore desirable to screen for compound interactions with the hERG channel at an early stage of drug development. Here, the authors report a cell-based fluorescence assay using membrane potential-sensitive fluorescent dyes and stably transfected hERG channels from CHO cells. The assay allows semiautomated screening of compounds for hERG activity on 384-well plates and is sufficiently rapid for testing a large number of compounds. The assay is robust as indicated by a Z' factor larger than 0.6. The throughput is in the range of 10,000 data points per day, which is significantly higher than any other method presently available for hERG. The data obtained with the fluorescence assay were in qualitative agreement with those from patch-clamp electrophysiological analysis. There were no false-positive hits, and the rate of false-negative compounds is currently 12% but might be further reduced by testing compounds at higher concentration. Quantitative differences between fluorescence and electrophysiological methods may be due to the use- or voltage-dependent activity of the antagonists.
- Published
- 2005
47. MARK4 is a novel microtubule-associated proteins/microtubule affinity-regulating kinase that binds to the cellular microtubule network and to centrosomes
- Author
-
Andreas Ebneth, Miro Brajenovic, B. Trinczek, and Gerard Drewes
- Subjects
Cytoplasm ,DNA, Complementary ,Microtubule-associated protein ,Amino Acid Motifs ,Green Fluorescent Proteins ,Molecular Sequence Data ,tau Proteins ,CHO Cells ,Biology ,Protein Serine-Threonine Kinases ,Transfection ,Biochemistry ,Microtubules ,Cell Line ,Neuroblastoma ,Microtubule ,Alzheimer Disease ,Catalytic Domain ,Cell Line, Tumor ,Cricetinae ,Serine ,Animals ,Humans ,Amino Acid Sequence ,Cloning, Molecular ,Phosphorylation ,Molecular Biology ,Phylogeny ,Microtubule nucleation ,Centrosome ,Sequence Homology, Amino Acid ,Kinase ,Brain ,Microtubule organizing center ,Cell Differentiation ,Cell Biology ,Blotting, Northern ,Cosmids ,Precipitin Tests ,Cell biology ,Protein Structure, Tertiary ,Luminescent Proteins ,Microtubule-Associated Proteins ,Protein Binding - Abstract
The MARK protein kinases were originally identified by their ability to phosphorylate a serine motif in the microtubule-binding domain of tau that is critical for microtubule binding. Here, we report the cloning and expression of a novel human paralog, MARK4, which shares 75% overall homology with MARK1-3 and is predominantly expressed in brain. Homology is most pronounced in the catalytic domain (90%), and MARK4 readily phosphorylates tau and the related microtubule-associated protein 2 (MAP2) and MAP4. In contrast to the three paralogs that all exhibit uniform cytoplasmic localization, MARK4 colocalizes with the centrosome and with microtubules in cultured cells. Overexpression of MARK4 causes thinning out of the microtubule network, concomitant with a reorganization of microtubules into bundles. In line with these findings, we show that a tandem affinity-purified MARK4 protein complex contains alpha-, beta-, and gamma-tubulin. In differentiated neuroblastoma cells, MARK4 is localized prominently at the tips of neurite-like processes. We suggest that although the four MARK/PAR-1 kinases might play multiple cellular roles in concert with different targets, MARK4 is likely to be directly involved in microtubule organization in neuronal cells and may contribute to the pathological phosphorylation of tau in Alzheimer's disease.
- Published
- 2003
48. HTS techniques to investigate the potential effects of compounds on cardiac ion channels at early-stages of drug discovery
- Author
-
Rainer, Netzer, Ulrike, Bischoff, and Andreas, Ebneth
- Subjects
Long QT Syndrome ,Drug Design ,Myocardium ,Drug Evaluation, Preclinical ,Animals ,Humans ,Heart ,Rubidium ,Ion Channels ,Fluorescent Dyes - Abstract
Within the past few years, the high-throughput screening (HTS) of compounds targeting cardiac ion channels has been primarily focused on the testing of the HERG channel, which is involved in the termination of cardiac action potential. Interaction of drugs with this channel may induce QT interval prolongation and cardiac arrhythmia. These undesirable side effects have forced several pharmaceutical companies to terminate drug discovery and development projects. The screening of compounds for HERG-mediated activity early in the drug development process may thus help reduce the number of compounds that are withdrawn from late preclinical or early clinical trials due to cardiovascular side effects. However, early screening implies the ability to test large numbers of compounds. Therefore, tests have to be performed rapidly, combining high-throughput and low costs, and allow the use of small amounts of compounds. In this review, the HTS systems currently available to investigate the potential effects of compounds on the activity of the cardiac HERG ion channel will be described and compared.
- Published
- 2003
49. Ion channel screening technologies: will they revolutionize drug discovery?
- Author
-
Andreas Ebneth
- Subjects
Pharmacology ,Drug Industry ,Computer science ,business.industry ,Drug discovery ,High-throughput screening ,Drug Discovery ,Drug Evaluation, Preclinical ,Nanotechnology ,business ,Ion channel ,Ion Channels ,Biotechnology - Published
- 2002
50. Identification and functional characterization of a novel KCNE2 (MiRP1) mutation that alters HERG channel kinetics
- Author
-
Andreas Ebneth, Martin Borggrefe, Dirk Isbrandt, Patrick Friederich, Eric Schulze-Bahr, Harald Funke, Olaf Pongs, Anna Solth, Kathrin Sauter, Wilhelm Haverkamp, and Günter Breithardt
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,ERG1 Potassium Channel ,Potassium Channels ,Genotype ,Mutant ,hERG ,Mutation, Missense ,CHO Cells ,Biology ,medicine.disease_cause ,Sudden death ,Transcriptional Regulator ERG ,Cricetinae ,Drug Discovery ,medicine ,Missense mutation ,Myocyte ,Animals ,Humans ,cardiovascular diseases ,Cation Transport Proteins ,Genetics (clinical) ,Genetics ,Mutation ,Chinese hamster ovary cell ,KCNE2 ,Electric Stimulation ,Ether-A-Go-Go Potassium Channels ,Cell biology ,DNA-Binding Proteins ,Long QT Syndrome ,Potassium Channels, Voltage-Gated ,biology.protein ,Trans-Activators ,Molecular Medicine ,Female ,Subcellular Fractions - Abstract
Long-QT syndrome (LQTS) may cause syncope and sudden death due to cardiac tachyarrhythmia. Chromosome 7-linked LQTS (LQT2) has been correlated with mutations in the human ether-a-go-go-related gene (HERG). HERG forms voltage-gated K channels that may be associated with Mink-related peptide 1 (MiRP1), an auxiliary beta-subunit. The channels mediate currents that resemble native I(Kr). Mutations in the KCNE2 gene encoding MiRP1 may also cause LQTS. In this study, the frequency of mutations in KCNE2 of 150 unrelated LQTS patients without known genotype and of 100 controls was analyzed using single-strand conformation polymorphism analysis and direct sequencing. We identified a novel missense mutation, V65 M, in the KCNE2 gene of a 17-year-old female with syncope and LQTS. Expression studies in Chinese hamster ovary cells revealed that mutant and wild-type MiRP1 co-localized with HERG subunits and formed functional channels. However, mutant HERG/MiRP1(V65M) channels mediated currents with an accelerated inactivation time course compared with wild-type channels. The accelerated inactivation time course of HERG/MiRP1(V65M) channels may decrease I(Kr) current density of myocardial cells, thereby impairing the ability of myocytes to repolarize in response to sudden membrane depolarizations such as extrasystoles.
- Published
- 2001
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.