27 results on '"Müller-Schiffmann, A."'
Search Results
2. Viruses as ‘Truffle Hounds’: Molecular Tools for Untangling Brain Cellular Pathology
- Author
-
Svenja V. Trossbach, Carsten Korth, Vishwanath R. Lingappa, and Andreas Müller-Schiffmann
- Subjects
0301 basic medicine ,Cellular pathology ,Drug discovery ,viruses ,General Neuroscience ,Brain ,Host factors ,Computational biology ,Biology ,Protein aggregation ,Protein Aggregation, Pathological ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Proteostasis ,Multiprotein Complexes ,Viruses ,Humans ,Identification (biology) ,030217 neurology & neurosurgery ,Host protein - Abstract
The ability of viruses to evolve several orders of magnitude faster than their host cells has enabled them to exploit host cellular machinery by selectively recruiting multiprotein complexes (MPCs) for their catalyzed assembly and replication. This hijacking may depend on alternative, 'moonlighting' functions of host proteins that deviate from their canonical functions thereby inducing cellular pathology. Here, we posit that if virus-induced cellular pathology is similar to that of other, unknown (non-viral) causes, the identification and molecular characterization of the host proteins involved in virus-mediated cellular pathology can be leveraged to decipher the non-viral disease-relevant mechanisms. We focus on how virus-induced aberrant proteostasis and protein aggregation resemble the cellular pathology of sporadic neurodegenerative diseases (NDs) and how this can be exploited for drug discovery.
- Published
- 2021
- Full Text
- View/download PDF
3. The interaction of insoluble Amyloid‐β with soluble Amyloid‐β dimers decreases Amyloid‐β plaque numbers
- Author
-
Kathy Keyvani, Lothar Gremer, Arne Herring, Else F van Gerresheim, Andreas Müller-Schiffmann, and Carsten Korth
- Subjects
0301 basic medicine ,Genetically modified mouse ,Histology ,Amyloid beta ,Medizin ,Mice, Transgenic ,Plaque, Amyloid ,Oligomer ,Pathology and Forensic Medicine ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Alzheimer Disease ,In vivo ,Physiology (medical) ,Animals ,Humans ,Cognitive Dysfunction ,ddc:610 ,Cognitive decline ,Neuroinflammation ,Amyloid beta-Peptides ,biology ,Brain ,Peptide Fragments ,In vitro ,030104 developmental biology ,Neurology ,chemistry ,Neuroinflammatory Diseases ,Biophysics ,biology.protein ,Thioflavin ,Neurology (clinical) ,030217 neurology & neurosurgery - Abstract
Objectives: The heterogeneity of Amyloid-beta (Aβ) plaque load in patients with Alzheimer's disease (AD) has puzzled neuropathology. Since brain Aβ plaque load does not correlate with cognitive decline, neurotoxic soluble Aβ oligomers have been championed as disease-causing agents in early AD. So far, investigating molecular interactions between soluble oligomeric Aβ and insoluble Aβ in vivo has been difficult because of the abundance of Aβ oligomer species and the kinetic equilibrium in which they coexist. Here, we investigated whether Aβ plaque heterogeneity relates to interactions of different Aβ conformers. Materials and Methods: We took advantage of transgenic mice that generate exclusively Aβ dimers (tgDimer mice) but do not develop Aβ plaques or neuroinflammation during their lifetime, crossed them to the transgenic CRND8 mice that develop plaques after 90 days and measured Aβ plaque load using immunohistochemical and biochemical assays. Furthermore, we performed in vitro thioflavin T (ThT) aggregation assays titrating synthetic Aβ₄₂-S8C dimers into fibril-forming synthetic Aβ₄₂. Results: We observed a lower number of Aβ plaques in the brain of double transgenic mice compared to tgCRND8 mice alone while the average plaque size remained unaltered. Corroborating these in vivo findings, synthetic Aβ-S8C dimers inhibited fibril formation of wild-type Aβ also in vitro, seen by an increased half-time in the ThT assay. Conclusions: Our study indicates that Aβ dimers directly interfere with Aβ fibril formation in vivo and in vitro. The variable interaction of Aβ dimers with insoluble Aβ seeds could thus contribute to the heterogeneity of Aβ plaque load in AD patients.
- Published
- 2021
- Full Text
- View/download PDF
4. Disruption of cellular proteostasis by H1N1 influenza A virus causes α-synuclein aggregation
- Author
-
Svenja V. Trossbach, Shriya Sahu, Suganya Selvarajah, Andreas Müller-Schiffmann, Ana Raquel Moreira, Vishwanath R. Lingappa, Irina Soloviev, Ingrid Prikulis, Sebastian Hänsch, Carsten Korth, Rita Marreiros, and Stefanie Weidtkamp-Peters
- Subjects
0301 basic medicine ,Synucleinopathies ,Cell ,Nerve Tissue Proteins ,Protein aggregation ,Biology ,Virus ,Mice ,03 medical and health sciences ,Influenza A Virus, H1N1 Subtype ,0302 clinical medicine ,Orthomyxoviridae Infections ,Influenza, Human ,medicine ,Animals ,Humans ,Homeodomain Proteins ,Mice, Knockout ,Multidisciplinary ,Dopaminergic Neurons ,Autophagy ,Neurotoxicity ,medicine.disease ,Cell biology ,030104 developmental biology ,Proteostasis ,medicine.anatomical_structure ,PNAS Plus ,Knockout mouse ,alpha-Synuclein ,Female ,Protein Multimerization ,030217 neurology & neurosurgery - Abstract
Neurodegenerative diseases feature specific misfolded or misassembled proteins associated with neurotoxicity. The precise mechanisms by which protein aggregates first arise in the majority of sporadic cases have remained unclear. Likely, a first critical mass of misfolded proteins starts a vicious cycle of a prion-like expansion. We hypothesize that viruses, having evolved to hijack the host cellular machinery for catalyzing their replication, lead to profound disturbances of cellular proteostasis, resulting in such a critical mass of protein aggregates. Here, we investigated the effect of influenza virus (H1N1) strains on proteostasis of proteins associated with neurodegenerative diseases in Lund human mesencephalic dopaminergic cells in vitro and infection of Rag knockout mice in vivo. We demonstrate that acute H1N1 infection leads to the formation of α-synuclein and Disrupted-in-Schizophrenia 1 (DISC1) aggregates, but not of tau or TDP-43 aggregates, indicating a selective effect on proteostasis. Oseltamivir phosphate, an antiinfluenza drug, prevented H1N1-induced α-synuclein aggregation. As a cell pathobiological mechanism, we identified H1N1-induced blocking of autophagosome formation and inhibition of autophagic flux. In addition, α-synuclein aggregates appeared in infected cell populations connected to the olfactory bulbs following intranasal instillation of H1N1 in Rag knockout mice. We propose that H1N1 virus replication in neuronal cells can induce seeds of aggregated α-synuclein or DISC1 that may be able to initiate further detrimental downstream events and should thus be considered a risk factor in the pathogenesis of synucleinopathies or a subset of mental disorders. More generally, aberrant proteostasis induced by viruses may be an underappreciated factor in initiating protein misfolding.
- Published
- 2020
- Full Text
- View/download PDF
5. All
- Author
-
Eduard V, Bocharov, Lothar, Gremer, Anatoly S, Urban, Ivan S, Okhrimenko, Pavel E, Volynsky, Kirill D, Nadezhdin, Olga V, Bocharova, Daniil A, Kornilov, Yuliya A, Zagryadskaya, Anna V, Kamynina, Pavel K, Kuzmichev, Janine, Kutzsche, Najoua, Bolakhrif, Andreas, Müller-Schiffmann, Norbert A, Dencher, Alexander S, Arseniev, Roman G, Efremov, Valentin I, Gordeliy, and Dieter, Willbold
- Subjects
Amyloid beta-Protein Precursor ,Mice ,Alzheimer Disease ,Animals ,Humans ,Stereoisomerism ,Amino Acid Sequence ,Molecular Dynamics Simulation ,Oligopeptides ,Protein Binding - Abstract
Alzheimer's disease (AD) is a severe neurodegenerative pathology with no effective treatment known. Toxic amyloid-β peptide (Aβ) oligomers play a crucial role in AD pathogenesis. All
- Published
- 2021
6. Oxidized MIF is an Alzheimer’s Disease drug target relaying external risk factors to tau pathology
- Author
-
Andreas Müller-Schiffmann, Felix Torres, Anatolly Kitaygorodskyy, Anand Ramani, Argyro Alatza, Sarah K. Tschirner, Ingrid Prikulis, Shaofeng Yu, Debendranath Dey, Suguna Mallesh, Dharma Prasad, Dennis Solas, Verian Bader, Annemieke Rozemuller, Selina Wray, Jay Gopalakrishnan, Roland Riek, Vishwanath R. Lingappa, and Carsten Korth
- Subjects
Cellular pathology ,Herpes simplex virus ,Viral replication ,Cell culture ,medicine ,Macrophage migration inhibitory factor ,Alzheimer's disease ,Biology ,medicine.disease ,medicine.disease_cause ,Virology ,Virus ,Host factor - Abstract
Herpes virus infections are endemic and ubiquitous. While only rarely leading to overt encephalitis, subchronic or latent infections have been associated to a variety of conditions, including Alzheimers disease (AD). The cellular consequences of herpes virus infection are determined by the host proteins recruited during virus replication and assembly. Identifying such virus-recruited host proteins therefore allows the interrogation fundamental cellular events leading to associated "sporadic" diseases. A host protein-targeted small molecule drug highly active against herpes simplex virus 1 (HSV-1) infection in human brain organoids and cell lines was identified to interact with macrophage migration inhibitory factor (MIF) where it acted by intercalating between MIF units within a trimer, as determined by nuclear magnetic resonance (NMR). MIF knockout cells showed a decreased viral antigen/titer ratio corroborating its role in virus assembly. From post-mortem brain homogenates of patients with Braak 6-staged AD the small molecule lead compound specifically eluted a MIF subpopulation that correlated with the oxidized conformer of MIF (oxMIF). HSV-1 led to an increase in tau phosphorylation at distinct residues, and the lead compound decreased tau phosphorylation in recombinant cell lines expressing mutant tau and in neuron-differentiated iPSCs also in the absence of HSV-1 infection. We conclude that MIF is a cellular host factor involved in HSV-1 replication and a drug target with antiviral efficacy. At the same time, MIF also plays a role in tau phosphorylation and is enriched in an oxidized conformation in brains of AD patients. MIF thus presents as a molecular link connecting HSV-1 infection and cellular pathology characteristic of neurodegenerative diseases involving aberrant tau phosphorylation.
- Published
- 2021
- Full Text
- View/download PDF
7. A Pan-respiratory Antiviral Chemotype Targeting a Transient Host Multiprotein Complex
- Author
-
Andreas Müller-Schiffmann, Maya Michon, Anuradha F. Lingappa, Shao Feng Yu, Li Du, Fred Deiter, Sean Broce, Suguna Mallesh, Jackelyn Crabtree, Usha F. Lingappa, Amanda Macieik, Lisa Müller, Philipp Niklas Ostermann, Marcel Andrée, Ortwin Adams, Heiner Schaal, Robert J. Hogan, Ralph A. Tripp, Umesh Appaiah, Sanjeev K. Anand, Thomas W. Campi, Michael J. Ford, Jonathan C. Reed, Jim Lin, Olayemi Akintunde, Kiel Copeland, Christine Nichols, Emma Petrouski, A. Raquel Moreira, I-ting Jiang, Nicholas DeYarman, Ian Brown, Sharon Lau, Ilana Segal, Danielle Goldsmith, Shi Hong, Vinod Asundi, Erica M. Briggs, Ngwe Sin Phyo, Markus Froehlich, Bruce Onisko, Kent Matlack, Debendranath Dey, Jaisri R. Lingappa, M. Dharma Prasad, Anatoliy Kitaygorodskyy, Dennis Solas, Homer Boushey, John Greenland, Satish Pillai, Michael K. Lo, Joel M. Montgomery, Christina F. Spiropoulou, Carsten Korth, Suganya Selvarajah, Kumar Paulvannan, and Vishwanath R. Lingappa
- Subjects
Drug ,Innate immune system ,biology ,viruses ,media_common.quotation_subject ,Common cold ,medicine.disease ,biology.organism_classification ,Virology ,Article ,Virus ,Cell culture ,medicine ,Viral disease ,Porcine epidemic diarrhea virus ,Cytokine storm ,media_common - Abstract
Antiviral compounds displaying several remarkable features have been identified by a uniquely enabling drug screen and advanced through validation in two animal models, and in human primary bronchial epithelial cells grown to an air-liquid interface (ALI) and infected with SARS-CoV-2 (Brazil). Activity is observed in the nanomolar range in mammalian cells in vitro against the six viral families causing most human respiratory viral disease, irrespective of strain, including SARS-CoV-2 delta variant. A substantial barrier to development of viral resistance is demonstrated for influenza (FLUV). The drug target is an allosteric site on a novel host multi-protein complex (MPC) formed transiently, in an energy-dependent fashion, and composed of proteins implicated in viral lifecycles and innate immunity. The composition of this host MPC is modified in viral family-specific ways by FLUV and CoV, and substantially restored to the uninfected state with drug treatment. SQSTM1/p62, a key regulator of the autophagy pathway of innate immunity is i) found in the target MPC from uninfected cells, ii) lost upon infection, and iii) restored by drug treatment of infected cells, as assessed by drug resin affinity chromatography. A small subset of 14-3-3 is identified as the host protein to which the drug is bound. Advanced compounds with good oral bioavailability, half-life, lung exposure, and safety are approaching criteria for a Target Product Profile. We propose these novel drug targets to comprise a previously unappreciated molecular basis for homeostasis that is modified by viruses to facilitate their propagation and is restored by treatment with the therapeutic compounds presented. This discovery has transformative implications for treating respiratory viral-related disease, applicable to everything from COVID-19, seasonal influenza, common ‘winter viruses’ (respiratory syncytial virus, parainfluenza virus, rhinovirus, etc.), emerging respiratory viruses, and prevention of virus-induced asthma/COPD exacerbations. Treating respiratory viral disease with these host-targeted pan-respiratory viral family active compounds early, upon onset of symptoms of viral upper respiratory infection, irrespective of cause, should protect against progression to lower respiratory tract or systemic infection, the hallmarks of serious illness.
- Published
- 2021
- Full Text
- View/download PDF
8. Cellular Prion Protein Participates in Amyloid-β Transcytosis across the Blood—Brain Barrier
- Author
-
Benjamin Petsch, Carsten Korth, Sascha Weggen, Andreas Müller-Schiffmann, Thorsten Pflanzner, Sabrina Tschickardt, Bettina André-Dohmen, Lothar Stitz, and Claus U. Pietrzik
- Subjects
Amyloid β ,animal diseases ,Biology ,Brief Communication ,Blood–brain barrier ,Models, Biological ,Mice ,Alzheimer Disease ,mental disorders ,medicine ,Animals ,PrPC Proteins ,Prion protein ,Receptor ,Cells, Cultured ,Amyloid beta-Peptides ,Neurotoxicity ,medicine.disease ,Molecular biology ,Peptide Fragments ,In vitro ,nervous system diseases ,Cell biology ,medicine.anatomical_structure ,Neurology ,Transcytosis ,Blood-Brain Barrier ,Gene Knockdown Techniques ,biology.protein ,Neurology (clinical) ,Antibody ,Cardiology and Cardiovascular Medicine ,Protein Binding - Abstract
The blood—brain barrier (BBB) facilitates amyloid-β (Aβ) exchange between the blood and the brain. Here, we found that the cellular prion protein (PrPc), a putative receptor implicated in mediating Aβ neurotoxicity in Alzheimer's disease (AD), participates in Aβ transcytosis across the BBB. Using an in vitro BBB model, [125I]-Aβ1–40 transcytosis was reduced by genetic knockout of PrPc or after addition of a competing PrPc-specific antibody. Furthermore, we provide evidence that PrPc is expressed in endothelial cells and, that monomeric Aβ1–40 binds to PrPc. These observations provide new mechanistic insights into the role of PrPc in AD.
- Published
- 2012
- Full Text
- View/download PDF
9. Molecular linking of influenza infection to cellular pathology of protein misassembly: the case of disrupted-in-schizophrenia 1 (DISC1)
- Author
-
Ingrid Prikulis, V. Lingappa, I. Soloviev, A.R. Moreira, S. Sahu, S. Selvarajah, Carsten Korth, Andreas Müller-Schiffmann, and Rita Marreiros
- Subjects
Regulation of gene expression ,Pharmacology ,Cellular pathology ,biology ,business.industry ,Cellular homeostasis ,Disease ,medicine.disease ,Phenotype ,DISC1 ,Psychiatry and Mental health ,Proteostasis ,Neurology ,Schizophrenia ,Immunology ,medicine ,biology.protein ,Pharmacology (medical) ,Neurology (clinical) ,business ,Biological Psychiatry - Abstract
Chronic mental illnesses such as schizophrenia or recurrent affective disorders are heterogeneous in their biological causes behind the clinical, syndromatic diagnosis. Viral infection has been linked to chronic mental illnesses both by direct mechanisms and indirect mechanisms through immune stimulation. For example, studies showed that intrauterine influenza infection could be a risk factor for chronic mental illness [1]. It is also established that viral infection can lead to a change in proteostasis impairing regulatory protein networks and leading to disease. The most dramatic consequences of disrupted proteostasis are the so-called protein misfolding diseases that show as a phenotypical hallmark, microscopically visible deposits of one or several proteins, and cell degeneration. More subtle forms of proteostatic imbalance exist where protein deposits are not microscopically visible and the neurons do not die but remain functionally impaired, for example in some forms of chronic mental illness such as schizophrenia [2]. We previously reported insoluble Disrupted-in-schizophrenia 1 (DISC1) protein in a subpopulation of patients with chronic mental illness [3]. This gene/protein is involved in vulnerability to chronic mental disorders, and was discovered mutant and segregating in a large Scottish pedigree with a chromosomal translocation leading to a 3′ truncation of the gene [4]. Subsequent genetic association studies in multiple populations of different ethnicities also support the involvement of this protein in mental illness. For the present study, our hypothesis was that influenza infection could cause significant long-term impairments of specific proteostatic pathways leading to aggregated DISC1 protein in chronic mental illness patients [5]. Neuroblastoma cell lines expressing inducible full-length, non-mutant DISC1 protein were infected with WSN33 influenza (H1N1 strain). The effect of viral infection was investigated by immunocytochemistry, biochemical and functional assays. Furthermore, infected mice with WSN33 influenza where investigated for insoluble DISC1 deposits by immunohistochemistry. Antiviral drugs were tested in order to reverse the influenza-induced DISC1 aggregates. Influenza infection of cell lines induced DISC1 misassembly but not misassembly of the protein TDP43 known to be mislocalized in ALS, but not in mental illness or influenza, and therefore serving as a control for specificity of DISC1 mis-assembly upon influenza infection in cellular models. Appearance of DISC1 deposits was dependent on infectious influenza virus titer, and the infection time. Functional consequences of influenza-induced DISC1 misassembly will be reported, as well as our investigations of influenza-infected mice that are still ongoing. We conclude that influenza infection leads to a very specific proteostatic change, including the misassembly of DISC1, which has functional consequences for dopamine homeostasis. Thus a selective reprogramming of cellular homeostasis, as measured by proteostasis, may be a novel molecular mechanism by which influenza infection can lead to brain disease.
- Published
- 2019
- Full Text
- View/download PDF
10. Gesteigerte Wirksamkeit durch Synergismus: Verknüpfung unabhängiger Wirkstoffklassen zu Hybridsubstanzen
- Author
-
Anselm H. C. Horn, Marco Hellmert, Dirk Bartnik, Jolanta Polkowska, Christine Moriscot, Julia März-Berberich, Dieter Willbold, Guy Schoehn, Heinrich Sticht, Andreas Müller-Schiffmann, Aksana Andreyeva, S. Aileen Funke, Oleksandr Brener, Kurt Gottmann, Janine Kutzsche, Carsten Korth, Thomas Schrader, Luitgart Nagel-Steger, Raik Rönicke, and Klaus G. Reymann
- Subjects
Chemistry ,General Medicine - Published
- 2010
- Full Text
- View/download PDF
11. Oral Treatment with the <scp>d</scp>-Enantiomeric Peptide D3 Improves the Pathology and Behavior of Alzheimer’s Disease Transgenic Mice
- Author
-
Dirk Bartnik, Luitgard Nagel-Steger, Andreas Müller-Schiffmann, Carsten Korth, Renu Batra-Safferling, Inga Kadish, Torsten Sehl, Christine Moriscot, Oleksandr Brener, Thomas van Groen, Heinrich Sticht, Dieter Willbold, Guy Schoehn, Susanne Aileen Funke, and Anselm H. C. Horn
- Subjects
Genetically modified mouse ,Oral treatment ,Pathology ,medicine.medical_specialty ,Physiology ,Cognitive Neuroscience ,Administration, Oral ,Mice, Transgenic ,macromolecular substances ,Disease ,Biology ,Fibril ,Biochemistry ,Protein Structure, Secondary ,Pathogenesis ,Mice ,Cognition ,Alzheimer Disease ,medicine ,Animals ,Humans ,Maze Learning ,Oligopeptide ,Stereoisomerism ,Cell Biology ,General Medicine ,medicine.disease ,Protein Structure, Tertiary ,Disease Models, Animal ,Treatment Outcome ,Female ,Alzheimer's disease ,Enantiomer ,Oligopeptides - Abstract
Several lines of evidence suggest that the amyloid-β-peptide (Aβ) plays a central role in the pathogenesis of Alzheimer's disease (AD). Not only Aβ fibrils but also small soluble Aβ oligomers in particular are suspected to be the major toxic species responsible for disease development and progression. The present study reports on in vitro and in vivo properties of the Aβ targeting d-enantiomeric amino acid peptide D3. We show that next to plaque load and inflammation reduction, oral application of the peptide improved the cognitive performance of AD transgenic mice. In addition, we provide in vitro data elucidating the potential mechanism underlying the observed in vivo activity of D3. These data suggest that D3 precipitates toxic Aβ species and converts them into nonamyloidogenic, nonfibrillar, and nontoxic aggregates without increasing the concentration of monomeric Aβ. Thus, D3 exerts an interesting and novel mechanism of action that abolishes toxic Aβ oligomers and thereby supports their decisive role in AD development and progression.
- Published
- 2010
- Full Text
- View/download PDF
12. Vaccine Approaches to Prevent and Treat Prion Infection
- Author
-
Andreas Müller-Schiffmann and Carsten Korth
- Subjects
Immunogen ,Amyloid ,Prions ,medicine.drug_class ,animal diseases ,Scrapie ,Biology ,Monoclonal antibody ,Active immunization ,Epitope ,Prion Diseases ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Pharmacology ,Vaccines ,Vaccination ,Antibodies, Monoclonal ,Gene targeting ,General Medicine ,Virology ,nervous system diseases ,Disease Models, Animal ,Immunology ,biology.protein ,Antibody ,Biotechnology - Abstract
Prion diseases are transmissible neurodegenerative diseases of humans and animals. The prion agent consists of a misfolded protein, PrPSc (prion protein, scrapie form), of a glycosylphosphatidylinositol-anchored host protein, PrPC (PrP cellular form) of unknown function. During prion replication, PrPSc induces host PrPC to adopt its pathogenic conformation. Some PrPSc may aggregate to microscopically visible, extracellular prion plaques that stain for amyloid. The development of antiprion vaccines presents some challenges. While there is strong self-tolerance to an endogenous antibody response to PrPC and PrPSc, highly potent monoclonal antibodies (mAbs) have been raised in mice in which the prion protein gene has been deleted by gene targeting. These mAbs have been demonstrated to be antiprion-active in permanently scrapie-infected neuroblastoma (ScN2a) cells, primarily when bound to one of four epitopes (the octarepeat region, the region around codons 90-110, helix 1 region codons 145-160, and the extreme C-terminal codons 210-220). The mAbs directed against codon regions 90-110 or 145-160 are also antiprion-active in vivo, but only after intraperitoneal infection with prions, not intracerebral infection, suggesting their blood-brain barrier (BBB) impermeability. The challenge will be to make antibodies, or recombinant derivatives thereof, BBB permeable; this is preferably achieved by monovalent antibody fragments since divalent ones were found to be neurotoxic. Self-tolerance of wild-type animals to PrP immunizations was found to be of extrathymic origin. Even though antibodies raised in wild-type mice were found to display antiprion activity in ScN2a cells, these mice did not have significant extensions of incubation times when challenged intraperitoneally with prions. A general low affinity of these antibody responses to native surface-bound PrPC may account for this. Since wild-type mice were found to develop sufficient T-cell responses to codon regions 145-160 and 210-220, we believe that there is a theoretical chance of a successful vaccination therapy. The influence of the way the immunogen is presented has already been shown to be of major importance for the ensuing immune response, in that presentation of PrP with CpG oligodeoxynucleotides as adjuvant or viral packaging improved antibody responses. Major progress for active immunizations may therefore be expected in this field. Eradication programs will be one of the most important uses of active immunization protocols. For this purpose, vaccines will have to be inexpensive, easy to handle, and effective. In the short term, passive immunizations will likely be most promising for therapy of prion disease, including for human medical interventions. Active immunization protocols are less likely to succeed quickly, and will take years if not decades to be validated for domestic or free-ranging animals.
- Published
- 2008
- Full Text
- View/download PDF
13. Amyloid-β dimers in the absence of plaque pathology impair learning and synaptic plasticity
- Author
-
Andreas Müller-Schiffmann, Joseph P. Huston, Maria A. de Souza Silva, Anselm H. C. Horn, Carsten Korth, Heinrich Sticht, Olga A. Sergeeva, Laila Abdel-Hafiz, Arne Herring, A. N. Chepkova, Diana Wedel, Kurt Gottmann, Sandra Schäble, and Kathy Keyvani
- Subjects
0301 basic medicine ,Genetically modified mouse ,Male ,Pathology ,medicine.medical_specialty ,Amyloid ,BACE1-AS ,Medizin ,Mice, Transgenic ,Plaque, Amyloid ,Oligomer ,Hippocampus ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Organ Culture Techniques ,mental disorders ,medicine ,Amyloid precursor protein ,Animals ,Senile plaques ,Amyloid beta-Peptides ,Neuronal Plasticity ,biology ,Long-term potentiation ,Mice, Inbred C57BL ,030104 developmental biology ,chemistry ,Synaptic plasticity ,biology.protein ,Neurology (clinical) ,Protein Multimerization ,Cognition Disorders ,030217 neurology & neurosurgery - Abstract
Despite amyloid plaques, consisting of insoluble, aggregated amyloid-β peptides, being a defining feature of Alzheimer’s disease, their significance has been challenged due to controversial findings regarding the correlation of cognitive impairment in Alzheimer’s disease with plaque load. The amyloid cascade hypothesis defines soluble amyloid-β oligomers, consisting of multiple amyloid-β monomers, as precursors of insoluble amyloid-β plaques. Dissecting the biological effects of single amyloid-β oligomers, for example of amyloid-β dimers, an abundant amyloid-β oligomer associated with clinical progression of Alzheimer’s disease, has been difficult due to the inability to control the kinetics of amyloid-β multimerization. For investigating the biological effects of amyloid-β dimers, we stabilized amyloid-β dimers by an intermolecular disulphide bridge via a cysteine mutation in the amyloid-β peptide (Aβ-S8C) of the amyloid precursor protein. This construct was expressed as a recombinant protein in cells and in a novel transgenic mouse, termed tgDimer mouse. This mouse formed constant levels of highly synaptotoxic soluble amyloid-β dimers, but not monomers, amyloid-β plaques or insoluble amyloid-β during its lifespan. Accordingly, neither signs of neuroinflammation, tau hyperphosphorylation or cell death were observed. Nevertheless, these tgDimer mice did exhibit deficits in hippocampal long-term potentiation and age-related impairments in learning and memory, similar to what was observed in classical Alzheimer’s disease mouse models. Although the amyloid-β dimers were unable to initiate the formation of insoluble amyloid-β aggregates in tgDimer mice, after crossbreeding tgDimer mice with the CRND8 mouse, an amyloid-β plaque generating mouse model, Aβ-S8C dimers were sequestered into amyloid-β plaques, suggesting that amyloid-β plaques incorporate neurotoxic amyloid-β dimers that by themselves are unable to self-assemble. Our results suggest that within the fine interplay between different amyloid-β species, amyloid-β dimer neurotoxic signalling, in the absence of amyloid-β plaque pathology, may be involved in causing early deficits in synaptic plasticity, learning and memory that accompany Alzheimer’s disease.10.1093/brain/awv355_video_abstract awv355_video_abstract
- Published
- 2015
14. The E6 Protein of the Cutaneous Human Papillomavirus Type 8 Can Stimulate the Viral Early and Late Promoters by Distinct Mechanisms
- Author
-
Julia Beckmann, Andreas Müller-Schiffmann, and Gertrud Steger
- Subjects
Gene Expression Regulation, Viral ,Keratinocytes ,Immunology ,P300-CBP Transcription Factors ,Biology ,Transfection ,Microbiology ,Cell Line ,Virology ,Gene expression ,Coactivator ,medicine ,Transcriptional regulation ,Humans ,p300-CBP Transcription Factors ,Promoter Regions, Genetic ,Papillomaviridae ,Promoter ,Oncogene Proteins, Viral ,Epidermodysplasia verruciformis ,medicine.disease ,Molecular biology ,Genome Replication and Regulation of Viral Gene Expression ,Insect Science ,Mutagenesis, Site-Directed ,Trans-Activators - Abstract
The expression of the proteins encoded by human papillomaviruses (HPVs) is tightly linked to the differentiation program of the infected keratinocytes. The late promoter, expressing the structural proteins, becomes activated in the differentiated keratinocytes, while the early promoter is also active in the basal layers. We have shown previously that the viral transcriptional regulator E2 and the cellular coactivator p300 cooperate in activation of gene expression of HPV8, which infects the skin and is associated with epidermodysplasia verruciformis. Here we demonstrate that this activation is further stimulated after overexpression of the E6 oncoprotein of HPV8 (8E6). RNase protection experiments revealed that 8E6 efficiently cooperates with 8E2 and p300 in activation of the late promoter. In addition, the early promoter, which did not respond to 8E2 and/or p300, was stimulated more than fourfold by 8E6. Our data suggest that both promoters are activated via distinct mechanisms, since the activation of the early promoter was achieved by the N-terminal moiety of 8E6; in contrast, its C-terminal half was sufficient for late promoter activation. This was markedly reduced by the deletion of amino acids 132 to 136 of 8E6, which also abolished the binding to p300, indicating that a direct interaction between 8E6 and p300 is involved. Moreover, a 45-amino-acid segment within the C/H3 region of p300 is required for 8E6 to stimulate the coactivator function of p300. Our results demonstrate for the first time that an E6 oncoprotein of HPV directly contributes to the regulation of HPV gene expression.
- Published
- 2006
- Full Text
- View/download PDF
15. Viral capsid assembly as a model for protein aggregation diseases: Active processes catalyzed by cellular assembly machines comprising novel drug targets
- Author
-
Vishwanath R. Lingappa, Debendranath Dey, Rita Marreiros, Verian Bader, Carsten Korth, Suganya Selvarajah, and Andreas Müller-Schiffmann
- Subjects
Cancer Research ,Enzyme complex ,Viral capsid assembly ,Virus Assembly ,Cellular homeostasis ,Biology ,Protein aggregation ,Models, Biological ,Protein Aggregation, Pathological ,Virus ,Cell biology ,Infectious Diseases ,Stress granule ,Capsid ,Biochemistry ,Virus Diseases ,Virology ,Viruses ,Animals ,Humans ,Capsid Proteins ,Host factor - Abstract
Viruses can be conceptualized as self-replicating multiprotein assemblies, containing coding nucleic acids. Viruses have evolved to exploit host cellular components including enzymes to ensure their replicative life cycle. New findings indicate that also viral capsid proteins recruit host factors to accelerate their assembly. These assembly machines are RNA-containing multiprotein complexes whose composition is governed by allosteric sites. In the event of viral infection, the assembly machines are recruited to support the virus over the host and are modified to achieve that goal. Stress granules and processing bodies may represent collections of such assembly machines, readily visible by microscopy but biochemically labile and difficult to isolate by fractionation. We hypothesize that the assembly of protein multimers such as encountered in neurodegenerative or other protein conformational diseases, is also catalyzed by assembly machines. In the case of viral infection, the assembly machines have been modified by the virus to meet the virus' need for rapid capsid assembly rather than host homeostasis. In the case of the neurodegenerative diseases, it is the monomers and/or low n oligomers of the so-called aggregated proteins that are substrates of assembly machines. Examples for substrates are amyloid β peptide (Aβ) and tau in Alzheimer's disease, α-synuclein in Parkinson's disease, prions in the prion diseases, Disrupted-in-schizophrenia 1 (DISC1) in subsets of chronic mental illnesses, and others. A likely continuum between virus capsid assembly and cell-to-cell transmissibility of aggregated proteins is remarkable. Protein aggregation diseases may represent dysfunction and dysregulation of these assembly machines analogous to the aberrations induced by viral infection in which cellular homeostasis is pathologically reprogrammed. In this view, as for viral infection, reset of assembly machines to normal homeostasis should be the goal of protein aggregation therapeutics. A key basis for the commonality between viral and neurodegenerative disease aggregation is a broader definition of assembly as more than just simple aggregation, particularly suited for the crowded cytoplasm. The assembly machines are collections of proteins that catalytically accelerate an assembly reaction that would occur spontaneously but too slowly to be relevant in vivo. Being an enzyme complex with a functional allosteric site, appropriated for a non-physiological purpose (e.g. viral infection or conformational disease), these assembly machines present a superior pharmacological target because inhibition of their active site will amplify an effect on their substrate reaction. Here, we present this hypothesis based on recent proof-of-principle studies against Aβ assembly relevant in Alzheimer's disease.
- Published
- 2014
16. BRI2-BRICHOS is increased in human amyloid plaques in early stages of Alzheimer's disease
- Author
-
Lois-Lee Dekkers, Connie R. Jimenez, Annemieke J.M. Rozemuller, Charlotte E. Teunissen, Philip Scheltens, Marinus A. Blankenstein, Marta Del Campo, Jeroen J.M. Hoozemans, Carsten Korth, Andreas Müller-Schiffmann, Robert Veerhuis, Pathology, Neurology, Clinical chemistry, Medical oncology laboratory, NCA - Neurobiology of mental health, and NCA - neurodegeneration
- Subjects
Aging ,medicine.medical_specialty ,Pathology ,ADAM10 ,BACE1-AS ,Hippocampus ,Plaque, Amyloid ,Pathogenesis ,Amyloid beta-Protein Precursor ,Alzheimer Disease ,Internal medicine ,medicine ,Amyloid precursor protein ,Humans ,Furin ,Adaptor Proteins, Signal Transducing ,Amyloid beta-Peptides ,Membrane Glycoproteins ,biology ,General Neuroscience ,Immunohistochemistry ,Pathophysiology ,Protein Structure, Tertiary ,Endocrinology ,Multiprotein Complexes ,biology.protein ,Neurology (clinical) ,Geriatrics and Gerontology ,Protein Binding ,Developmental Biology - Abstract
BRI2 protein binds amyloid precursor protein to halt amyloid-β production and inhibits amyloid-β aggregation via its BRICHOS-domain suggesting a link between BRI2 and Alzheimer's disease (AD). Here, we investigate the possible involvement of BRI2 in human AD pathogenesis. BRI2 containing BRICHOS-domain was increased up to 3-fold in AD hippocampus (p = 0.003, n = 14/group). Immunohistochemistry showed BRI2 deposits associated with amyloid-β plaques in early pathologic stages (Braak-III; Thal-2/3). We observed a decrease in the protein levels of ADAM10 (p = 0.02) and furin (p = 0.066), as well as an increase in SPPL2b (p < 0.0001) in AD hippocampus. Because these enzymes are involved in BRI2 processing, their changes may lead to aberrant processing of BRI2 promoting its deposition and likely affecting BRI2 function. Loss of BRI2 function in AD was supported by the decreased presence of BRI2-amyloid precursor protein complexes in the hippocampus of AD patients compared with control subjects. In conclusion, our data obtained from human samples indicate that in early stages of AD there is an increased deposition of BRI2, which likely leads to impaired BRI2 function thereby influencing AD pathophysiology.
- Published
- 2014
- Full Text
- View/download PDF
17. P3–066: The increase of BRI2 in early stages of Alzheimer's disease correlates with reduced levels of ADAM10 and may prevent its binding to APP
- Author
-
Philip Scheltens, R. Veerhuis, Marinus A. Blankenstein, Jeroen J.M. Hoozemans, Andreas Müller-Schiffmann, Charlotte E. Teunissen, Marta Del Campo Milan, Connie R. Jimenez, and Carsten Korth
- Subjects
medicine.medical_specialty ,Epidemiology ,business.industry ,Health Policy ,ADAM10 ,Disease ,Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Endocrinology ,Developmental Neuroscience ,Internal medicine ,Medicine ,Neurology (clinical) ,Geriatrics and Gerontology ,business - Published
- 2013
- Full Text
- View/download PDF
18. Hybrid molecules synergistically acting against protein aggregation diseases
- Author
-
Andreas Müller-Schiffmann, Ralf Klingenstein, and Carsten Korth
- Subjects
Models, Molecular ,Protein Folding ,Molecular Structure ,PrPSc Proteins ,Chemistry ,Drug discovery ,Drug Synergism ,General Medicine ,Protein aggregation ,Small molecule ,In vitro ,Prion Diseases ,Small Molecule Libraries ,Chimera (genetics) ,Biochemistry ,In vivo ,Postsynaptic potential ,Heterocyclic Compounds ,Drug Discovery ,Excitatory postsynaptic potential ,Animals ,Humans ,Peptides - Abstract
An emerging common feature of the age-associated neurodegenerative disorders like Alzheimer's disease (AD) and Creutzfeldt-Jakob disease (CJD) is the ability of many disease-associated protein aggregates to induce conversion of a normal counterpart conformer leading to an acceleration of disease progression. Curative pharmacotherapy has not been achieved so far despite successes in elucidating pathomechanisms. Here, we review the pharmaceutical strategy of generating hybrid compounds, i.e. compounds consisting of several independently acting moieties with synergistic effects, on key molecular players in AD and CJD. For prion diseases, we review hybrid compounds consisting of two different heterocyclic compounds, their synergistic effects on prion replication in a cell culture model and their ability to prolong survival of experimentally prion-infected mice in vivo. While a combination therapy of several antiprion compounds including quinacrine, clomipramine, simvastatin and tocopherol prolonged survival time to 10-25%, administration of hybrid compound quinpramine alone, a chimera of acridine and iminodibenzyl scaffolds, led to 10% survival time extension. For AD, we review a hybrid compound consisting of an Aβ recognizing D-peptide fused to a small molecule β-sheet breaker, an aminopyrazole. This molecule was able to diminish Aβ oligomers in cell culture and significantly decrease synaptotoxicity as measured by miniature excitatory postsynaptic responses in vitro. Hybrid compounds can dramatically increase potency of their single moieties and lead to novel functions when they act in a simultaneous or sequential manner thereby revealing synergistic properties. Their systematic generation combining different classes of compounds from peptides to small molecules has the potential to significantly accelerate drug discovery.
- Published
- 2013
19. P4‐203: Expression and characterization of a novel Aβ oligomer and fibril conformer‐specific single‐chain variable fragment (scFv) that was selected using Aβ1‐16 linear sequence
- Author
-
Dieter Willbold, Carsten Korth, Andreas Müller-Schiffmann, Susanne Aileen Funke, Silke Dornieden, and Heinrich Sticht
- Subjects
Epidemiology ,Stereochemistry ,Health Policy ,Linear sequence ,Fibril ,Oligomer ,Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Developmental Neuroscience ,chemistry ,Single-chain variable fragment ,Neurology (clinical) ,Geriatrics and Gerontology ,Conformational isomerism - Published
- 2012
- Full Text
- View/download PDF
20. C-terminal fragment of N-cadherin accelerates synapse destabilization by amyloid-β
- Author
-
Katja Nieweg, Carsten Korth, Kurt Gottmann, Aksana Andreyeva, Simon D. Klapper, Katharina Horstmann, and Andreas Müller-Schiffmann
- Subjects
Gene Expression ,AMPA receptor ,Presenilin ,Synapse ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Alzheimer Disease ,Antigens, CD ,medicine ,Presenilin-1 ,Animals ,Humans ,Cells, Cultured ,030304 developmental biology ,Aged ,Aged, 80 and over ,Neurons ,0303 health sciences ,Amyloid beta-Peptides ,biology ,Cadherin ,Miniature Postsynaptic Potentials ,Dipeptides ,medicine.disease ,Cadherins ,Peptide Fragments ,Cell biology ,Mice, Inbred C57BL ,Silent synapse ,Proteolysis ,Synapses ,Excitatory postsynaptic potential ,biology.protein ,Female ,Neurology (clinical) ,Carbamates ,Alzheimer's disease ,Amyloid Precursor Protein Secretases ,Amyloid precursor protein secretase ,Neuroscience ,Protein Processing, Post-Translational ,030217 neurology & neurosurgery - Abstract
The aetiology of Alzheimer's disease is thought to include functional impairment of synapses and synapse loss as crucial pathological events leading to cognitive dysfunction and memory loss. Oligomeric amyloid-β peptides are well known to induce functional damage, destabilization and loss of brain synapses. However, the complex molecular mechanisms of amyloid-β action resulting ultimately in synapse elimination are incompletely understood, thus limiting knowledge of potential therapeutic targets. Under physiological conditions, long-term synapse stability is mediated by trans-synaptically interacting adhesion molecules such as the homophilically binding N-cadherin/catenin complexes. In this study, we addressed whether inhibition of N-cadherin function affects amyloid-β-induced synapse impairment. We found that blocking N-cadherin function, both by specific peptides interfering with homophilic binding and by expression of a dominant-negative, ectodomain-deleted N-cadherin mutant, resulted in a strong acceleration of the effect of amyloid-β on synapse function in cultured cortical neurons. The frequency of AMPA (alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptor-mediated miniature excitatory postsynaptic currents was reduced upon amyloid-β application much earlier than observed in controls. We further hypothesized that ectodomain-shed, transmembrane C-terminal fragments that are generated during N-cadherin proteolytic processing might similarly enhance amyloid-β-induced synapse damage. Indeed, expression of human N-cadherin C-terminal fragment 1 strongly accelerated amyloid-β-triggered synapse impairment. Ectodomain-shed N-cadherin C-terminal fragment 1 is further proteolytically cleaved by γ-secretase. Therefore, both pharmacological inhibition of γ-secretase and expression of the dominant-negative presenilin 1 mutant L166P were used to increase the presence of endogeneous N-cadherin C-terminal fragment 1. Under these conditions, we again found a strong acceleration of amyloid-β-induced synapse impairment, which could be compensated by over-expression of full-length N-cadherin. Intriguingly, western blot analysis of post-mortem brains from patients with Alzheimer's disease revealed an enhanced presence of N-cadherin C-terminal fragment 1. Thus, an inhibition of N-cadherin function by proteolytically generated N-cadherin C-terminal fragment 1 might play an important role in Alzheimer's disease progression by accelerating amyloid-β-triggered synapse damage.
- Published
- 2012
21. Hybrid compounds: from simple combinations to nanomachines
- Author
-
Heinrich Sticht, Carsten Korth, and Andreas Müller-Schiffmann
- Subjects
Aptamer ,Chemistry, Pharmaceutical ,Peptide ,Biology ,01 natural sciences ,03 medical and health sciences ,Molecular recognition ,Drug Discovery ,Moiety ,Humans ,Pharmacology (medical) ,Molecular Targeted Therapy ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,Pharmacology ,chemistry.chemical_classification ,0303 health sciences ,010405 organic chemistry ,General Medicine ,Small molecule ,0104 chemical sciences ,Drug Combinations ,Drug development ,chemistry ,Biochemistry ,Models, Chemical ,Nucleic acid ,Nanoparticles ,Biotechnology ,Signal Transduction - Abstract
The combination of two different and independently acting compounds into one covalently linked hybrid compound can convey synergy from the effects of both independently acting moieties to the new composite compound, leading to a pharmacological potency greater than the sum of each individual moiety's potencies. Here, we review a variety of such hybrid compounds, which can consist of various functional parts, molecular recognition or subcellular targeting moieties, or combinations thereof, acting either simultaneously or sequentially. Such moieties within a hybrid compound can consist of a variety of substance classes, including small organic molecules, polypeptides or nucleic acids identified either via rational molecular design or selection from libraries. Precedent for hybrid compounds comes from naturally occurring proteins and small molecules, such as botulinum toxin and bleomycin, which are secreted by micro-organisms. We review the high degree of suitability of hybrid compounds for the treatment of multifactorial diseases by simultaneously hitting several targets along an identified disease pathway. Examples are hybrid compounds against Alzheimer's disease, against the cancer-relevant phosphoinisitide-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) and epidermal growth factor signaling cascade, or in antimalarial therapy via simultaneous hitting of different mechanisms of hemozoin formation. Molecular recognition by peptides or aptamers (recognition-specific RNA or peptide sequences) can be combined with the transport of small molecule β-sheet breakers or toxins, or targeting to ubiquitin-dependent proteolysis. The vision of molecular nanomachines is currently realized in sequentially acting modular nanotransporters, consisting of four modules including a target, a membrane and nuclear translocation sequence, as well as a drug attachment domain. Through the rational combination of existing drugs and the synergy of their effects, a rapid amplification of their potency may be achieved, greatly accelerating drug development. A further enhancement of simultaneous multitarget action is enabled through the design of multifunctional hybrid drugs with sequential effects that make these hybrid molecules resemble intelligent nanomachines.
- Published
- 2012
22. P2‐446: Oligomeric Aβ‐targeting D‐peptides and peptide‐aminopyrazole‐hybrids for treatment of Alzheimer's disease
- Author
-
Carsten Korth, Susanne Aileen Funke, Kurt Gottmann, Dieter Willbold, Thomas Schrader, Klaus G. Reymann, Oleksandr Brener, Luitgard Nagel-Steger, Inga Kadish, Dirk Bartnik, Heinrich Sticht, Thomas van Groen, Anselm H. C. Horn, Julia Maerz-Berberich, and Andreas Müller-Schiffmann
- Subjects
chemistry.chemical_classification ,Epidemiology ,Chemistry ,Health Policy ,Peptide ,Disease ,Pharmacology ,Cleavage (embryo) ,Highly sensitive ,Psychiatry and Mental health ,Cellular and Molecular Neuroscience ,Immune system ,Cerebrospinal fluid ,Developmental Neuroscience ,Pharmacodynamics ,Toxicity ,Neurology (clinical) ,Geriatrics and Gerontology - Abstract
Background: The g-secretase complex generates the C-terminus of As from APP and thus determines the potential for oligomerization, formation and propensity for deposition. Inhibition of g-secretase activity blocks As production and is being tested as disease modifying treatment for AD. However, g-secretase inhibitors (GSIs) also block the cleavage of other g-secretase substrates such as Notch, which can lead to gastrointestinal and immune related toxicity. g-secretase modulators (GSMs), on the other hand, reduce specifically the formation of As42, without affecting the signaling functions of other g-secretase substrates. This selective reduction of As42 levels after GSM treatment is accompanied by an increase in the production of the shorter and less toxic As species. Methods: We investigate the effect of GSMs and GSIs on As levels in cerebrospinal fluid (CSF) from beagle dogs and utilize specific and sensitive immunoassays for the quantification of As species of different lengths. Results: We show that GSM treatment leads to decreased levels of As42 and increased levels of shorter As peptides in dog CSF. Our data support that quantification of shorter As peptides can be used as a pharmacodynamic marker of GSM treatment. As expected, the results show that GSIs are able to decrease the levels of all As peptides in dog CSF. Conclusions:We have produced highly sensitive and specific assays that can detect As species in dog CSF. Our data show that GSMs are able to selectively reduce the levels of the As42 toxic species and increase the levels of shorter As species, thus representing a promising approach for a disease-modifying treatment of AD.
- Published
- 2011
- Full Text
- View/download PDF
23. Pharmacological prion protein silencing accelerates central nervous system autoimmune disease via T cell receptor signalling
- Author
-
Andreas Breil, Jeffrey L. Elliott, Lawrence P. Kane, Amy E. Lovett-Racke, Todd N. Eagar, Petra D. Cravens, Carsten Korth, Olaf Stüve, Rehana Z. Hussain, Mahendra Pal Singh, Krishna Puttaparthi, Andreas Müller-Schiffmann, Benjamin Petsch, Anne R. Gocke, Lothar Stitz, Michael K. Racke, Wei Hu, Bernhard Hemmer, Li Hong Ben, S. Rutger Leliveld, and Stefan Nessler
- Subjects
Small interfering RNA ,Prions ,T cell ,animal diseases ,Receptors, Antigen, T-Cell ,Mice, Transgenic ,Demyelinating Autoimmune Diseases, CNS ,prion ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Gene Silencing ,RNA, Small Interfering ,Receptor ,030304 developmental biology ,0303 health sciences ,immunosuppression ,biology ,Effector ,Experimental autoimmune encephalomyelitis ,Original Articles ,medicine.disease ,Molecular biology ,small interfering RNA ,3. Good health ,Myelin basic protein ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,biology.protein ,Female ,Neurology (clinical) ,T cell signaling ,Signal transduction ,030215 immunology ,Signal Transduction - Abstract
The primary biological function of the endogenous cellular prion protein has remained unclear. We investigated its biological function in the generation of cellular immune responses using cellular prion protein gene-specific small interfering ribonucleic acid in vivo and in vitro. Our results were confirmed by blocking cellular prion protein with monovalent antibodies and by using cellular prion protein-deficient and -transgenic mice. In vivo prion protein gene-small interfering ribonucleic acid treatment effects were of limited duration, restricted to secondary lymphoid organs and resulted in a 70% reduction of cellular prion protein expression in leukocytes. Disruption of cellular prion protein signalling augmented antigen-specific activation and proliferation, and enhanced T cell receptor signalling, resulting in zeta-chain-associated protein-70 phosphorylation and nuclear factor of activated T cells/activator protein 1 transcriptional activity. In vivo prion protein gene-small interfering ribonucleic acid treatment promoted T cell differentiation towards pro-inflammatory phenotypes and increased survival of antigen-specific T cells. Cellular prion protein silencing with small interfering ribonucleic acid also resulted in the worsening of actively induced and adoptively transferred experimental autoimmune encephalomyelitis. Finally, treatment of myelin basic protein(1-11) T cell receptor transgenic mice with prion protein gene-small interfering ribonucleic acid resulted in spontaneous experimental autoimmune encephalomyelitis. Thus, central nervous system autoimmune disease was modulated at all stages of disease: the generation of the T cell effector response, the elicitation of T effector function and the perpetuation of cellular immune responses. Our findings indicate that cellular prion protein regulates T cell receptor-mediated T cell activation, differentiation and survival. Defects in autoimmunity are restricted to the immune system and not the central nervous system. Our data identify cellular prion protein as a regulator of cellular immunological homoeostasis and suggest cellular prion protein as a novel potential target for therapeutic immunomodulation.
- Published
- 2010
24. Complementarity determining regions of an anti-prion protein scFv fragment orchestrate conformation specificity and antiprion activity
- Author
-
Janine Muyrers, S. Rutger Leliveld, Carsten Korth, Stephan Schwarzinger, Agnieska Salwierz, Benjamin Petsch, Lothar Stitz, Detlev Riesner, Andreas Müller-Schiffmann, and Christian Mangels
- Subjects
PrPSc Proteins ,medicine.drug_class ,Protein Conformation ,animal diseases ,medicine.medical_treatment ,Immunology ,Molecular Sequence Data ,Antibody Affinity ,Peptide ,Complementarity determining region ,Monoclonal antibody ,medicine.disease_cause ,law.invention ,Cell Line ,Mice ,law ,medicine ,Bioassay ,Animals ,PrPC Proteins ,Amino Acid Sequence ,Molecular Biology ,Escherichia coli ,chemistry.chemical_classification ,Mice, Knockout ,Protease ,Chemistry ,Molecular biology ,Complementarity Determining Regions ,nervous system diseases ,Dissociation constant ,Recombinant DNA ,Peptides - Abstract
The prion protein, PrP, exists in several stable conformations, with the presence of one conformation, PrPSc, associated with transmissible neurodegenerative diseases. Targeting PrP by high-affinity ligands has been proven to be an effective way of preventing peripheral prion infections. Here, we have generated bacterially expressed single chain fragments of the variable domains (scFv) of a monoclonal antibody in Escherichia coli, originally raised against purified PrPSc that recognizes both PrPc and PrPSc. This scFv fragment had a dissociation constant (K-D) with recombinant PrP of 2 nM and cleared prions in ScN2a cells at 4 nM, as demonstrated by a mouse prion bioassay. A peptide corresponding to the complementarity determining region 3 of the heavy chain (CDR3H) selectively bound PrPSc but had lost antiprion activity. However, synthesis and application of an improved peptide mimicking side chain topology of CDR3H while exhibiting increased protease resistance, a retro-inverso D-peptide of CDR3H, still bound PrPSc and reinstated antiprion activity. We conclude that (1) scFvW226 is so far the smallest polypeptide with bioassay confirmed antiprion activity, and (2) differential conformation specificity and bioactivity can be regulated by orchestrating the participation of different CDRs. (C) 2008 Elsevier Ltd. All rights reserved
- Published
- 2008
25. Protein tyrosine phosphatase H1 is a target of the E6 oncoprotein of high-risk genital human papillomaviruses
- Author
-
Gertrud Steger, Andreas Müller-Schiffmann, Konstantin Matentzoglu, Stephanie Töpffer, and Martin Scheffner
- Subjects
Chromatin Immunoprecipitation ,Somatic cell ,Ubiquitin-Protein Ligases ,PDZ domain ,Endogeny ,Protein tyrosine phosphatase ,Biology ,Cell Line ,ddc:150 ,Virology ,Two-Hybrid System Techniques ,Humans ,Psychological repression ,chemistry.chemical_classification ,Human papillomavirus 16 ,Human papillomavirus 18 ,Protein Tyrosine Phosphatase, Non-Receptor Type 3 ,Oncogene Proteins, Viral ,Molecular biology ,Ubiquitin ligase ,DNA-Binding Proteins ,Repressor Proteins ,Enzyme ,chemistry ,Cell culture ,biology.protein ,Protein Binding - Abstract
The E6 proteins of high-risk genital human papillomaviruses (HPV), such as HPV types 16 and 18, possess a conserved C-terminal PDZ-binding motif, which mediates interaction with some cellular PDZ domain proteins. The binding of E6 usually results in their ubiquitin-mediated degradation. The ability of E6 to bind to PDZ domain proteins correlates with the oncogenic potential. Using a yeast two-hybrid system, GST pull-down experiments and coimmunoprecipitations, we identified the protein tyrosine phosphatase H1 (PTPH1/PTPN3) as a novel target of the PDZ-binding motif of E6 of HPV16 and 18. PTPH1 has been suggested to function as tumour suppressor protein, since mutational analysis revealed somatic mutations in PTPH1 in a minor fraction of various human tumours. We show here that HPV16 E6 accelerated the proteasome-mediated degradation of PTPH1, which required the binding of E6 to the cellular ubiquitin ligase E6-AP and to PTPH1. The endogenous levels of PTPH1 were particularly low in HPV-positive cervical carcinoma cell lines. The reintroduction of the E2 protein into the HPV16-positive cervical carcinoma cell line SiHa, known to lead to a sharp repression of E6 expression and to induce growth suppression, resulted in an increase of the amount of PTPH1. Our data suggest that reducing the level of PTPH1 may contribute to the oncogenic activity of high-risk genital E6 proteins.
- Published
- 2007
26. Differential Effects of Cell-derived Amyloid-beta Monomers and Dimers on Spontaneous Neuronal Network Activity
- Author
-
Carsten Korth, A. Müller-Schiffmann, Christian Lange-Asschenfeldt, Philipp Görtz, and Uwe Henning
- Subjects
biology ,Amyloid beta ,Chemistry ,Cell ,Transfection ,Small molecule ,Pathogenesis ,Psychiatry and Mental health ,medicine.anatomical_structure ,Cell culture ,mental disorders ,biology.protein ,Biophysics ,Biological neural network ,medicine ,Function (biology) - Abstract
Introduction Growing evidence suggests that soluble amyloid-beta (Abeta) peptides play a pivotal role in Alzheimer’s disease(AD) pathogenesis by mediating synaptotoxic effects particularly at early disease stages. Objectives/ Aims We quantified the effects of different order Abeta assemblies on spontaneous firing dynamics ofneuronal networks cultured on multielectrode arrays ('neurochips”) as a read-out. We used naturally secreted, stable and conformationally highly homogenous Abeta monomers, dimers, and a mixture of different low-noligomers, derived from permanently transfected cell lines. Results Abeta dimers promoted a dose-dependent suppression of overall activity and network synchrony and altered the burst structure already in the low picomolar dose range. By contrast, Abeta monomers exhibited no effect on overall activity, but only a slight effect on burst structure and a moderate effect on network synchrony. A yet different response pattern was seen for a mixture of various low-n Abeta oligomers. Thus, multiparametric assessment of electrical activity changes on neurochips revealed characteristic signatures of the network response for the different Abeta assemblies. Since alterations of Network function likely occur in initial disease stages, these results confirm the pivotal role of Abeta dimers in early AD pathogenesis. Conclusions Neurochip recordings of toxic dimeric Abeta species may serve as a valuable diagnostic read-out in early AD and may also be applicable for future testing of drugs, antibodies, or small molecules aiming at Abeta dimers.
- Published
- 2015
- Full Text
- View/download PDF
27. Molecular engineering of a secreted, highly homogeneous, and neurotoxic aβ dimer
- Author
-
Carsten Korth, Heinrich Sticht, Aksana Andreyeva, Anselm H. C. Horn, Kurt Gottmann, and Andreas Müller-Schiffmann
- Subjects
Amyloid beta-Peptides ,Physiology ,Chemistry ,Cognitive Neuroscience ,Dimer ,Mutant ,Wild type ,Computational Biology ,Cell Biology ,General Medicine ,Transfection ,Biochemistry ,In vitro ,Cell Line ,Molecular engineering ,Folding (chemistry) ,chemistry.chemical_compound ,Alzheimer Disease ,Mutation ,Humans ,Cysteine ,Dimerization ,Secretory pathway - Abstract
Aβ oligomers play a key role in the pathophysiology of Alzheimer's disease. Research into structure-function relationships of Aβ oligomers has been hampered by the lack of large amounts of homogeneous and stable material. Using computational chemistry, we designed conservative cysteine substitutions in Aβ aiming at accelerating and stabilizing assembly of Aβ dimers by an intermolecular disulfide bond without changing its folding. Molecular dynamics simulations suggested that mutants AβS8C and AβM35C exhibited structural properties similar to those of Aβ wildtype dimers. Full length, mutant APP was stably expressed in transfected cell lines to study assembly of Aβ oligomers in the physiological, secretory pathway and to avoid artifacts resulting from simultaneous in vitro oxidation and aggregation. Biochemical and neurophysiological analysis of supernatants indicated that AβS8C generated an exclusive, homogeneous, and neurotoxic dimer, whereas AβM35C assembled into dimers, tetramers, and higher oligomers. Thus, molecular engineering enabled generation of bioactive, homogeneous, and correctly processed Aβ dimers in vivo.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.