22 results on '"Müller-Schiffmann, A."'
Search Results
2. Soluble amyloid‐β dimers are resistant to amyloid‐β prion conversion in vivo suggesting antiprion properties.
- Author
-
van Gerresheim, Else F., Müller‐Schiffmann, Andreas, Schäble, Sandra, Koopmans, Bastijn, Loos, Maarten, and Korth, Carsten
- Published
- 2023
- Full Text
- View/download PDF
3. The interaction of insoluble Amyloid‐β with soluble Amyloid‐β dimers decreases Amyloid‐β plaque numbers.
- Author
-
Gerresheim, Else F., Herring, Arne, Gremer, Lothar, Müller‐Schiffmann, Andreas, Keyvani, Kathy, and Korth, Carsten
- Subjects
DIMERS ,ALZHEIMER'S patients ,TRANSGENIC mice ,OLIGOMERS ,MOLECULAR interactions - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
4. SARS‐CoV‐2 targets neurons of 3D human brain organoids.
- Author
-
Ramani, Anand, Müller, Lisa, Ostermann, Philipp N, Gabriel, Elke, Abida‐Islam, Pranty, Müller‐Schiffmann, Andreas, Mariappan, Aruljothi, Goureau, Olivier, Gruell, Henning, Walker, Andreas, Andrée, Marcel, Hauka, Sandra, Houwaart, Torsten, Dilthey, Alexander, Wohlgemuth, Kai, Omran, Heymut, Klein, Florian, Wieczorek, Dagmar, Adams, Ortwin, and Timm, Jörg
- Subjects
SARS-CoV-2 ,COVID-19 pandemic ,NEURONS ,VIRAL tropism ,COVID-19 ,NEUROTOXICOLOGY ,SYMPTOMS - Abstract
COVID‐19 pandemic caused by SARS‐CoV‐2 infection is a public health emergency. COVID‐19 typically exhibits respiratory illness. Unexpectedly, emerging clinical reports indicate that neurological symptoms continue to rise, suggesting detrimental effects of SARS‐CoV‐2 on the central nervous system (CNS). Here, we show that a Düsseldorf isolate of SARS‐CoV‐2 enters 3D human brain organoids within 2 days of exposure. We identified that SARS‐CoV‐2 preferably targets neurons of brain organoids. Imaging neurons of organoids reveal that SARS‐CoV‐2 exposure is associated with altered distribution of Tau from axons to soma, hyperphosphorylation, and apparent neuronal death. Our studies, therefore, provide initial insights into the potential neurotoxic effect of SARS‐CoV‐2 and emphasize that brain organoids could model CNS pathologies of COVID‐19. Synopsis: Modelling coronavirus exposure of the central nervous system is critical to assess the cellular tropism and potential neurological consequences of infection. Here, a Düsseldorf isolate of SARS‐CoV‐2 is shown to enter human cerebral organoids and preferably target neuronal cells. Clinical SARS‐CoV-2 strain targets neurons of 3D human brain organoids.SARS‐CoV-2 does not appear to actively proliferate in neurons.SARS‐CoV-2 is associated with Tau abnormalities in neurons.SARS‐CoV-2 induces neuronal cell death. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
5. Disruption of cellular proteostasis by H1N1 influenza A virus causes α-synuclein aggregation.
- Author
-
Marreiros, Rita, Müller-Schiffmann, Andreas, Trossbach, Svenja V., Prikulis, Ingrid, Hänsch, Sebastian, Weidtkamp-Peters, Stefanie, Moreira, Ana Raquel, Sahu, Shriya, Soloviev, Irina, Selvarajah, Suganya, Lingappa, Vishwanath R., and Korth, Carsten
- Subjects
H1N1 influenza ,INFLUENZA A virus, H1N1 subtype ,INFLUENZA A virus ,PATHOLOGY ,KNOCKOUT mice - 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 a-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 a-synuclein aggregation. As a cell pathobiological mechanism, we identified H1N1-induced blocking of autophagosome formation and inhibition of autophagic flux. In addition, a-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 a-synuclein or DISC1 thatmay 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. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
6. Biophysical insights from a single chain camelid antibody directed against the Disrupted-in-Schizophrenia 1 protein.
- Author
-
Yerabham, Antony S. K., Müller-Schiffmann, Andreas, Ziehm, Tamar, Stadler, Andreas, Köber, Sabrina, Indurkhya, Xela, Marreiros, Rita, Trossbach, Svenja V., Bradshaw, Nicholas J., Prikulis, Ingrid, Willbold, Dieter, Weiergräber, Oliver H., and Korth, Carsten
- Subjects
NEURAL development ,GENETIC polymorphisms ,PROTEIN-protein interactions ,SURFACE plasmon resonance ,MOLECULAR models - Abstract
Accumulating evidence suggests an important role for the Disrupted-in-Schizophrenia 1 (DISC1) protein in neurodevelopment and chronic mental illness. In particular, the C-terminal 300 amino acids of DISC1 have been found to mediate important protein-protein interactions and to harbor functionally important phosphorylation sites and disease-associated polymorphisms. However, long disordered regions and oligomer-forming subdomains have so far impeded structural analysis. V
H H domains derived from camelid heavy chain only antibodies are minimal antigen binding modules with appreciable solubility and stability, which makes them well suited for the stabilizing proteins prior to structural investigation. Here, we report on the generation of a VH H domain derived from an immunized Lama glama, displaying high affinity for the human DISC1 C region (aa 691–836), and its characterization by surface plasmon resonance, size exclusion chromatography and immunological techniques. The VH H-DISC1 (C region) complex was also used for structural investigation by small angle X-ray scattering analysis. In combination with molecular modeling, these data support predictions regarding the three-dimensional fold of this DISC1 segment as well as its steric arrangement in complex with our VH H antibody. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
7. Amyloid-β dimers in the absence of plaque pathology impair learning and synaptic plasticity.
- Author
-
Müller-Schiffmann, Andreas, Herring, Arne, Abdel-Hafiz, Laila, Chepkova, Aisa N., Schäble, Sandra, Wedel, Diana, Horn, Anselm H. C., Sticht, Heinrich, de Souza Silva, Maria A., Gottmann, Kurt, Sergeeva, Olga A., Huston, Joseph P., Keyvani, Kathy, and Korth, Carsten
- Subjects
AMYLOID ,DIMERS ,AMYLOID plaque ,GENETICS of Alzheimer's disease ,CASCADES (Fluid dynamics) - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
8. BRI2 ectodomain affects Aβ fibrillation and tau truncation in human neuroblastoma cells.
- Author
-
Campo, M., Oliveira, C., Scheper, W., Zwart, R., Korth, C., Müller-Schiffmann, A., Kostallas, G., Biverstal, H., Presto, J., Johansson, J., Hoozemans, J., Pereira, C., and Teunissen, C.
- Subjects
MEMBRANE proteins ,ATRIAL fibrillation ,TAU proteins ,NEUROBLASTOMA ,ALZHEIMER'S disease ,AMYLOID beta-protein ,PHOSPHORYLATION - Abstract
Alzheimer's disease (AD) is pathologically characterized by the presence of misfolded proteins such as amyloid beta (Aβ) in senile plaques, and hyperphosphorylated tau and truncated tau in neurofibrillary tangles (NFT). The BRI2 protein inhibits Aβ aggregation via its BRICHOS domain and regulates critical proteins involved in initiating the amyloid cascade, which has been hypothesized to be central in AD pathogenesis. We recently detected the deposition of BRI2 ectodomain associated with Aβ plaques and concomitant changes in its processing enzymes in early stages of AD. Here, we aimed to investigate the effects of recombinant BRI2 ectodomain (rBRI2) on Aβ aggregation and on important molecular pathways involved in early stages of AD, including the unfolded protein response (UPR), phosphorylation and truncation of tau, as well as apoptosis. We found that rBRI2 delays Aβ fibril formation, although less efficiently than the BRI2 BRICHOS domain (BRI2 residues 113-231). In human neuroblastoma SH-SY5Y cells, rBRI2 slightly decreased cell viability and increased up to two-fold the Bax/Bcl-2 ratio and the subsequent activity of caspases 3 and 9, indicating activation of apoptosis. rBRI2 upregulated the chaperone BiP but did not modify the mRNA expression of other UPR markers (CHOP and Xbp-1). Strikingly, rBRI2 induced the activation of GSK3β but not the phosphorylation of tau. However, exposure to rBRI2 significantly induced the truncation of tau, indicating that BRI2 ectodomain can contribute to NFT formation. Since BRI2 can also regulate the metabolism of Aβ, the current data suggests that BRI2 ectodomain is a potential nexus between Aβ, tau pathology and neurodegeneration. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
9. Hybridization of an Aβ-specific antibody fragment with aminopyrazole-based β-sheet ligands displays striking enhancement of target affinity.
- Author
-
Hellmert, Marco, Müller-Schiffmann, Andreas, Peters, Max Sena, Korth, Carsten, and Schrader, Thomas
- Published
- 2015
- Full Text
- View/download PDF
10. Characterization of a Single-Chain Variable Fragment Recognizing a Linear Epitope of Aβ: A Biotechnical Tool for Studies on Alzheimer’s Disease?
- Author
-
Dornieden, Silke, Müller-Schiffmann, Andreas, Sticht, Heinrich, Jiang, Nan, Cinar, Yeliz, Wördehoff, Michael, Korth, Carsten, Funke, Susanne Aileen, and Willbold, Dieter
- Subjects
ALZHEIMER'S disease ,EPITOPES ,DISEASE progression ,NEURODEGENERATION ,IMMUNOTHERAPY ,NEUROANATOMY ,CLINICAL trials - Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder with devastating effects. Currently, therapeutic options are limited to symptomatic treatment. For more than a decade, research focused on immunotherapy for the causal treatment of AD. However, clinical trials with active immunization using Aβ encountered severe complications, for example meningoencephalitis. Consequently, attention focused on passive immunization using antibodies. As an alternative to large immunoglobulins (IgGs), Aβ binding single-chain variable fragments (scFvs) were used for diagnostic and therapeutic research approaches. scFvs can be expressed in E. coli and may provide improved pharmacokinetic properties like increased blood-brain barrier permeability or reduced side-effects in vivo. In this study, we constructed an scFv from an Aβ binding IgG, designated IC16, which binds the N-terminal region of Aβ (Aβ(1-8)). scFv-IC16 was expressed in E. coli, purified and characterized with respect to its interaction with different Aβ species and its influence on Aβ fibril formation. We were able to show that scFv-IC16 strongly influenced the aggregation behavior of Aβ and could be applied as an Aβ detection probe for plaque staining in the brains of transgenic AD model mice. The results indicate potential for therapy and diagnosis of AD. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
11. C-terminal fragment of N-cadherin accelerates synapse destabilization by amyloid-[beta].
- Author
-
Andreyeva A, Nieweg K, Horstmann K, Klapper S, Müller-Schiffmann A, Korth C, and Gottmann K
- Published
- 2012
12. C-terminal fragment of N-cadherin accelerates synapse destabilization by amyloid-β.
- Author
-
Andreyeva, Aksana, Nieweg, Katja, Horstmann, Katharina, Klapper, Simon, Müller-Schiffmann, Andreas, Korth, Carsten, and Gottmann, Kurt
- Subjects
ETIOLOGY of Alzheimer's disease ,CADHERINS ,SYNAPSES ,AMYLOID beta-protein ,COGNITION disorders ,MEMORY loss ,SECRETASE inhibitors ,WESTERN immunoblotting - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
13. Cellular prion protein participates in amyloid-β transcytosis across the blood-brain barrier.
- Author
-
Pflanzner, Thorsten, Petsch, Benjamin, André-Dohmen, Bettina, Müller-Schiffmann, Andreas, Tschickardt, Sabrina, Weggen, Sascha, Stitz, Lothar, Korth, Carsten, and Pietrzik, Claus U
- Subjects
PRIONS ,AMYLOID beta-protein ,TRANSCYTOSIS ,BLOOD-brain barrier ,NEUROTOXICOLOGY ,ALZHEIMER'S disease - 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 (PrP
c ), 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, [125 I]-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. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
14. Hybrid Compounds.
- Author
-
Müller-Schiffmann, Andreas, Sticht, Heinrich, and Korth, Carsten
- Subjects
PHARMACOLOGY ,EPIDERMAL growth factor ,ALZHEIMER'S disease ,RAPAMYCIN ,PEPTIDES ,BLEOMYCIN ,BOTULINUM toxin - 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. [ABSTRACT FROM AUTHOR]
- Published
- 2012
15. Combining Independent Drug Classes into Superior, Synergistically Acting Hybrid Molecules.
- Author
-
Müller-Schiffmann, Andreas, März-Berberich, Julia, Andreyeva, Aksana, Rönicke, Raik, Bartnik, Dirk, Brener, Oleksandr, Kutzsche, Janine, Horn, Anselm H. C., Hellmert, Marco, Polkowska, Jolanta, Gottmann, Kurt, Reymann, Klaus G., Funke, S. Aileen, Nagel-Steger, Luitgart, Moriscot, Christine, Schoehn, Guy, Sticht, Heinrich, Willbold, Dieter, Schrader, Thomas, and Korth, Carsten
- Published
- 2010
- Full Text
- View/download PDF
16. Gesteigerte Wirksamkeit durch Synergismus: Verknüpfung unabhängiger Wirkstoffklassen zu Hybridsubstanzen.
- Author
-
Müller-Schiffmann, Andreas, März-Berberich, Julia, Andreyeva, Aksana, Rönicke, Raik, Bartnik, Dirk, Brener, Oleksandr, Kutzsche, Janine, Horn, Anselm H. C., Hellmert, Marco, Polkowska, Jolanta, Gottmann, Kurt, Reymann, Klaus G., Funke, S. Aileen, Nagel-Steger, Luitgart, Moriscot, Christine, Schoehn, Guy, Sticht, Heinrich, Willbold, Dieter, Schrader, Thomas, and Korth, Carsten
- Published
- 2010
- Full Text
- View/download PDF
17. Pharmacological prion protein silencing accelerates central nervous system autoimmune disease via T cell receptor signalling.
- Author
-
Wei Hu, Nessler, Stefan, Hemmer, Bernhard, Eagar, Todd N., Kane, Lawrence P., Leliveld, S. Rutger, Müller-Schiffmann, Andreas, Gocke, Anne R., Lovett-Racke, Amy, Li-Hong Ben, Hussain, Rehana Z., Breil, Andreas, Elliott, Jeffrey L., Puttaparthi, Krishna, Cravens, Petra D., Singh, Mahendra P., Petsch, Benjamin, Stitz, Lothar, Racke, Michael K., and Korth, Carsten
- Subjects
PRIONS ,CENTRAL nervous system ,AUTOIMMUNE diseases ,T cell receptors ,IMMUNOSUPPRESSION - 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 protein1–11 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. [ABSTRACT FROM PUBLISHER]
- Published
- 2010
- Full Text
- View/download PDF
18. Vaccine Approaches to Prevent and Treat Prion Infection: Progress and Challenges.
- Author
-
Müller-Schiffmann, Andreas and Korth, Carsten
- Subjects
VACCINES ,PRION diseases ,PROTEINS ,IMMUNIZATION ,PREVENTIVE medicine - Abstract
Prion diseases are transmissible neurodegenerative diseases of humans and animals. The prion agent consists of a misfolded protein, PrP (prion protein, scrapie form), of a glycosylphosphatidylinositol-anchored host protein, PrP (PrP cellular form) of unknown function. During prion replication, PrP induces host PrP to adopt its pathogenic conformation. Some PrP 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 PrP and PrP, 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 PrP 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. [ABSTRACT FROM AUTHOR]
- Published
- 2008
19. IPSC-Derived Neuronal Cultures Carrying the Alzheimer's Disease Associated TREM2 R47H Variant Enables the Construction of an Aβ-Induced Gene Regulatory Network.
- Author
-
Martins, Soraia, Müller-Schiffmann, Andreas, Erichsen, Lars, Bohndorf, Martina, Wruck, Wasco, Sleegers, Kristel, Van Broeckhoven, Christine, Korth, Carsten, and Adjaye, James
- Subjects
ALZHEIMER'S disease ,ENDOPLASMIC reticulum ,INDUCED pluripotent stem cells ,GENE regulatory networks ,GENE expression profiling - Abstract
Genes associated with immune response and inflammation have been identified as genetic risk factors for late-onset Alzheimer´s disease (LOAD). The rare R47H variant within triggering receptor expressed on myeloid cells 2 (TREM2) has been shown to increase the risk for developing Alzheimer's disease (AD) 2–3-fold. Here, we report the generation and characterization of a model of late-onset Alzheimer's disease (LOAD) using lymphoblast-derived induced pluripotent stem cells (iPSCs) from patients carrying the TREM2 R47H mutation, as well as from control individuals without dementia. All iPSCs efficiently differentiated into mature neuronal cultures, however AD neuronal cultures showed a distinct gene expression profile. Furthermore, manipulation of the iPSC-derived neuronal cultures with an Aβ-S8C dimer highlighted metabolic pathways, phagosome and immune response as the most perturbed pathways in AD neuronal cultures. Through the construction of an Aβ-induced gene regulatory network, we were able to identify an Aβ signature linked to protein processing in the endoplasmic reticulum (ER), which emphasized ER-stress, as a potential causal role in LOAD. Overall, this study has shown that our AD-iPSC based model can be used for in-depth studies to better understand the molecular mechanisms underlying the etiology of LOAD and provides new opportunities for screening of potential therapeutic targets. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
20. P.48: Abeta dimers are sufficient to cause cognitive impairments in the absence of plaques.
- Author
-
Müller-Schiffmann, Andreas, Herring, Arne, Abdel-Hafiz, Laila, Schäble, Sandra, Wedel, Diana, van Stegen, Bernd, Horn, Anselm H. C., Sticht, Heinrich, Gottmann, Kurt, de Souza Silva, Maria Angelica, Keyvani, Kathy, and Korth, Carsten
- Published
- 2014
21. 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
-
Funke, Susanne, Dornieden, Silke, Müller-Schiffmann, Andreas, Sticht, Heinrich, Korth, Carsten, and Willbold, Dieter
- Published
- 2012
- Full Text
- View/download PDF
22. Oligomeric Aβ-targeting D-peptides and peptide-aminopyrazole-hybrids for treatment of Alzheimer's disease
- Author
-
Bartnik, Dirk, van Groen, Thomas, Kadish, Inga, Nagel-Steger, Luitgard, Maerz-Berberich, Julia, Müller-Schiffmann, Andreas, Brener, Oleksandr, Horn, Anselm H., Reymann, Klaus, Gottmann, Kurt, Schrader, Thomas, Sticht, Heinrich, Korth, Carsten, Funke, Susanne, and Willbold, Dieter
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.