Network


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

Hotspot


Dive into the research topics where Susanne Krasemann is active.

Publication


Featured researches published by Susanne Krasemann.


Annals of Neurology | 2015

Targeting miR-155 restores abnormal microglia and attenuates disease in SOD1 mice.

Oleg Butovsky; Mark P. Jedrychowski; Ron Cialic; Susanne Krasemann; Gopal Murugaiyan; Zain Fanek; David J. Greco; Pauline M. Wu; Camille E. Doykan; Olga Kiner; Robert Lawson; Matthew P. Frosch; Nathalie Pochet; Rachid El Fatimy; Anna M. Krichevsky; Steven P. Gygi; Hans Lassmann; James D. Berry; Merit Cudkowicz; Howard L. Weiner

To investigate miR‐155 in the SOD1 mouse model and human sporadic and familial amyotrophic lateral sclerosis (ALS).


Journal of Immunological Methods | 1996

Induction of antibodies against human prion proteins (PrP) by DNA-mediated immunization of PrP00 mice

Susanne Krasemann; Martin H. Groschup; Gerhard Hunsmann; Walter Bodemer

Prion diseases are neurodegenerative disorders, affecting humans and animals. The human diseases include kuru, Creutzfeldt-Jakob disease (CJD), Gerstmann-Sträussler-Scheinker syndrome (GSS), and fatal familial insomnia (FFI). To generate monospecific antisera against human prion proteins we have immunized mice with DNA coding for different human prion proteins. We constructed immunization vectors expressing individual genotypes of either the cellular prion gene (PRNP) or mutant forms under appropriate promoters. This approach avoids the preparation of infectious material for immunization. To circumvent immunological tolerance prion protein-deficient PrP0/0 mice were used for the DNA-mediated immunization. Thereby monospecific sera were raised capable of specifically precipitating in vitro synthesized human prion proteins. With prion protein-specific peptide ELISAs, we found that antibodies are predominantly directed against the octapeptide repeat region and to a lesser extent to regions comprising the signal peptide, the neurotoxic domain or the GPI anchor. In contrast, prion gene-positive (PrP+/+) BALB/c mice immunized under the same experimental conditions as the PrP0/0 mice did not respond with antibody formation against the human prion protein. This is the first report clearly showing that immune competent prion protein-deficient mice react with a vigorous polyclonal immune response after DNA-mediated immunization with human prion gene sequences.


Journal of Biotechnology | 1999

Generation of monoclonal antibodies against prion proteins with an unconventional nucleic acid-based immunization strategy.

Susanne Krasemann; Thomas Jürgens; Walter Bodemer

Prion diseases belong to a group of neurodegenerative disorders affecting humans and animals. The human diseases include kuru, Creutzfeldt-Jakob disease (CJD), Gerstmann-Sträussler-Scheinker syndrome (GSS) and fatal familial insomnia (FFI). The pathomechanisms of the prion diseases are not yet understood. Therefore, monoclonal antibodies (mAbs) would provide valuable tools in diagnostics as well as in basic research of these diseases. In contrast to conventional strategies we have developed an immunization protocol based on nucleic acid injection into non tolerant PrP0/0-mice. DNA or RNA coding for different human prion proteins including the mutated sequences associated with CJD, GSS and FFI were injected into muscle tissue. The mice were primarily inoculated with DNA-plasmids encoding PRNP and boosted either with DNA, RNA or recombinant Semliki Forest virus (SFV) particles expressing PRNP. After hybridoma preparation, different mAbs against prion proteins were obtained and their binding behaviour was analysed by peptide-ELISA, Western blot, immunofluorescence and immunoprecipitation. Our mAbs are directed against four different linear epitopes and may also recognize discontinuous regions of the native prion protein. It could, therefore, be demonstrated that immunization of non tolerant mice with DNA and live attenuated SF virus is a valuable means to induce a broad immune response leading eventually to the generation of a panel of mAbs for basic science as well as for diagnostics.


PLOS Neglected Tropical Diseases | 2014

Evaluation of Antiviral Efficacy of Ribavirin, Arbidol, and T-705 (Favipiravir) in a Mouse Model for Crimean-Congo Hemorrhagic Fever

Lisa Oestereich; Toni Rieger; Melanie Neumann; Christian Bernreuther; Maria Lehmann; Susanne Krasemann; Stephanie Wurr; Petra Emmerich; Xavier de Lamballerie; Stephan Ölschläger; Stephan Günther

Background Mice lacking the type I interferon receptor (IFNAR−/− mice) reproduce relevant aspects of Crimean-Congo hemorrhagic fever (CCHF) in humans, including liver damage. We aimed at characterizing the liver pathology in CCHF virus-infected IFNAR−/− mice by immunohistochemistry and employed the model to evaluate the antiviral efficacy of ribavirin, arbidol, and T-705 against CCHF virus. Methodology/Principal Findings CCHF virus-infected IFNAR−/− mice died 2–6 days post infection with elevated aminotransferase levels and high virus titers in blood and organs. Main pathological alteration was acute hepatitis with extensive bridging necrosis, reactive hepatocyte proliferation, and mild to moderate inflammatory response with monocyte/macrophage activation. Virus-infected and apoptotic hepatocytes clustered in the necrotic areas. Ribavirin, arbidol, and T-705 suppressed virus replication in vitro by ≥3 log units (IC50 0.6–2.8 µg/ml; IC90 1.2–4.7 µg/ml). Ribavirin [100 mg/(kg×d)] did not increase the survival rate of IFNAR−/− mice, but prolonged the time to death (p<0.001) and reduced the aminotransferase levels and the virus titers. Arbidol [150 mg/(kg×d)] had no efficacy in vivo. Animals treated with T-705 at 1 h [15, 30, and 300 mg/(kg×d)] or up to 2 days [300 mg/(kg×d)] post infection survived, showed no signs of disease, and had no virus in blood and organs. Co-administration of ribavirin and T-705 yielded beneficial rather than adverse effects. Conclusions/Significance Activated hepatic macrophages and monocyte-derived cells may play a role in the proinflammatory cytokine response in CCHF. Clustering of infected hepatocytes in necrotic areas without marked inflammation suggests viral cytopathic effects. T-705 is highly potent against CCHF virus in vitro and in vivo. Its in vivo efficacy exceeds that of the current standard drug for treatment of CCHF, ribavirin.


Brain | 2014

High molecular mass assemblies of amyloid-β oligomers bind prion protein in patients with Alzheimer’s disease

Frank Dohler; Diego Sepulveda-Falla; Susanne Krasemann; Hermann Altmeppen; Hartmut Schlüter; Diana Hildebrand; Inga Zerr; Jakob Matschke; Markus Glatzel

Alzheimers disease is the most common form of dementia and the generation of oligomeric species of amyloid-β is causal to the initiation and progression of it. Amyloid-β oligomers bind to the N-terminus of plasma membrane-bound cellular prion protein (PrP(C)) initiating a series of events leading to synaptic degeneration. Composition of bound amyloid-β oligomers, binding regions within PrP(C), binding affinities and modifiers of this interaction have been almost exclusively studied in cell culture or murine models of Alzheimers disease and our knowledge on PrP(C)-amyloid-β interaction in patients with Alzheimers disease is limited regarding occurrence, binding regions in PrP(C), and size of bound amyloid-β oligomers. Here we employed a PrP(C)-amyloid-β binding assay and size exclusion chromatography on neuropathologically characterized Alzheimers disease and non-demented control brains (n = 15, seven female, eight male, average age: 79.2 years for Alzheimers disease and n = 10, three female, seven male, average age: 66.4 years for controls) to investigate amyloid-β-PrP(C) interaction. PrP(C)-amyloid-β binding always occurred in Alzheimers disease brains and was never detected in non-demented controls. Neither expression level of PrP(C) nor known genetic modifiers of Alzheimers disease, such as the PrP(C) codon 129 polymorphism, influenced this interaction. In Alzheimers disease brains, binding of amyloid-β to PrP(C) occurred via the PrP(C) N-terminus. For synthetic amyloid-β42, small oligomeric species showed prominent binding to PrP(C), whereas in Alzheimers disease brains larger protein assemblies containing amyloid-β42 bound efficiently to PrP(C). These data confirm Alzheimers disease specificity of binding of amyloid-β to PrP(C) via its N-terminus in a large cohort of Alzheimers disease/control brains. Differences in sizes of separated protein fractions between synthetic and brain-derived amyloid-β binding to PrP(C) suggest that larger assemblies of amyloid-β or additional non-amyloid-β components may play a role in binding of amyloid-β42 to PrP(C) in Alzheimers disease.


Journal of Neurochemistry | 2016

Exosomal cellular prion protein drives fibrillization of amyloid beta and counteracts amyloid beta-mediated neurotoxicity.

Clemens Falker; Alexander Hartmann; Inga Guett; Frank Dohler; Hermann Altmeppen; Christian Betzel; Robin Schubert; Dana Thurm; Florian Wegwitz; Pooja Joshi; Claudia Verderio; Susanne Krasemann; Markus Glatzel

Alzheimers disease is a common neurodegenerative, progressive, and fatal disorder. Generation and deposition of amyloid beta (Aβ) peptides associate with its pathogenesis and small soluble Aβ oligomers show the most pronounced neurotoxic effects and correlate with disease initiation and progression. Recent findings showed that Aβ oligomers bind to the cellular prion protein (PrPC) eliciting neurotoxic effects. The role of exosomes, small extracellular vesicles of endosomal origin, in Alzheimers disease is only poorly understood. Besides serving as disease biomarkers they may promote Aβ plaque formation, decrease Aβ‐mediated synaptotoxicity, and enhance Aβ clearance. Here, we explore how exosomal PrPC connects to protective functions attributed to exosomes in Alzheimers disease. To achieve this, we generated a mouse neuroblastoma PrPC knockout cell line using transcription activator‐like effector nucleases. Using these, as well as SH‐SY5Y human neuroblastoma cells, we show that PrPC is highly enriched on exosomes and that exosomes bind amyloid beta via PrPC. Exosomes showed highest binding affinity for dimeric, pentameric, and oligomeric Aβ species. Thioflavin T assays revealed that exosomal PrPC accelerates fibrillization of amyloid beta, thereby reducing neurotoxic effects imparted by oligomeric Aβ. Our study provides further evidence for a protective role of exosomes in Aβ‐mediated neurodegeneration and highlights the importance of exosomal PrPC in molecular mechanisms of Alzheimers disease.


EMBO Reports | 2017

TREM2 deficiency impairs chemotaxis and microglial responses to neuronal injury

F. Mazaheri; Nicolas Snaidero; Gernot Kleinberger; Charlotte Madore; Anna Daria; Georg Werner; Susanne Krasemann; Anja Capell; Dietrich Trümbach; Wolfgang Wurst; Bettina Brunner; Sebastian Bultmann; Sabina Tahirovic; Martin Kerschensteiner; Thomas Misgeld; Oleg Butovsky; Christian Haass

Sequence variations in the triggering receptor expressed on myeloid cells 2 (TREM2) have been linked to an increased risk for neurodegenerative disorders such as Alzheimers disease and frontotemporal lobar degeneration. In the brain, TREM2 is predominantly expressed in microglia. Several disease‐associated TREM2 variants result in a loss of function by reducing microglial phagocytosis, impairing lipid sensing, preventing binding of lipoproteins and affecting shielding of amyloid plaques. We here investigate the consequences of TREM2 loss of function on the microglia transcriptome. Among the differentially expressed messenger RNAs in wild‐type and Trem2−/− microglia, gene clusters are identified which represent gene functions in chemotaxis, migration and mobility. Functional analyses confirm that loss of TREM2 impairs appropriate microglial responses to injury and signals that normally evoke chemotaxis on multiple levels. In an ex vivo organotypic brain slice assay, absence of TREM2 reduces the distance migrated by microglia. Moreover, migration towards defined chemo‐attractants is reduced upon ablation of TREM2 and can be rescued by TREM2 re‐expression. In vivo, microglia lacking TREM2 migrate less towards injected apoptotic neurons, and outgrowth of microglial processes towards sites of laser‐induced focal CNS damage in the somatosensory cortex is slowed. The apparent lack of chemotactic stimulation upon depletion of TREM2 is consistent with a stable expression profile of genes characterizing the homoeostatic signature of microglia.


FEBS Journal | 2013

Roles of endoproteolytic α‐cleavage and shedding of the prion protein in neurodegeneration

Hermann Altmeppen; Johannes Prox; Berta Puig; Frank Dohler; Clemens Falker; Susanne Krasemann; Markus Glatzel

The cellular prion protein (PrPC) plays important roles in neurodegenerative diseases. First, it is the well‐established substrate for the conformational conversion into its pathogenic isoform (PrPSc) giving rise to progressive and fatal prion diseases. Moreover, several recent reports highlight important roles of PrPC in other neurodegenerative conditions such as Alzheimers disease. Since PrPC is subject to proteolytic processing, here we discuss the two main cleavage events under physiological conditions, α‐cleavage and shedding. We focus on how these cleavages and the resulting fragments may impact prion diseases as well as other neurodegenerative proteinopathies. Finally, we discuss the recently identified sheddase of PrPC, namely the metalloprotease ADAM10, with regard to therapeutic potential against neurodegenerative diseases.


eLife | 2015

The sheddase ADAM10 is a potent modulator of prion disease

Hermann Altmeppen; Johannes Prox; Susanne Krasemann; Berta Puig; Katharina Kruszewski; Frank Dohler; Christian Bernreuther; Ana Hoxha; Luise Linsenmeier; Beata Sikorska; Pawel P. Liberski; Udo Bartsch; Paul Saftig; Markus Glatzel

The prion protein (PrPC) is highly expressed in the nervous system and critically involved in prion diseases where it misfolds into pathogenic PrPSc. Moreover, it has been suggested as a receptor mediating neurotoxicity in common neurodegenerative proteinopathies such as Alzheimers disease. PrPC is shed at the plasma membrane by the metalloprotease ADAM10, yet the impact of this on prion disease remains enigmatic. Employing conditional knockout mice, we show that depletion of ADAM10 in forebrain neurons leads to posttranslational increase of PrPC levels. Upon prion infection of these mice, clinical, biochemical, and morphological data reveal that lack of ADAM10 significantly reduces incubation times and increases PrPSc formation. In contrast, spatiotemporal analysis indicates that absence of shedding impairs spread of prion pathology. Our data support a dual role for ADAM10-mediated shedding and highlight the role of proteolytic processing in prion disease. DOI: http://dx.doi.org/10.7554/eLife.04260.001


The Journal of Infectious Diseases | 2016

Efficacy of Favipiravir Alone and in Combination With Ribavirin in a Lethal, Immunocompetent Mouse Model of Lassa Fever

Lisa Oestereich; Toni Rieger; Anja Lüdtke; Paula Ruibal; Stephanie Wurr; Elisa Pallasch; Sabrina Bockholt; Susanne Krasemann; César Muñoz-Fontela; Stephan Günther

We studied the therapeutic potential of favipiravir (T-705) for Lassa fever, both alone and in combination with ribavirin. Favipiravir suppressed Lassa virus replication in cell culture by 5 log10 units. In a novel lethal mouse model, it lowered the viremia level and the virus load in organs and normalized levels of cell-damage markers. Treatment with 300 mg/kg per day, commenced 4 days after infection, when the viremia level had reached 4 log10 virus particles/mL, rescued 100% of Lassa virus–infected mice. We found a synergistic interaction between favipiravir and ribavirin in vitro and an increased survival rate and extended survival time when combining suboptimal doses in vivo.

Collaboration


Dive into the Susanne Krasemann's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Inga Zerr

German Center for Neurodegenerative Diseases

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

César Muñoz-Fontela

Bernhard Nocht Institute for Tropical Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge