Network


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

Hotspot


Dive into the research topics where Severin Filser is active.

Publication


Featured researches published by Severin Filser.


PLOS ONE | 2012

Long-Term In Vivo Imaging of Fibrillar Tau in the Retina of P301S Transgenic Mice

Christian Schön; Nadine Hoffmann; Simon M. Ochs; Steffen Burgold; Severin Filser; Sonja Steinbach; Mathias W. Seeliger; Thomas Arzberger; Michel Goedert; Hans A. Kretzschmar; Boris Schmidt; Jochen Herms

Tauopathies are widespread neurodegenerative disorders characterised by the intracellular accumulation of hyperphosphorylated tau. Especially in Alzheimers disease, pathological alterations in the retina are discussed as potential biomarkers to improve early diagnosis of the disease. Using mice expressing human mutant P301S tau, we demonstrate for the first time a straightforward optical approach for the in vivo detection of fibrillar tau in the retina. Longitudinal examinations of individual animals revealed the fate of single cells containing fibrillar tau and the progression of tau pathology over several months. This technique is most suitable to monitor therapeutic interventions aimed at reducing the accumulation of fibrillar tau. In order to evaluate if this approach can be translated to human diagnosis, we tried to detect fibrillar protein aggregates in the post-mortem retinas of patients that had suffered from Alzheimers disease or Progressive Supranuclear Palsy. Even though we could detect hyperphosphorylated tau, we did not observe any fibrillar tau or Aß aggregates. In contradiction to previous studies, our observations do not support the notion that Aβ or tau in the retina are of diagnostic value in Alzheimers disease.


Biological Psychiatry | 2015

Pharmacological Inhibition of BACE1 Impairs Synaptic Plasticity and Cognitive Functions

Severin Filser; Saak V. Ovsepian; Mercè Masana; Lidia Blazquez-Llorca; Anders Brandt Elvang; Christiane Volbracht; Marianne B. Müller; Christian K.E. Jung; Jochen Herms

BACKGROUND BACE1 (beta site amyloid precursor protein cleaving enzyme 1) is the rate limiting protease in amyloid β production, hence a promising drug target for the treatment of Alzheimers disease. Inhibition of BACE1, as the major β-secretase in vivo with multiple substrates, however is likely to have mechanism-based adverse effects. We explored the impact of long-term pharmacological inhibition of BACE1 on dendritic spine dynamics, synaptic functions, and cognitive performance of adult mice. METHODS Sandwich enzyme-linked immunosorbent assay was used to assess Aβ40 levels in brain and plasma after oral administration of BACE1 inhibitors SCH1682496 or LY2811376. In vivo two-photon microscopy of the somatosensory cortex was performed to monitor structural dynamics of dendritic spines while synaptic functions and plasticity were measured via electrophysiological recordings of excitatory postsynaptic currents and hippocampal long-term potentiation in brain slices. Finally, behavioral tests were performed to analyze the impact of pharmacological inhibition of BACE1 on cognitive performance. RESULTS Dose-dependent decrease of Aβ40 levels in vivo confirmed suppression of BACE1 activity by both inhibitors. Prolonged treatment caused a reduction in spine formation of layer V pyramidal neurons, which recovered after withdrawal of inhibitors. Congruently, the rate of spontaneous and miniature excitatory postsynaptic currents in pyramidal neurons and hippocampal long-term potentiation were reduced in animals treated with BACE1 inhibitors. These effects were not detected in Bace1(-/-) mice treated with SCH1682496, confirming BACE1 as the pharmacological target. Described structural and functional changes were associated with cognitive deficits as revealed in behavioral tests. CONCLUSIONS Our findings indicate important functions to BACE1 in structural and functional synaptic plasticity in the mature brain, with implications for cognition.


Molecular Psychiatry | 2015

Loss of neuronal GSK3β reduces dendritic spine stability and attenuates excitatory synaptic transmission via β-catenin

Simon M. Ochs; Mario M. Dorostkar; G Aramuni; Christian Schön; Severin Filser; Julia Pöschl; Anneke Kremer; F. Van Leuven; Saak V. Ovsepian; Jochen Herms

Central nervous glycogen synthase kinase 3β (GSK3β) is implicated in a number of neuropsychiatric diseases, such as bipolar disorder, depression, schizophrenia, fragile X syndrome or anxiety disorder. Many drugs employed to treat these conditions inhibit GSK3β either directly or indirectly. We studied how conditional knockout of GSK3β affected structural synaptic plasticity. Deletion of the GSK3β gene in a subset of cortical and hippocampal neurons in adult mice led to reduced spine density. In vivo imaging revealed that this was caused by a loss of persistent spines, whereas stabilization of newly formed spines was reduced. In electrophysiological recordings, these structural alterations correlated with a considerable drop in the frequency and amplitude of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor-dependent miniature excitatory postsynaptic currents. Expression of constitutively active β-catenin caused reduction in spine density and electrophysiological alterations similar to GSK3β knockout, suggesting that the effects of GSK3β knockout were mediated by the accumulation of β-catenin. In summary, changes of dendritic spines, both in quantity and in morphology, are correlates of experience-dependent synaptic plasticity; thus, these results may help explain the mechanism of action of psychotropic drugs inhibiting GSK3β.


Brain | 2014

Immunotherapy alleviates amyloid-associated synaptic pathology in an Alzheimer’s disease mouse model

Mario M. Dorostkar; Steffen Burgold; Severin Filser; Stefan Barghorn; Boris Schmidt; Upendra Rao Anumala; Heinz Hillen; Corinna Klein; Jochen Herms

Accumulation of amyloid-beta leads to loss of functional synapses in Alzheimer’s disease. Dorostkar et al. report that immunotherapy against oligomeric amyloid-beta in the Tg2576 mouse model attenuates synapse loss near plaques, and abolishes it elsewhere. Sequestering oligomeric amyloid-beta may counteract synaptic pathology, even while fibrillar amyloid load remains unchanged.


Acta neuropathologica communications | 2014

In vivo imaging reveals sigmoidal growth kinetic of β-amyloid plaques

Steffen Burgold; Severin Filser; Mario M. Dorostkar; Boris Schmidt; Jochen Herms

A major neuropathological hallmark of Alzheimer’s disease is the deposition of amyloid plaques in the brains of affected individuals. Amyloid plaques mainly consist of fibrillar β-amyloid, which is a cleavage product of the amyloid precursor protein. The amyloid-cascade-hypothesis postulates Aβ accumulation as the central event in initiating a toxic cascade leading to Alzheimer’s disease pathology and, ultimately, loss of cognitive function. We studied the kinetics of β-amyloid deposition in Tg2576 mice, which overexpress human amyloid precursor protein with the Swedish mutation. Utilizing long-term two-photon imaging we were able to observe the entire kinetics of plaque growth in vivo. Essentially, we observed that plaque growth follows a sigmoid-shaped curve comprising a cubic growth phase, followed by saturation. In contrast, plaque density kinetics exhibited an asymptotic progression. Taking into account the fact that a critical concentration of Aβ is required to seed new plaques, we can propose the following kinetic model of β-amyloid deposition in vivo. In the early cubic phase, plaque growth is not limited by Aβ concentration and plaque density increases very fast. During the transition phase, plaque density stabilizes whereas plaque volume increases strongly reflecting a robust growth of the plaques. In the late asymptotic phase, Aβ peptide production becomes rate-limiting for plaque growth. In conclusion, the present study offers a direct link between in vitro and in vivo studies facilitating the translation of Aβ-lowering strategies from laboratory models to patients.


Biological Psychiatry | 2016

Beta-Site Amyloid Precursor Protein Cleaving Enzyme 1 Inhibition Impairs Synaptic Plasticity via Seizure Protein 6

Kaichuan Zhu; Xianyuan Xiang; Severin Filser; Petar Marinković; Mario M. Dorostkar; Sophie Crux; Ulf Neumann; Derya R. Shimshek; Gerhard Rammes; Christian Haass; Stefan F. Lichtenthaler; Jenny M. Gunnersen; Jochen Herms

BACKGROUND Beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) is a promising drug target for the treatment of Alzheimers disease. Prolonged BACE1 inhibition interferes with structural and functional synaptic plasticity in mice, most likely by altering the metabolism of BACE1 substrates. Seizure protein 6 (SEZ6) is predominantly cleaved by BACE1, and Sez6 knockout mice share some phenotypes with BACE1 inhibitor-treated mice. We investigated whether SEZ6 is involved in BACE1 inhibition-induced structural and functional synaptic alterations. METHODS The function of NB-360, a novel blood-brain barrier penetrant and orally available BACE1 inhibitor, was verified by immunoblotting. In vivo microscopy was applied to monitor the impact of long-term pharmacological BACE1 inhibition on dendritic spines in the cerebral cortex of constitutive and conditional Sez6 knockout mice. Finally, synaptic functions were characterized using electrophysiological field recordings in hippocampal slices. RESULTS BACE1 enzymatic activity was strongly suppressed by NB-360. Prolonged NB-360 treatment caused a reversible spine density reduction in wild-type mice, but it did not affect Sez6-/- mice. Knocking out Sez6 in a small subset of mature neurons also prevented the structural postsynaptic changes induced by BACE1 inhibition. Hippocampal long-term potentiation was decreased in both chronic BACE1 inhibitor-treated wild-type mice and vehicle-treated Sez6-/- mice. However, chronic NB-360 treatment did not alter long-term potentiation in CA1 neurons of Sez6-/- mice. CONCLUSIONS Our results suggest that SEZ6 plays an important role in maintaining normal dendritic spine dynamics. Furthermore, SEZ6 is involved in BACE1 inhibition-induced structural and functional synaptic alterations.


Scientific Reports | 2015

A30P α-Synuclein interferes with the stable integration of adult-born neurons into the olfactory network

Johanna Neuner; Severin Filser; Stylianos Michalakis; Martin Biel; Jochen Herms

Impaired olfaction is an early symptom in Parkinson disease (PD), although the exact cause is as yet unknown. Here, we investigated the link between PD-related mutant α-Synuclein (α-SYN) pathology and olfactory deficit, by examining the integration of adult-born neurons in the olfactory bulb (OB) of A30P α-SYN overexpressing mice. To this end, we chose to label one well-known vulnerable subpopulation of adult-born cells, the dopaminergic neurons. Using in vivo two-photon imaging, we followed the dynamic process of neuronal turnover in transgenic A30P α-SYN and wild-type mice over a period of 2.5 months. Our results reveal no difference in the number of cells that reach, and possibly integrate into, the glomerular layer in the OB. However, in mutant transgenic mice these new neurons have a significantly shortened survival, resulting in an overall reduction in the addition of neurons to the glomerular layer over time. We therefore propose unstable integration and impaired homeostasis of functional new neurons as a likely contributor to odour discrimination deficits in mutant α-SYN mice.


Acta Neuropathologica | 2018

BACE1 inhibition more effectively suppresses initiationthan progression of β-amyloid pathology

Finn Peters; Hazal Salihoglu; Eva Ferreira Rodrigues; Etienne Herzog; Tanja Blume; Severin Filser; Mario M. Dorostkar; Derya R. Shimshek; Nils Brose; Ulf Neumann; Jochen Herms

BACE1 is the rate-limiting protease in the production of synaptotoxic β-amyloid (Aβ) species and hence one of the prime drug targets for potential therapy of Alzheimer’s disease (AD). However, so far pharmacological BACE1 inhibition failed to rescue the cognitive decline in mild-to-moderate AD patients, which indicates that treatment at the symptomatic stage might be too late. In the current study, chronic in vivo two-photon microscopy was performed in a transgenic AD model to monitor the impact of pharmacological BACE1 inhibition on early β-amyloid pathology. The longitudinal approach allowed to assess the kinetics of individual plaques and associated presynaptic pathology, before and throughout treatment. BACE1 inhibition could not halt but slow down progressive β-amyloid deposition and associated synaptic pathology. Notably, the data revealed that the initial process of plaque formation, rather than the subsequent phase of gradual plaque growth, is most sensitive to BACE1 inhibition. This finding of particular susceptibility of plaque formation has profound implications to achieve optimal therapeutic efficacy for the prospective treatment of AD.


Nature Communications | 2014

Pathological α-synuclein impairs adult-born granule cell development and functional integration in the olfactory bulb

Johanna Neuner; Saak V. Ovsepian; Mario M. Dorostkar; Severin Filser; Aayush Gupta; Stylianos Michalakis; Martin Biel; Jochen Herms

Although the role of noxious α-synuclein (α-SYN) in the degeneration of midbrain dopaminergic neurons and associated motor deficits of Parkinson’s disease is recognized, its impact on non-motor brain circuits and related symptoms remains elusive. Through combining in vivo two-photon imaging with time-coded labelling of neurons in the olfactory bulb of A30P α-SYN transgenic mice, we show impaired growth and branching of dendrites of adult-born granule cells (GCs), with reduced gain and plasticity of dendritic spines. The spine impairments are especially pronounced during the critical phase of integration of new neurons into existing circuits. Functionally, retarded dendritic expansion translates into reduced electrical capacitance with enhanced intrinsic excitability and responsiveness of GCs to depolarizing inputs, while the spine loss correlates with decreased frequency of AMPA-mediated miniature EPSCs. Changes described here are expected to interfere with the functional integration and survival of new GCs into bulbar networks, contributing towards olfactory deficits and related behavioural impairments.


Scientific Reports | 2017

Neurogenesis from Sox2 expressing cells in the adult cerebellar cortex

Julia Ahlfeld; Severin Filser; Felix Schmidt; Annika K. Wefers; Daniel Merk; Rainer Glaß; Jochen Herms; Ulrich Schüller

We identified a rare undifferentiated cell population that is intermingled with the Bergmann glia of the adult murine cerebellar cortex, expresses the stem cell markers Sox2 and Nestin, and lacks markers of glial or neuronal differentiation. Interestingly, such Sox2+ S100− cells of the adult cerebellum expanded after adequate physiological stimuli in mice (exercise), and Sox2+ precursors acquired positivity for the neuronal marker NeuN over time and integrated into cellular networks. In human patients, SOX2+ S100− cells similarly increased in number after relevant pathological insults (infarcts), suggesting a similar expansion of cells that lack terminal glial differentiation.

Collaboration


Dive into the Severin Filser's collaboration.

Top Co-Authors

Avatar

Jochen Herms

German Center for Neurodegenerative Diseases

View shared research outputs
Top Co-Authors

Avatar

Boris Schmidt

Technische Universität Darmstadt

View shared research outputs
Top Co-Authors

Avatar

Finn Peters

German Center for Neurodegenerative Diseases

View shared research outputs
Top Co-Authors

Avatar

Christian Schön

Center for Integrated Protein Science Munich

View shared research outputs
Top Co-Authors

Avatar

Daniel Merk

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kaichuan Zhu

German Center for Neurodegenerative Diseases

View shared research outputs
Top Co-Authors

Avatar

Lidia Blazquez-Llorca

German Center for Neurodegenerative Diseases

View shared research outputs
Top Co-Authors

Avatar

Martin Biel

Center for Integrated Protein Science Munich

View shared research outputs
Top Co-Authors

Avatar

Stylianos Michalakis

Center for Integrated Protein Science Munich

View shared research outputs
Researchain Logo
Decentralizing Knowledge