Network


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

Hotspot


Dive into the research topics where Fabio Canneva is active.

Publication


Featured researches published by Fabio Canneva.


PLOS ONE | 2012

Altered hypothalamic protein expression in a rat model of Huntington's disease.

Wei-na Cong; Huan Cai; Rui Wang; Caitlin M. Daimon; Stuart Maudsley; Kerstin Raber; Fabio Canneva; Stephan von Hörsten; Bronwen Martin

Huntingtons disease (HD) is a neurodegenerative disorder, which is characterized by progressive motor impairment and cognitive alterations. Changes in energy metabolism, neuroendocrine function, body weight, euglycemia, appetite function, and circadian rhythm can also occur. It is likely that the locus of these alterations is the hypothalamus. We used the HD transgenic (tg) rat model bearing 51 CAG repeats, which exhibits similar HD symptomology as HD patients to investigate hypothalamic function. We conducted detailed hypothalamic proteome analyses and also measured circulating levels of various metabolic hormones and lipids in pre-symptomatic and symptomatic animals. Our results demonstrate that there are significant alterations in HD rat hypothalamic protein expression such as glial fibrillary acidic protein (GFAP), heat shock protein-70, the oxidative damage protein glutathione peroxidase (Gpx4), glycogen synthase1 (Gys1) and the lipid synthesis enzyme acylglycerol-3-phosphate O-acyltransferase 1 (Agpat1). In addition, there are significant alterations in various circulating metabolic hormones and lipids in pre-symptomatic animals including, insulin, leptin, triglycerides and HDL, before any motor or cognitive alterations are apparent. These early metabolic and lipid alterations are likely prodromal signs of hypothalamic dysfunction. Gaining a greater understanding of the hypothalamic and metabolic alterations that occur in HD, could lead to the development of novel therapeutics for early interventional treatment of HD.


PLOS ONE | 2013

Characterization of acid sphingomyelinase activity in human cerebrospinal fluid.

Christiane Mühle; Hagen B. Huttner; Silke Walter; Martin Reichel; Fabio Canneva; Piotr Lewczuk; Erich Gulbins; Johannes Kornhuber

Background As a key enzyme in sphingolipid metabolism, acid sphingomyelinase (ASM) is involved in the regulation of cell fate and signaling via hydrolysis of sphingomyelin to form ceramide. While increased activity of the lysosomal form has been associated with various pathological conditions, there are few studies on secretory ASM limited only to cell models, plasma or serum. Methods An optimized assay based on a fluorescent substrate was applied to measure the ASM activity in cerebrospinal fluid (CSF) collected from mice and from 42 patients who were classified as controls based on normal routine CSF values. Results We have detected ASM activity in human CSF, established a sensitive quantitative assay and characterized the enzyme’s properties. The enzyme resembles plasmatic ASM including protein stability and Zn2+-dependence but the assays differ considerably in the optimal detergent concentration. Significantly increased activities in the CSF of ASM transgenic mice and undetectable levels in ASM knock-out mice prove that the measured ASM activity originates from the ASM-encoding gene SMPD1. CSF localized ASM activities were comparable to corresponding serum ASM levels at their respective optimal reaction conditions, but no correlation was observed. The large variance in ASM activity was independent of sex, age or analyzed routine CSF parameters. Conclusions Human and mouse CSF contain detectable levels of secretory ASM, which are unrelated to serum ASM activities. Further investigations in humans and in animal models will help to elucidate the role of this enzyme in human disease and to assess its value as a potential biomarker for disease type, severity, progress or therapeutic success.


BMC Neuroscience | 2013

Glutaminyl cyclase-mediated toxicity of pyroglutamate-beta amyloid induces striatal neurodegeneration

Andreas Becker; Stephanie Kohlmann; Anca Alexandru; Wolfgang Jagla; Fabio Canneva; Christoph Bäuscher; Holger Cynis; Reinhard Sedlmeier; Sigrid Graubner; Stephan Schilling; Hans-Ulrich Demuth; Stephan von Hörsten

BackgroundPosttranslational modifications of beta amyloid (Aβ) have been shown to affect its biophysical and neurophysiological properties. One of these modifications is N-terminal pyroglutamate (pE) formation. Enzymatic glutaminyl cyclase (QC) activity catalyzes cyclization of truncated Aβ(3-x), generating pE3-Aβ. Compared to unmodified Aβ, pE3-Aβ is more hydrophobic and neurotoxic. In addition, it accelerates aggregation of other Aβ species. To directly investigate pE3-Aβ formation and toxicity in vivo, transgenic (tg) ETNA (E at the truncated N-terminus of Aβ) mice expressing truncated human Aβ(3–42) were generated and comprehensively characterized. To further investigate the role of QC in pE3-Aβ formation in vivo, ETNA mice were intercrossed with tg mice overexpressing human QC (hQC) to generate double tg ETNA-hQC mice.ResultsExpression of truncated Aβ(3–42) was detected mainly in the lateral striatum of ETNA mice, leading to progressive accumulation of pE3-Aβ. This ultimately resulted in astrocytosis, loss of DARPP-32 immunoreactivity, and neuronal loss at the sites of pE3-Aβ formation. Neuropathology in ETNA mice was associated with behavioral alterations. In particular, hyperactivity and impaired acoustic sensorimotor gating were detected. Double tg ETNA-hQC mice showed similar Aβ levels and expression sites, while pE3-Aβ were significantly increased, entailing increased astrocytosis and neuronal loss.ConclusionsETNA and ETNA-hQC mice represent novel mouse models for QC-mediated toxicity of truncated and pE-modified Aβ. Due to their significant striatal neurodegeneration these mice can also be used for analysis of striatal regulation of basal locomotor activity and sensorimotor gating, and possibly for DARPP-32-dependent neurophysiology and neuropathology. The spatio-temporal correlation of pE3-Aβ and neuropathology strongly argues for an important role of this Aβ species in neurodegenerative processes in these models.


Developmental Neurobiology | 2016

Effects of In utero environment and maternal behavior on neuroendocrine and behavioral alterations in a mouse model of prenatal trauma.

Y. Golub; Fabio Canneva; R. Funke; S. Frey; Joerg Distler; S. von Hörsten; C.M. Freitag; Oliver Kratz; Gunther H. Moll; J. Solati

Maternal posttraumatic stress disorder (PTSD) following trauma exposure during pregnancy is associated with an increased risk of affective disorders in children. To investigate the mechanisms by which prenatal trauma and/or maternal PTSD affect brain development and behavior we established a mouse model of prenatal traumatic (PT) experience based on the application of an electric foot shock to C57Bl/6N female mice on the gestational day 12 during their pregnancy. The model is based on a previously validated animal model of PTSD. We found high anxiety levels and poor maternal care along with reduced serum prolactin and increased corticosterone levels in dams following maternal trauma (MT). PT‐pups were born smaller and stayed smaller throughout their life. We show increased time and frequency of ultrasonic calls in PT‐pups when separated from the mothers on the postnatal day (PND) 9. Cross‐fostering experiments reveal lower anxiety levels in PT pups raised by healthy mothers as compared to trauma‐naive pups raised by MT‐dams.


Aging Cell | 2016

Differential transgene expression patterns in Alzheimer mouse models revealed by novel human amyloid precursor protein-specific antibodies.

Corinna Höfling; Markus Morawski; Ulrike Zeitschel; Elisa R. Zanier; Katrin Moschke; Alperen Serdaroglu; Fabio Canneva; Stephan von Hörsten; Maria Grazia De Simoni; Gianluigi Forloni; Carsten Jäger; Elisabeth Kremmer; Steffen Roßner; Stefan F. Lichtenthaler; Peer-Hendrik Kuhn

Alzheimers disease (AD) is histopathologically characterized by neurodegeneration, the formation of intracellular neurofibrillary tangles and extracellular Aβ deposits that derive from proteolytic processing of the amyloid precursor protein (APP). As rodents do not normally develop Aβ pathology, various transgenic animal models of AD were designed to overexpress human APP with mutations favouring its amyloidogenic processing. However, these mouse models display tremendous differences in the spatial and temporal appearance of Aβ deposits, synaptic dysfunction, neurodegeneration and the manifestation of learning deficits which may be caused by age‐related and brain region‐specific differences in APP transgene levels. Consequentially, a comparative temporal and regional analysis of the pathological effects of Aβ in mouse brains is difficult complicating the validation of therapeutic AD treatment strategies in different mouse models. To date, no antibodies are available that properly discriminate endogenous rodent and transgenic human APP in brains of APP‐transgenic animals. Here, we developed and characterized rat monoclonal antibodies by immunohistochemistry and Western blot that detect human but not murine APP in brains of three APP‐transgenic mouse and one APP‐transgenic rat model. We observed remarkable differences in expression levels and brain region‐specific expression of human APP among the investigated transgenic mouse lines. This may explain the differences between APP‐transgenic models mentioned above. Furthermore, we provide compelling evidence that our new antibodies specifically detect endogenous human APP in immunocytochemistry, FACS and immunoprecipitation. Hence, we propose these antibodies as standard tool for monitoring expression of endogenous or transfected APP in human cells and APP expression in transgenic animals.


Journal of Psychopharmacology | 2017

Capturing schizophrenia-like prodromal symptoms in a spinocerebellar ataxia-17 transgenic rat

Davide Amato; Fabio Canneva; Huu Phuc Nguyen; Peter Bauer; Olaf Riess; Stephan von Hörsten; Christian P. Müller

Rationale: The polyglutamine disease spinocerebellar ataxia type 17 (SCA17) is a neurodegenerative disease leading to severe neurological symptoms during development. Additionally, patients affected by SCA17 display psychosis earlier than their motor disorders. Objective: Here the putative psychotic phenotype and endophenotype of transgenic SCA17 rats was examined. Methods: The expression of schizophrenia-like symptoms was evaluated over a longitudinal period before and after the onset of neurological symptoms in SCA17. To this end, transgenic SCA17 rats’ monoamine neurotransmission was investigated along with their locomotion at baseline and in response to amphetamine using in-vivo microdialysis in free moving conditions, their sensorimotor gating using pre-pulse inhibition of startle reaction, and their object memory using the novel object recognition test as an index of cognitive impairments. Results: Presymptomatic SCA17 rats displayed dysregulated monoamine levels at baseline and in response to amphetamine compared with control wild-type (wt) rats. At that stage, neither amphetamine-induced hyperlocomotion nor sensorimotor gating differed from that in wt rats. Symptomatic SCA17 rats developed sensorimotor gating deficits and also showed an impaired object memory, while their monoaminergic responses remained supersensitive to amphetamine. Conclusions: The data of the present study demonstrate a neurochemical endophenotype in SCA17 rats resembling that of prodromal schizophrenia. These findings suggest that a sensitization of the monoamine systems arises early in adulthood in SCA17 rats and may predispose them to express schizophrenia-like symptoms later in life.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Early postnatal behavioral, cellular, and molecular changes in models of Huntington disease are reversible by HDAC inhibition

Florian A. Siebzehnrubl; Kerstin Raber; Yvonne K. Urbach; Anja Schulze-Krebs; Fabio Canneva; Sandra Moceri; Johanna Habermeyer; Dalila Achoui; Bhavana Gupta; Dennis A. Steindler; Michael Stephan; Huu Phuc Nguyen; Michael Bonin; Olaf Riess; Andreas Bauer; Ludwig Aigner; Sebastien Couillard-Despres; Martin Paucar; Per Svenningsson; Alexander P. Osmand; Alexander Andreew; Claus Zabel; Andreas Weiss; Rainer Kuhn; Saliha Moussaoui; Ines Blockx; Annemie Van der Linden; Rachel Y. Cheong; Laurent Roybon; Åsa Petersén

Significance In Huntington disease (HD) gene carriers the disease-causing mutant Huntingtin (mHTT) is already present during early developmental stages, but, surprisingly, HD patients develop clinical symptoms only many years later. While a developmental role of Huntingtin has been described, so far new therapeutic approaches targeting those early neurodevelopmental processes are lacking. Here, we show that behavioral, cellular, and molecular changes associated with mHTT in the postnatal period of genetic animal models of HD can be reverted using low-dose treatment with a histone deacetylation inhibitor. Our findings support a neurodevelopmental basis for HD and provide proof of concept that pre-HD symptoms, including aberrant neuronal differentiation, are reversible by early therapeutic intervention in vivo. Huntington disease (HD) is an autosomal dominant neurodegenerative disorder caused by expanded CAG repeats in the huntingtin gene (HTT). Although mutant HTT is expressed during embryonic development and throughout life, clinical HD usually manifests later in adulthood. A number of studies document neurodevelopmental changes associated with mutant HTT, but whether these are reversible under therapy remains unclear. Here, we identify very early behavioral, molecular, and cellular changes in preweaning transgenic HD rats and mice. Reduced ultrasonic vocalization, loss of prepulse inhibition, and increased risk taking are accompanied by disturbances of dopaminergic regulation in vivo, reduced neuronal differentiation capacity in subventricular zone stem/progenitor cells, and impaired neuronal and oligodendrocyte differentiation of mouse embryo-derived neural stem cells in vitro. Interventional treatment of this early phenotype with the histone deacetylase inhibitor (HDACi) LBH589 led to significant improvement in behavioral changes and markers of dopaminergic neurotransmission and complete reversal of aberrant neuronal differentiation in vitro and in vivo. Our data support the notion that neurodevelopmental changes contribute to the prodromal phase of HD and that early, presymptomatic intervention using HDACi may represent a promising novel treatment approach for HD.


Molecular Psychiatry | 2018

A dopaminergic mechanism of antipsychotic drug efficacy, failure, and failure reversal: the role of the dopamine transporter

Davide Amato; Fabio Canneva; Paul Cumming; Simone Maschauer; Dominik Groos; Jana Katharina Dahlmanns; Teja W. Grömer; Lisa Chiofalo; Marc Dahlmanns; Fang Zheng; Johannes Kornhuber; Olaf Prante; Christian Alzheimer; Stephan von Hörsten; Christian P. Müller

Antipsychotic drugs are effective interventions in schizophrenia. However, the efficacy of these agents often decreases over time, which leads to treatment failure and symptom recurrence. We report that antipsychotic efficacy in rat models declines in concert with extracellular striatal dopamine levels rather than insufficient dopamine D2 receptor occupancy. Antipsychotic efficacy was associated with a suppression of dopamine transporter activity, which was reversed during failure. Antipsychotic failure coincided with reduced dopamine neuron firing, which was not observed during antipsychotic efficacy. Synaptic field responses in dopamine target areas declined during antipsychotic efficacy and showed potentiation during failure. Antipsychotics blocked synaptic vesicle release during efficacy but enhanced this release during failure. We found that the pharmacological inhibition of the dopamine transporter rescued antipsychotic drug treatment outcomes, supporting the hypothesis that the dopamine transporter is a main target of antipsychotic drugs and predicting that dopamine transporter blockers may be an adjunct treatment to reverse antipsychotic treatment failure.


Journal of Neuroscience Methods | 2018

Dynamic footprint based locomotion sway assessment in α-synucleinopathic mice using Fast Fourier Transform and Low Pass Filter

Ivanna K. Timotius; Fabio Canneva; Georgia Minakaki; Cristian Pasluosta; Sandra Moceri; Nicolas Casadei; Olaf Riess; Jürgen Winkler; Jochen Klucken; Stephan von Hörsten; Bjoern M. Eskofier

BACKGROUND Sway is a crucial gait characteristic tightly correlated with the risk of falling in patients with Parkinsońs disease (PD). So far, the swaying pattern during locomotion has not been investigated in rodent models using the analysis of dynamic footprint recording obtained from the CatWalk gait recording and analysis system. NEW METHODS We present three methods for describing locomotion sway and apply them to footprint recordings taken from C57BL6/N wild-type mice and two different α-synuclein transgenic PD-relevant mouse models (α-synm-ko, α-synm-koxα-synh-tg). Individual locomotion data were subjected to three different signal processing analytical approaches: the first two methods are based on Fast Fourier Transform (FFT), while the third method uses Low Pass Filters (LPF). These methods use the information associated with the locomotion sway and generate sway-related parameters. RESULTS The three proposed methods were successfully applied to the footprint recordings taken from all paws as well as from front/hind-paws separately. Nine resulting sway-related parameters were generated and successfully applied to differentiate between the mouse models under study. Namely, α-synucleinopathic mice revealed higher sway and sway itself was significantly higher in the α-synm-koxα-synh-tg mice compared to their wild-type littermates in eight of the nine sway-related parameters. COMPARISON WITH EXISTING METHOD Previous locomotion sway index computation is based on the estimated center of mass position of mice. CONCLUSIONS The methods presented in this study provide a sway-related gait characterization. Their application is straightforward and may lead to the identification of gait pattern derived biomarkers in rodent models of PD.


Behavioural Brain Research | 2019

Role of hypothalamus-pituitary-adrenal axis modulation in the stress-resilient phenotype of DPP4-deficient rats

Yulia Golub; Eva-Maria Schildbach; Chadi Touma; Oliver Kratz; Gunther H. Moll; Stephan von Hörsten; Fabio Canneva

Background: Dipeptidyl peptidase 4 (DPP4, CD26) is a moonlighting enzyme responsible for the proteolytic inactivation of neuropeptide Y (NPY), a peptide known for its anxiolytic effect in the central nervous system. Our previous work revealed a stress‐resilient phenotype and a potentiation of short‐term fear extinction in a congenic rat model deficient for DPP4 activity (DPP4mut). Here, we investigated neuroendocrine mechanisms underlying the phenotype of the DPP4mut animals. We studied the function of the hypothalamus–pituitary–adrenal (HPA) axis including the expression levels of its key genes and explored the possibility of structural NPY system changes. Methods and results: We find decreased expression of Nr3c1 (glucocorticoid receptor ‐ GR) and Fkbp5 (FK506 binding protein 5) in the amygdala and the hypothalamus of the DPP4mut rats, as well as the lower stress‐induced peripheral corticosterone (CORT) levels. We detect no significant alterations in basal and DEX‐induced CORT levels in the DPP4mut animals. The abundance of NPY‐ergic neurons in the basolateral amygdala, dentate gyrus and hippocampus did not differ between the DPP4mut and their wild type littermates. Conclusion: DPP4mut rats show blunted CORT response in line with their lower behavioral stress‐response profile. These results are consistent with the hypothesis that increased central NPY levels elevate the threshold of stress response. We suggest that changes in the expression levels of key HPA axis genes (Nr3c1 and Fkbp5) are a consequence of the altered stress‐perception of DPP4mut animals, thus further contributing to the stress‐resilient phenotype. HighlightsDipeptidyl peptidase 4 (DPP4, CD26) is an enzyme responsible for the inactivation of Neuropeptide Y.DPP4 deficiency in rats results in a stress‐resilient phenotype.DPP4mut rats show a blunted corticosterone response to stress.DPP4mut rats show no alterations in basal and Dexamethasone‐ induced corticosterone levels.This phenotype goes along with an altered expression of the hypothalamus–pituitary–adrenal‐axis genes.

Collaboration


Dive into the Fabio Canneva's collaboration.

Top Co-Authors

Avatar

Stephan von Hörsten

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Julia Dobner

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Olaf Riess

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar

Davide Amato

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Johannes Kornhuber

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Kerstin Raber

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Anne-Christine Plank

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Olaf Prante

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Sandra Moceri

University of Erlangen-Nuremberg

View shared research outputs
Researchain Logo
Decentralizing Knowledge