Network


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

Hotspot


Dive into the research topics where Peter Borghgraef is active.

Publication


Featured researches published by Peter Borghgraef.


American Journal of Pathology | 2008

Amyloid Activates GSK-3β to Aggravate Neuronal Tauopathy in Bigenic Mice

Dick Terwel; David Muyllaert; Ilse Dewachter; Peter Borghgraef; Sophie Croes; Herman Devijver; Fred Van Leuven

The hypothesis that amyloid pathology precedes and induces the tau pathology of Alzheimers disease is experimentally supported here through the identification of GSK-3 isozymes as a major link in the signaling pathway from amyloid to tau pathology. This study compares two novel bigenic mouse models: APP-V717I x Tau-P301L mice with combined amyloid and tau pathology and GSK-3beta x Tau-P301L mice with tauopathy only. Extensive and remarkable parallels were observed between these strains including 1) aggravation of tauopathy with highly fibrillar tangles in the hippocampus and cortex; 2) prolonged survival correlated to alleviated brainstem tauopathy; 3) development of severe cognitive and behavioral defects in young adults before the onset of amyloid deposition or tauopathy; and 4) presence of pathological phospho-epitopes of tau, including the characteristic GSK-3beta motif at S396/S404. Both GSK-3 isozymes were activated in the brain of parental APP-V717I amyloid mice, even at a young age when cognitive and behavioral defects are evident but before amyloid deposition. The data indicate that amyloid induces tauopathy through activation of GSK-3 and suggest a role for the kinase in maintaining the functional integrity of adult neurons.


The Journal of Neuroscience | 2006

Improved Long-Term Potentiation and Memory in Young Tau-P301L Transgenic Mice before Onset of Hyperphosphorylation and Tauopathy

Karin Boekhoorn; Dick Terwel; Barbara Biemans; Peter Borghgraef; Olof Wiegert; Ger J. A. Ramakers; Koos de Vos; Harm J. Krugers; Takami Tomiyama; Hiroshi Mori; Marian Joëls; Fred Van Leuven; Paul J. Lucassen

The microtubule binding protein tau is implicated in neurodegenerative tauopathies, including frontotemporal dementia (FTD) with Parkinsonism caused by diverse mutations in the tau gene. Hyperphosphorylation of tau is considered crucial in the age-related formation of neurofibrillary tangles (NFTs) correlating well with neurotoxicity and cognitive defects. Transgenic mice expressing FTD mutant tau-P301L recapitulate the human pathology with progressive neuronal impairment and accumulation of NFT. Here, we studied tau-P301L mice for parameters of learning and memory at a young age, before hyperphosphorylation and tauopathy were apparent. Unexpectedly, in young tau-P301L mice, increased long-term potentiation in the dentate gyrus was observed in parallel with improved cognitive performance in object recognition tests. Neither tau phosphorylation, neurogenesis, nor other morphological parameters that were analyzed could account for these cognitive changes. The data demonstrate that learning and memory processes in the hippocampus of young tau-P301L mice are not impaired and actually improved in the absence of marked phosphorylation of human tau. We conclude that protein tau plays an important beneficial role in normal neuronal processes of hippocampal memory, and conversely, that not tau mutations per se, but the ensuing hyperphosphorylation must be critical for cognitive decline in tauopathies.


Magnetic Resonance in Medicine | 2005

Noninvasive in vivo MRI detection of neuritic plaques associated with iron in APP[V717I] transgenic mice, a model for Alzheimer's disease

Greetje Vanhoutte; Ilse Dewachter; Peter Borghgraef; F. Van Leuven; A. Van der Linden

Transgenic mice overexpressing the London mutant of human amyloid precursor protein (APP[V717I]) in neurons develop amyloid plaques in the brain, thus demonstrating the most prominent neuropathological hallmark of Alzheimers disease. In vivo 3D T2*‐weighted MRI on these mice (24 months of age) revealed hypointense brain inclusions that affected the thalamus almost exclusively. Upon correlating these MRI observations with a panel of different histologic staining techniques, it appeared that only plaques that were positive for both thioflavin‐S and iron were visible on the MR images. Numerous thioflavin‐S‐positive plaques in the cortex that did not display iron staining remained invisible to MRI. The in vivo detection of amyloid plaques in this mouse model, using the intrinsic MRI contrast arising from the iron associated with the plaques, creates an unexpected opportunity for the noninvasive investigation of the longitudinal development of the plaques in the same animal. Thus, this work provides further research opportunities for analyzing younger APP[V717I] mouse models with the knowledge of the final outcome at 24 months of age. Magn Reson Med 53:607–613, 2005.


Genes, Brain and Behavior | 2008

Glycogen synthase kinase‐3β, or a link between amyloid and tau pathology?

David Muyllaert; Anneke Kremer; Tomasz Jaworski; Peter Borghgraef; Herman Devijver; Sophie Croes; Ilse Dewachter; F. Van Leuven

Phosphorylation is the most common post‐translational modification of cellular proteins, essential for most physiological functions. Deregulation of phosphorylation has been invoked in disease mechanisms, and the case of Alzheimer’s disease (AD) is no exception: both in the amyloid pathology and in the tauopathy are kinases deeply implicated. The glycogen synthase kinase‐3 (GSK‐3) isozymes participate in diverse cellular processes and important signalling pathways and have been implicitly linked to diverse medical problems, i.e. from diabetes and cancer to mood disorders and schizophrenia, and in the neurodegeneration of AD. Here, we review specific aspects of GSK‐3 isozymes in the framework of recent data that we obtained in novel transgenic mouse models that robustly recapitulate the pathology and mechanistical problems of AD.


PLOS ONE | 2013

Efficacy and Safety of A Liposome-Based Vaccine against Protein Tau, Assessed in Tau.P301L Mice That Model Tauopathy

Clara Theunis; Natalia Crespo-Biel; Valerie Gafner; Maria Pihlgren; María Pilar López-Deber; Pedro Reis; David T. Hickman; Oskar Adolfsson; Nathalie Chuard; Dorin Mlaki Ndao; Peter Borghgraef; Herman Devijver; Fred Van Leuven; Andrea Pfeifer; Andreas Muhs

Progressive aggregation of protein Tau into oligomers and fibrils correlates with cognitive decline and synaptic dysfunction, leading to neurodegeneration in vulnerable brain regions in Alzheimers disease. The unmet need of effective therapy for Alzheimers disease, combined with problematic pharmacological approaches, led the field to explore immunotherapy, first against amyloid peptides and recently against protein Tau. Here we adapted the liposome-based amyloid vaccine that proved safe and efficacious, and incorporated a synthetic phosphorylated peptide to mimic the important phospho-epitope of protein Tau at residues pS396/pS404. We demonstrate that the liposome-based vaccine elicited, rapidly and robustly, specific antisera in wild-type mice and in Tau.P301L mice. Long-term vaccination proved to be safe, because it improved the clinical condition and reduced indices of tauopathy in the brain of the Tau.P301L mice, while no signs of neuro-inflammation or other adverse neurological effects were observed. The data corroborate the hypothesis that liposomes carrying phosphorylated peptides of protein Tau have considerable potential as safe and effective treatment against tauopathies, including Alzheimers disease.


Neurobiology of Aging | 2009

Deregulation of NMDA-receptor function and down-stream signaling in APP(V717I) transgenic mice

Ilse Dewachter; Robert K. Filipkowski; Christina Priller; Laurence Ris; J. Neyton; Sophie Croes; D. Terwel; M Gysemans; Herman Devijver; Peter Borghgraef; Emile Godaux; Leszek Kaczmarek; Jochen Herms; F. Van Leuven

Evidence is accumulating for a role for amyloid peptides in impaired synaptic plasticity and cognition, while the underlying mechanisms remain unclear. We here analyzed the effects of amyloid peptides on NMDA-receptor function in vitro and in vivo. A synthetic amyloid peptide preparation containing monomeric and oligomeric A beta (1-42) peptides was used and demonstrated to bind to synapses expressing NMDA-receptors in cultured hippocampal and cortical neurons. Pre-incubation of primary neuronal cultures with A beta peptides significantly inhibited NMDA-receptor function, albeit not by a direct pharmacological inhibition of NMDA-receptors, since acute application of A beta peptides did not change NMDA-receptor currents in autaptic hippocampal cultures nor in xenopus oocytes expressing recombinant NMDA-receptors. Pre-incubation of primary neuronal cultures with A beta peptides however decreased NR2B-immunoreactive synaptic spines and surface expression of NR2B containing NMDA-receptors. Furthermore, we extended these findings for the first time in vivo, demonstrating decreased concentrations of NMDA-receptor subunit NR2B and PSD-95 as well as activated alpha-CaMKII in postsynaptic density preparations of APP[V717I] transgenic mice. This was associated with impaired NMDA-dependent LTP and decreased NMDA- and AMPA-receptor currents in hippocampal CA1 region in APP[V717I] transgenic mice. In addition, induction of c-Fos following cued and contextual fear conditioning was significantly impaired in the basolateral amygdala and hippocampus of APP[V717I] transgenic mice. Our data demonstrate defects in NMDA-receptor function and learning dependent signaling cascades in vivo in APP[V717I] transgenic mice and point to decreased surface expression of NMDA-receptors as a mechanism involved in early synaptic defects in APP[V717I] transgenic mice in vivo.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Mice lacking phosphatase PP2A subunit PR61/B’δ (Ppp2r5d) develop spatially restricted tauopathy by deregulation of CDK5 and GSK3β

Justin Vijay Louis; Ellen Martens; Peter Borghgraef; Caroline Lambrecht; Ward Sents; Sari Longin; Karen Zwaenepoel; R. Pijnenborg; Isabelle Landrieu; Guy Lippens; Birgit Ledermann; Jürgen Götz; Fred Van Leuven; Jozef Goris; Veerle Janssens

Functional diversity of protein phosphatase 2A (PP2A) enzymes mainly results from their association with distinct regulatory subunits. To analyze the functions of one such holoenzyme in vivo, we generated mice lacking PR61/B’δ (B56δ), a subunit highly expressed in neural tissues. In PR61/B’δ-null mice the microtubule-associated protein tau becomes progressively phosphorylated at pathological epitopes in restricted brain areas, with marked immunoreactivity for the misfolded MC1-conformation but without neurofibrillary tangle formation. Behavioral tests indicated impaired sensorimotor but normal cognitive functions. These phenotypical characteristics were further underscored in PR61/B’δ-null mice mildly overexpressing human tau. PR61/B’δ-containing PP2A (PP2AT61δ) poorly dephosphorylates tau in vitro, arguing against a direct dephosphorylation defect. Rather, the activity of glycogen synthase kinase-3β, a major tau kinase, was found increased, with decreased phosphorylation of Ser-9, a putative cyclin-dependent kinase 5 (CDK5) target. Accordingly, CDK5 activity is decreased, and its cellular activator p35, strikingly absent in the affected brain areas. As opposed to tau, p35 is an excellent PP2AT61δ substrate. Our data imply a nonredundant function for PR61/B’δ in phospho-tau homeostasis via an unexpected spatially restricted mechanism preventing p35 hyperphosphorylation and its subsequent degradation.


Neurobiology of Disease | 2009

GSK3ß, a centre-staged kinase in neuropsychiatric disorders, modulates long term memory by inhibitory phosphorylation at Serine-9

Ilse Dewachter; Laurence Ris; Tomasz Jaworski; Claire Marie Seymour; Anneke Kremer; Peter Borghgraef; H De Vijver; Emile Godaux; F. Van Leuven

Accumulating evidence implicates deregulation of GSK3ss as a converging pathological event in Alzheimers disease and in neuropsychiatric disorders, including bipolar disorder and schizophrenia. Although these neurological disorders share cognitive dysfunction as a hallmark, the role of GSK3ss in learning and memory remains to be explored in depth. We here report increased phosphorylation of GSK3ss at Serine-9 following cognitive training in two different hippocampus dependent cognitive tasks, i.e. inhibitory avoidance and novel object recognition task. Conversely, transgenic mice expressing the phosphorylation defective mutant GSK3ss[S9A] show impaired memory in these tasks. Furthermore, GSK3ss[S9A] mice displayed impaired hippocampal L-LTP and facilitated LTD. Application of actinomycin, but not anisomycin, mimicked GSK3ss[S9A] induced defects in L-LTP, suggesting that transcriptional activation is affected. This was further supported by decreased expression of the immediate early gene c-Fos, a target gene of CREB. The combined data demonstrate a role for GSK3ss in long term memory formation, by inhibitory phosphorylation at Serine-9. The findings are fundamentally important and relevant in the search for therapeutic strategies in neurological disorders associated with cognitive impairment and deregulated GSK3ss signaling, including AD, bipolar disorder and schizophrenia.


The Journal of Neuroscience | 2010

Upper Airway Dysfunction of Tau-P301L Mice Correlates with Tauopathy in Midbrain and Ponto-Medullary Brainstem Nuclei

Mathias Dutschmann; Clément Menuet; Georg M. Stettner; Christian Gestreau; Peter Borghgraef; Herman Devijver; Lies Gielis; Gérard Hilaire; Fred Van Leuven

Tauopathy comprises hyperphosphorylation of the microtubule-associated protein tau, causing intracellular aggregation and accumulation as neurofibrillary tangles and neuropil treads. Some primary tauopathies are linked to mutations in the MAPT gene coding for protein tau, but most are sporadic with unknown causes. Also, in Alzheimers disease, the most frequent secondary tauopathy, neither the cause nor the pathological mechanisms and repercussions are understood. Transgenic mice expressing mutant Tau-P301L suffer cognitive and motor defects and die prematurely from unknown causes. Here, in situ electrophysiology in symptomatic Tau-P301L mice (7–8 months of age) revealed reduced postinspiratory discharges of laryngeal motor outputs that control laryngeal constrictor muscles. Under high chemical drive (hypercapnia), postinspiratory discharge was nearly abolished, whereas laryngeal inspiratory discharge was increased disproportionally. The latter may suggest a shift of postinspiratory laryngeal constrictor activity into inspiration. In vivo double-chamber plethysmography of Tau-P301L mice showed significantly reduced respiratory airflow but significantly increased chest movements during baseline breathing, but particularly in hypercapnia, confirming a significant increase in inspiratory resistive load. Histological analysis demonstrated hyperphosphorylated tau in brainstem nuclei, directly or indirectly involved in upper airway motor control (i.e., the Kölliker–Fuse, periaqueductal gray, and intermediate reticular nuclei). In contrast, young Tau-P301L mice did not show breathing disorders or brainstem tauopathy. Consequently, in aging Tau-P301L mice, progressive upper airway dysfunction is linked to progressive tauopathy in identified neural circuits. Because patients with tauopathy suffer from upper airway dysfunction, the Tau-P301L mice can serve as an experimental model to study disease-specific synaptic dysfunction in well defined functional neural circuits.


PLOS ONE | 2009

AAV-Tau Mediates Pyramidal Neurodegeneration by Cell-Cycle Re-Entry without Neurofibrillary Tangle Formation in Wild-Type Mice

Tomasz Jaworski; Ilse Dewachter; Benoit Lechat; Sophie Croes; Annelies Termont; David Demedts; Peter Borghgraef; Herman Devijver; Robert K. Filipkowski; Leszek Kaczmarek; Sebastian Kügler; Fred Van Leuven

In Alzheimers disease tauopathy is considered secondary to amyloid, and the duality obscures their relation and the definition of their respective contributions. Transgenic mouse models do not resolve this problem conclusively, i.e. the relative hierarchy of amyloid and tau pathology depends on the actual model and the genes expressed or inactivated. Here, we approached the problem in non-transgenic models by intracerebral injection of adeno-associated viral vectors to express protein tau or amyloid precursor protein in the hippocampus in vivo. AAV-APP mutant caused neuronal accumulation of amyloid peptides, and eventually amyloid plaques at 6 months post-injection, but with only marginal hippocampal cell-death. In contrast, AAV-Tau, either wild-type or mutant P301L, provoked dramatic degeneration of pyramidal neurons in CA1/2 and cortex within weeks. Tau-mediated neurodegeneration proceeded without formation of large fibrillar tau-aggregates or tangles, but with increased expression of cell-cycle markers. We present novel AAV-based models, which demonstrate that protein tau mediates pyramidal neurodegeneration in vivo. The data firmly support the unifying hypothesis that post-mitotic neurons are forced to re-enter the cell-cycle in primary and secondary tauopathies, including Alzheimers disease.

Collaboration


Dive into the Peter Borghgraef's collaboration.

Top Co-Authors

Avatar

Fred Van Leuven

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Herman Devijver

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Ilse Dewachter

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Benoit Lechat

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

David Muyllaert

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Clara Theunis

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

F. Van Leuven

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Tomasz Jaworski

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Hervé Maurin

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Natalia Crespo-Biel

Katholieke Universiteit Leuven

View shared research outputs
Researchain Logo
Decentralizing Knowledge