Network


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

Hotspot


Dive into the research topics where Anke Van der Perren is active.

Publication


Featured researches published by Anke Van der Perren.


The EMBO Journal | 2013

Endonuclease G mediates α‐synuclein cytotoxicity during Parkinson's disease

Sabrina Büttner; Lukas Habernig; Filomena Broeskamp; Doris Ruli; F.-Nora Vögtle; Manolis Vlachos; Francesca Macchi; Victoria Küttner; Didac Carmona-Gutierrez; Tobias Eisenberg; Julia Ring; Maria Markaki; Asli Aras Taskin; Stefan Benke; Christoph Ruckenstuhl; Ralf J. Braun; Chris Van den Haute; Tine Bammens; Anke Van der Perren; Kai-Uwe Fröhlich; Joris Winderickx; Guido Kroemer; Veerle Baekelandt; Nektarios Tavernarakis; Gabor G. Kovacs; Jörn Dengjel; Chris Meisinger; Stephan J. Sigrist; Frank Madeo

Malfunctioning of the protein α‐synuclein is critically involved in the demise of dopaminergic neurons relevant to Parkinsons disease. Nonetheless, the precise mechanisms explaining this pathogenic neuronal cell death remain elusive. Endonuclease G (EndoG) is a mitochondrially localized nuclease that triggers DNA degradation and cell death upon translocation from mitochondria to the nucleus. Here, we show that EndoG displays cytotoxic nuclear localization in dopaminergic neurons of human Parkinson‐diseased patients, while EndoG depletion largely reduces α‐synuclein‐induced cell death in human neuroblastoma cells. Xenogenic expression of human α‐synuclein in yeast cells triggers mitochondria‐nuclear translocation of EndoG and EndoG‐mediated DNA degradation through a mechanism that requires a functional kynurenine pathway and the permeability transition pore. In nematodes and flies, EndoG is essential for the α‐synuclein‐driven degeneration of dopaminergic neurons. Moreover, the locomotion and survival of α‐synuclein‐expressing flies is compromised, but reinstalled by parallel depletion of EndoG. In sum, we unravel a phylogenetically conserved pathway that involves EndoG as a critical downstream executor of α‐synuclein cytotoxicity.


Neurobiology of Aging | 2015

Longitudinal follow-up and characterization of a robust rat model for Parkinson's disease based on overexpression of alpha-synuclein with adeno-associated viral vectors

Anke Van der Perren; Jaan Toelen; Cindy Casteels; Francesca Macchi; Anne-Sophie Van Rompuy; Sophie Sarre; Nicolas Casadei; Silke Nuber; Uwe Himmelreich; Maria Isabel Osorio Garcia; Yvette Michotte; Rudi D'Hooge; Guy Bormans; Koen Van Laere; Rik Gijsbers; Chris Van den Haute; Zeger Debyser; Veerle Baekelandt

Testing of new therapeutic strategies for Parkinsons disease (PD) is currently hampered by the lack of relevant and reproducible animal models. Here, we developed a robust rat model for PD by injection of adeno-associated viral vectors (rAAV2/7) encoding α-synuclein into the substantia nigra, resulting in reproducible nigrostriatal pathology and behavioral deficits in a 4-week time period. Progressive dopaminergic dysfunction was corroborated by histopathologic and biochemical analysis, motor behavior testing and in vivo microdialysis. L-DOPA treatment was found to reverse the behavioral phenotype. Non-invasive positron emission tomography imaging and magnetic resonance spectroscopy allowed longitudinal monitoring of neurodegeneration. In addition, insoluble α-synuclein aggregates were formed in this model. This α-synuclein rat model shows improved face and predictive validity, and therefore offers the possibility to reliably test novel therapeutics. Furthermore, it will be of great value for further research into the molecular pathogenesis of PD and the importance of α-synuclein aggregation in the disease process.


Neurobiology of Aging | 2015

FK506 reduces neuroinflammation and dopaminergic neurodegeneration in an α-synuclein-based rat model for Parkinson's disease.

Anke Van der Perren; Francesca Macchi; Jaan Toelen; Marianne Carlon; Michael Maris; Henriette de Loor; Dirk Kuypers; Rik Gijsbers; Chris Van den Haute; Zeger Debyser; Veerle Baekelandt

Alpha-synuclein (α-synuclein) is considered a key player in Parkinsons disease (PD), but the exact relationship between α-synuclein aggregation and dopaminergic neurodegeneration remains unresolved. There is increasing evidence that neuroinflammatory processes are closely linked to dopaminergic cell death, but whether the inflammatory process is causally involved in PD or rather reflects secondary consequences of nigrostriatal pathway injury is still under debate. We evaluated the therapeutic effect of the immunophilin ligand FK506 in a rAAV2/7 α-synuclein overexpression rat model. Treatment with FK506 significantly increased the survival of dopaminergic neurons in a dose-dependent manner. No reduction in α-synuclein aggregation was apparent in this time window, but FK506 significantly lowered the infiltration of both T helper and cytotoxic T cells and the number and subtype of microglia and macrophages. These data suggest that the anti-inflammatory properties of FK506 decrease neurodegeneration in this α-synuclein-based PD model, pointing to a causal role of neuroinflammation in the pathogenesis of PD.


The Journal of Nuclear Medicine | 2011

A PET Brain Reporter Gene System Based on Type 2 Cannabinoid Receptors

Caroline Vandeputte; Nele Evens; Jaan Toelen; Christophe Deroose; Barbara Bosier; Abdelilah Ibrahimi; Anke Van der Perren; Rik Gijsbers; Peter Janssen; Didier M. Lambert; Alfons Verbruggen; Zeger Debyser; Guy Bormans; Veerle Baekelandt; Koen Van Laere

PET of gene expression in the brain may greatly facilitate neuroscience research and potential clinical implementation of gene or cell therapy of central nervous system diseases. To date, no adequate PET reporter system is available for the central nervous system because available tracers either do not cross the intact blood–brain barrier or have high background signals. Here we report the first, to our knowledge, PET reporter system for imaging gene expression in the intact brain. Methods: We selected the human type 2 cannabinoid receptor (hCB2) as a reporter because of its low basal expression in the brain. An inactive mutant (D80N) was chosen so as not to interfere with signal transduction. As a reporter probe we used the 11C-labeled CB2 ligand, 11C-GW405833, which readily crosses the blood–brain barrier. Dual-modality imaging lentiviral and adeno-associated viral vectors encoding both hCB2(D80N) and firefly luciferase or enhanced green fluorescent protein were engineered and validated in cell culture. Next, hCB2(D80N) was locoregionally overexpressed in rat striatum by stereotactic injection of lentiviral and adeno-associated viral vectors. Results: Kinetic PET revealed specific and reversible CB2 binding of 11C-GW405833 in the transduced rat striatum. hCB2 and firefly luciferase expression was followed until 9 mo and showed similar kinetics. Both hCB2 expression and enhanced green fluorescent protein expression were confirmed by immunohistochemistry. Conclusion: Dual-modality imaging viral vectors encoding hCB2(D80N) were engineered, and the reporter system was validated in different animal species. The results support the potential future clinical use of CB2 as a PET reporter in the intact brain.


Frontiers in Behavioral Neuroscience | 2015

Nigral proteasome inhibition in mice leads to motor and non-motor deficits and increased expression of Ser129 phosphorylated α-synuclein

Eduard Bentea; Anke Van der Perren; Joeri Van Liefferinge; Anissa El Arfani; Giulia Albertini; Thomas Demuyser; Ellen Merckx; Yvette Michotte; Ilse Smolders; Veerle Baekelandt; Ann Massie

Parkinsons disease is a neurodegenerative disorder characterized by motor and non-motor disturbances. Various pathogenic pathways drive disease progression including oxidative stress, mitochondrial dysfunction, α-synuclein aggregation and impairment of protein degradation systems. Dysfunction of the ubiquitin-proteasome system in the substantia nigra of Parkinsons disease patients is believed to be one of the causes of protein aggregation and cell death associated with this disorder. Lactacystin, a potent inhibitor of the proteasome, was previously delivered to the nigrostriatal pathway of rodents to model nigrostriatal degeneration. Although lactacystin-treated animals develop parkinsonian motor impairment, it is currently unknown whether they also develop non-motor symptoms characteristic of this disorder. In order to further describe the proteasome inhibition model of Parkinsons disease, we characterized the unilateral lactacystin model, performed by stereotaxic injection of the toxin in the substantia nigra of mice. We studied the degree of neurodegeneration and the behavioral phenotype 1 and 3 weeks after lactacystin lesion both in terms of motor impairment, as well as non-motor symptoms. We report that unilateral administration of 3 μg lactacystin to the substantia nigra of mice leads to partial (~40%) dopaminergic cell loss and concurrent striatal dopamine depletion, accompanied by increased expression of Ser129-phosphorylated α-synuclein. Behavioral characterization of the model revealed parkinsonian motor impairment, as well as signs of non-motor disturbances resembling early stage Parkinsons disease including sensitive and somatosensory deficits, anxiety-like behavior, and perseverative behavior. The consistent finding of good face validity, together with relevant construct validity, warrant a further evaluation of proteasome inhibition models of Parkinsons disease in pre-clinical research and validation of therapeutic targets.


Neurobiology of Aging | 2014

Alpha-synuclein-induced neurodegeneration is exacerbated in PINK1 knockout mice

Marusela Oliveras-Salvá; Francesca Macchi; Valérie Coessens; Angélique Deleersnijder; Melanie Gérard; Anke Van der Perren; Chris Van den Haute; Veerle Baekelandt

Loss-of-function mutations in the PINK1 gene lead to recessive forms of Parkinsons disease. Animal models with depleted PINK1 expression have failed to reproduce significant nigral dopaminergic neurodegeneration and clear alpha-synuclein pathology, main characteristics of the disease. In this study, we investigated whether alpha-synuclein pathology is altered in the absence of PINK1 in cell culture and in vivo. We observed that downregulation of PINK1 enhanced alpha-synuclein aggregation and apoptosis in a neuronal cell culture model for synucleinopathy. Silencing of PINK1 expression in mouse substantia nigra using recombinant adeno-associated viral vectors did not induce dopaminergic neurodegeneration in a long-term study up to 10 months, nor did it enhance or accelerate dopaminergic neurodegeneration after alpha-synuclein overexpression. However, in PINK1 knockout mice, overexpression of alpha-synuclein in the substantia nigra resulted in enhanced dopaminergic neurodegeneration as well as significantly higher levels of alpha-synuclein phosphorylation at serine 129 at 4 weeks postinjection. In conclusion, our results demonstrate that total loss of PINK1 leads to an increased sensitivity to alpha-synuclein-induced neuropathology and cell death in vivo.


Molecular Therapy | 2010

Efficient gene transfer into the mouse lung by fetal intratracheal injection of rAAV2/6.2.

Marianne Carlon; Jaan Toelen; Anke Van der Perren; Luk H. Vandenberghe; Veerle Reumers; L Sbragia; Rik Gijsbers; Veerle Baekelandt; Uwe Himmelreich; James M. Wilson; Jan Deprest; Zeger Debyser

Fetal gene therapy is one of the possible new therapeutic strategies for congenital or perinatal diseases with high mortality or morbidity. We developed a novel delivery strategy to inject directly into the fetal mouse trachea. Intratracheal (i.t.) injection at embryonic day 18 (E18) was more efficient in targeting the fetal lung than conventional intra-amniotic (i.a.) delivery. Viral vectors derived from adeno-associated virus serotype 6.2, with tropism for the airway epithelium and not earlier tested in the fetal mouse lung, were injected into the fetal trachea. Bioluminescence (BL) imaging (BLI) was combined with magnetic resonance (MR) imaging (MRI) for noninvasive and accurate localization of transgene expression in vivo. Histological analysis for β-galactosidase (β-gal) revealed 17.5% of epithelial cells transduced in the conducting airways and 1.5% in the alveolar cells. Stable gene expression was observed up to 1 month after injection. This study demonstrates that direct injection of rAAV2/6.2 in the fetal mouse trachea is superior to i.a. delivery for transducing the lung. Second, as stable gene transfer was detected up to 1 postnatal month, this approach may be useful to evaluate fetal gene therapy for pulmonary diseases such as cystic fibrosis, requiring both substantial numbers of transduced cells as well as prolonged gene expression to obtain a stable phenotypic effect.


Current topics in behavioral neurosciences | 2014

Viral Vector-Based Models of Parkinson’s Disease

Anke Van der Perren; Chris Van den Haute; Veerle Baekelandt

In order to study the molecular pathways of Parkinsons disease (PD) and to develop novel therapeutic strategies, scientific investigators rely on animal models. The identification of PD-associated genes has led to the development of genetic PD models as an alternative to toxin-based models. Viral vector-mediated loco-regional gene delivery provides an attractive way to express transgenes in the central nervous system. Several vector systems based on various viruses have been developed. In this chapter, we give an overview of the different viral vector systems used for targeting the CNS. Further, we describe the different viral vector-based PD models currently available based on overexpression strategies for autosomal dominant genes such as α-synuclein and LRRK2, and knockout or knockdown strategies for autosomal recessive genes, such as parkin, DJ-1, and PINK1. Models based on overexpression of α-synuclein are the most prevalent and extensively studied, and therefore the main focus of this chapter. Many efforts have been made to increase the expression levels of α-synuclein in the dopaminergic neurons. The best α-synuclein models currently available have been developed from a combined approach using newer AAV serotypes and optimized vector constructs, production, and purification methods. These third-generation α-synuclein models show improved face and predictive validity, and therefore offer the possibility to reliably test novel therapeutics.


Journal of Neuropathology and Experimental Neurology | 2014

Long-term overexpression of human wild-type and T240R mutant Parkin in rat substantia nigra induces progressive dopaminergic neurodegeneration.

Anne-Sophie Van Rompuy; Evy Lobbestael; Anke Van der Perren; Chris Van den Haute; Veerle Baekelandt

Mutations in the parkin gene are the most common cause of early-onset autosomal recessive Parkinson disease (PD). The pathogenic mechanisms of how parkin mutations lead to the development of PD are not fully understood. Studies of cell cultures and of Drosophila have suggested a dominant negative effect for the clinical parkin mutant T240R. Conversely, the neuroprotective capacity of parkin has been widely reported; this suggests that the parkin protein may have a potential therapeutic role in PD. Here, we aimed to develop a novel genetic rodent model of PD by overexpression of T240R-parkin and human wild-type parkin as a control in the dopaminergic neurons of adult rats using adeno-associated viral vectors (rAAV2/8). Surprisingly, we found that overexpression not only of T240R-parkin but also of human wild-type parkin induced progressive and dose-dependent dopaminergic cell death in rats, starting from 8 weeks after injection. This degeneration was specific for parkin because similar overexpressionof enhanced green fluorescent protein did not lead to nigral degeneration. Our results warrant caution to the development of therapeutic strategies for PD based on overexpression of parkin or enhancing parkin activity because this might be deleterious for dopaminergic neurons in the long-term.


Clinical & Developmental Immunology | 2018

Linking Neuroinflammation and Neurodegeneration in Parkinson’s Disease

Géraldine Gelders; Veerle Baekelandt; Anke Van der Perren

Neurodegenerative diseases such as Parkinsons disease (PD) and Alzheimers disease (AD) impose a pressing burden on our developed and consequently aging society. Misfolded protein aggregates are a critical aspect of several neurodegenerative diseases. Nevertheless, several questions remain unanswered regarding the role of misfolded protein aggregates and the cause of neuronal cell death. Recently, it has been postulated that neuroinflammatory processes might play a crucial role in the pathogenesis of PD. Numerous postmortem, brain imaging, epidemiological, and animal studies have documented the involvement of the innate and adaptive immunity in neurodegeneration. Whether these inflammatory processes are directly involved in the etiology of PD or represent secondary consequences of nigrostriatal pathway injury is the subject of intensive research. Immune alterations in response to extracellular α-synuclein may play a critical role in modulating Parkinsons disease progression. In this review, we address the current concept of neuroinflammation and its involvement in PD-associated neurodegeneration.

Collaboration


Dive into the Anke Van der Perren's collaboration.

Top Co-Authors

Avatar

Veerle Baekelandt

Catholic University of Leuven

View shared research outputs
Top Co-Authors

Avatar

Zeger Debyser

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Jaan Toelen

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Chris Van den Haute

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rik Gijsbers

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Anne-Sophie Van Rompuy

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Cindy Casteels

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Francesca Macchi

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Koen Van Laere

Ghent University Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge