Network


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

Hotspot


Dive into the research topics where Caroline Van Cauwenberghe is active.

Publication


Featured researches published by Caroline Van Cauwenberghe.


Lancet Neurology | 2012

A C9orf72 promoter repeat expansion in a Flanders-Belgian cohort with disorders of the frontotemporal lobar degeneration-amyotrophic lateral sclerosis spectrum: a gene identification study

Ilse Gijselinck; Tim Van Langenhove; Julie van der Zee; Kristel Sleegers; Stéphanie Philtjens; Gernot Kleinberger; Jonathan Janssens; Karolien Bettens; Caroline Van Cauwenberghe; Sandra Pereson; Sebastiaan Engelborghs; Anne Sieben; Rik Vandenberghe; Patrick Santens; Jan De Bleecker; Githa Maes; Veerle Bäumer; Lubina Dillen; Geert Joris; Ivy Cuijt; Ellen Corsmit; Ellen Elinck; Jasper Van Dongen; Steven Vermeulen; Marleen Van den Broeck; Carolien Vaerenberg; Maria Mattheijssens; Karin Peeters; Wim Robberecht; Patrick Cras

BACKGROUND Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) are extremes of a clinically, pathologically, and genetically overlapping disease spectrum. A locus on chromosome 9p21 has been associated with both disorders, and we aimed to identify the causal gene within this region. METHODS We studied 305 patients with FTLD, 137 with ALS, and 23 with concomitant FTLD and ALS (FTLD-ALS) and 856 controls from Flanders (Belgium); patients were identified from a hospital-based cohort and were negative for mutations in known FTLD and ALS genes. We also examined the family of one patient with FTLD-ALS previously linked to 9p21 (family DR14). We analysed 130 kbp at 9p21 in association and segregation studies, genomic sequencing, repeat genotyping, and expression studies to identify the causal mutation. We compared genotype-phenotype correlations between mutation carriers and non-carriers. FINDINGS In the patient-control cohort, the single-nucleotide polymorphism rs28140707 within the 130 kbp region of 9p21 was associated with disease (odds ratio [OR] 2·6, 95% CI 1·5-4·7; p=0·001). A GGGGCC repeat expansion in C9orf72 completely co-segregated with disease in family DR14. The association of rs28140707 with disease in the patient-control cohort was abolished when we excluded GGGGCC repeat expansion carriers. In patients with familial disease, six (86%) of seven with FTLD-ALS, seven (47%) of 15 with ALS, and 12 (16%) of 75 with FTLD had the repeat expansion. In patients without known familial disease, one (6%) of 16 with FTLD-ALS, six (5%) of 122 with ALS, and nine (4%) of 230 with FTLD had the repeat expansion. Mutation carriers primarily presented with classic ALS (10 of 11 individuals) or behavioural variant FTLD (14 of 15 individuals). Mean age at onset of FTLD was 55·3 years (SD 8·4) in 21 mutation carriers and 63·2 years (9·6) in 284 non-carriers (p=0·001); mean age at onset of ALS was 54·5 years (9·9) in 13 carriers and 60·4 years (11·4) in 124 non-carriers. Postmortem neuropathological analysis of the brains of three mutation carriers with FTLD showed a notably low TDP-43 load. In brain at postmortem, C9orf72 expression was reduced by nearly 50% in two carriers compared with nine controls (p=0·034). In familial patients, 14% of FTLD-ALS, 50% of ALS, and 62% of FTLD was not accounted for by known disease genes. INTERPRETATION We identified a pathogenic GGGGCC repeat expansion in C9orf72 on chromosome 9p21, as recently also reported in two other studies. The GGGGCC repeat expansion is highly penetrant, explaining all of the contribution of chromosome 9p21 to FTLD and ALS in the Flanders-Belgian cohort. Decreased expression of C9orf72 in brain suggests haploinsufficiency as an underlying disease mechanism. Unidentified genes probably also contribute to the FTLD-ALS disease spectrum. FUNDING Full funding sources listed at end of paper (see Acknowledgments).


Genetics in Medicine | 2016

The genetic landscape of Alzheimer disease: clinical implications and perspectives

Caroline Van Cauwenberghe; Christine Van Broeckhoven; Kristel Sleegers

The search for the genetic factors contributing to Alzheimer disease (AD) has evolved tremendously throughout the years. It started from the discovery of fully penetrant mutations in Amyloid precursor protein, Presenilin 1, and Presenilin 2 as a cause of autosomal dominant AD, the identification of the ɛ4 allele of Apolipoprotein E as a strong genetic risk factor for both early-onset and late-onset AD, and evolved to the more recent detection of at least 21 additional genetic risk loci for the genetically complex form of AD emerging from genome-wide association studies and massive parallel resequencing efforts. These advances in AD genetics are positioned in light of the current endeavor directing toward translational research and personalized treatment of AD. We discuss the current state of the art of AD genetics and address the implications and relevance of AD genetics in clinical diagnosis and risk prediction, distinguishing between monogenic and multifactorial AD. Furthermore, the potential and current limitations of molecular reclassification of AD to streamline clinical trials in drug development and biomarker studies are addressed.Genet Med 18 5, 421–430.


Lancet Neurology | 2015

Mutations in ABCA7 in a Belgian cohort of Alzheimer's disease patients: a targeted resequencing study

Elise Cuyvers; Arne De Roeck; Tobi Van den Bossche; Caroline Van Cauwenberghe; Karolien Bettens; Steven Vermeulen; Maria Mattheijssens; Karin Peeters; Sebastiaan Engelborghs; Mathieu Vandenbulcke; Rik Vandenberghe; Peter Paul De Deyn; Christine Van Broeckhoven; Kristel Sleegers

BACKGROUND ABCA7 was identified as a risk gene for Alzheimers disease in genome-wide association studies (GWAS). It was one of the genes most strongly associated with risk of Alzheimers disease in a Belgian cohort. Using targeted resequencing, we investigated ABCA7 in this cohort with the aim to directly detect rare and common variations in this gene associated with Alzheimers disease pathogenesis. METHODS We did massive parallel resequencing of ABCA7 after HaloPlex target enrichment of the exons, introns, and regulatory regions in 772 unrelated patients with Alzheimers disease (mean age at onset 74·6 years [SD 8·9]) recruited at two memory clinics in Flanders, Belgium, and 757 geographically matched community-dwelling controls (mean age at inclusion 73·9 years [8·0]). After bioinformatic processing, common variants were analysed with conditional logistic regression and rare variant association analysis was done in Variant Association Tools. To explore an observed founder effect, additional unrelated patients with Alzheimers disease (n=183, mean age at onset 78·8 years [SD 6·0]) and control individuals (n=265, mean age at inclusion 56·9 years [10·8]) from the same cohort who had not been included in massive parallel resequencing because of insufficient biosamples were screened for the ABCA7 frameshift mutation Glu709fs with Sanger sequencing. The effect of loss-of-function mutations on ABCA7 expression was investigated with quantitative real-time PCR in post-mortem brains of patients (n=3) and control individuals (n=4); nonsense mediated mRNA decay was investigated in lymphoblast cell lines from three predicted loss-of-function mutation carriers from the cohort of 772 patients with Alzheimers disease. FINDINGS An intronic low-frequency variant rs78117248 (minor allele frequency 3·8% in 58 patients with Alzheimers disease and in controls 1·8% in 28 controls) showed strongest association with Alzheimers disease (odds ratio 2·07, 95% CI 1·31-3·27; p=0·0016), and remained significant after conditioning for the GWAS top single nucleotide polymorphisms rs3764650, rs4147929, and rs3752246 (2·00, 1·22-3·26; p=0·006). We identified an increased frequency of predicted loss-of-function mutations in the patients compared with the controls (relative risk 4·03, 95% CI 1·75-9·29; p=0·0002). One frameshift mutation (Glu709fs) showed a founder effect in the study population, and was found to segregate with disease in a family with autosomal dominant inheritance of Alzheimers disease. Expression of ABCA7 was reduced in the two carriers of loss-of-function mutations found only in patients with Alzheimers disease (Glu709fs and Trp1214*) compared with four non-carrier controls (relative expression 0·45, 95% CI 0·25-0·84; p=0·002) and in lymphoblast cell lines from three carriers of Glu709fs compared with those from two non-carrier controls. INTERPRETATION We propose that a low-frequency variant can explain the association between ABCA7 and Alzheimers disease, and the evidence of loss-of-function mutations in this risk gene suggests that partial loss-of-function of ABCA7 could be a potential pathogenetic mechanism of Alzheimers disease. FUNDING Belgian Science Policy Office Interuniversity Attraction Poles program P7/16, Alzheimer Research Foundation, King Baudouin Foundation AB Fund, Methusalem Excellence Program initiative of the Flemish Government, Flanders Impulse Program on Networks for Dementia Research, Research Foundation Flanders, Agency for Innovation by Science and Technology Flanders, University of Antwerp Research Fund, and European Unions Seventh Framework Programme for Research, Technological development and Demonstration (AgedBrainSYSBIO).


Neurobiology of Aging | 2012

Genetic association of CR1 with Alzheimer's disease: A tentative disease mechanism

Lili-Naz Hazrati; Caroline Van Cauwenberghe; Patricia L. Brooks; Nathalie Brouwers; Mahdi Ghani; Christine Sato; Marc Cruts; Kristel Sleegers; Peter St George-Hyslop; Christine Van Broeckhoven; Ekaterina Rogaeva

CR1 is a novel Alzheimers disease (AD) gene identified by genome-wide association studies (GWAS). Recently, we showed that AD risk could be explained by an 18-kilobase insertion responsible for the complement component (3b/4b) receptor 1 (CR1)-S isoform. We investigated the relevance of the CR1 isoforms to AD in a Canadian dataset. Also, we genotyped rs4844610 tagging the GWAS-significant CR1 single nucleotide polymorphisms. Individuals with F/S genotype had a 1.8 times increased risk for AD compared with F/F genotype (p-adjusted = 0.003), while rs4844610 was only marginally significant (p-adjusted = 0.024). The analyses of brain samples demonstrated that the CR1-S isoform is expressed at lower protein levels than CR1-F (p < 0.0001) hence likely associated with increased complement activation. Intriguingly, our neuropathological results show that the pattern of CR1 expression in neurons is different between the F/F and F/S genotypes (filiform vs. vesicular-like profiles). Furthermore, double labeling studies supported a differential distribution of CR1 in neurons (endoplasmic reticulum intermediate compartment vs. lysosomes). These observations indicate that the CR1-S and CR1-F isoforms could be processed in different ways in neurons. In conclusion, our results support that the CR1-S isoform explains the GWAS signals and open a novel prospect for the investigation of CR1-related disease mechanisms.


Neurobiology of Aging | 2013

C9orf72 G4C2 repeat expansions in Alzheimer's disease and mild cognitive impairment.

Rita Cacace; Caroline Van Cauwenberghe; Karolien Bettens; Ilse Gijselinck; Julie van der Zee; Sebastiaan Engelborghs; Mathieu Vandenbulcke; Jasper Van Dongen; Veerle Bäumer; Lubina Dillen; Maria Mattheijssens; Karin Peeters; Marc Cruts; Rik Vandenberghe; Peter Paul De Deyn; Christine Van Broeckhoven; Kristel Sleegers

C9orf72 G4C2 repeat expansion is a major cause of amyotrophic lateral sclerosis and frontotemporal lobar degeneration. Its role in Alzheimers disease (AD) is less clear. We assessed the prevalence of G4C2 pathogenic repeat expansions in Flanders-Belgian patients with clinical AD or mild cognitive impairment (MCI). In addition, we studied the effect of non-pathogenic G4C2 repeat length variability on susceptibility to AD, and on AD cerebrospinal fluid (CSF) biomarker levels. A pathogenic repeat expansion was identified in 5 of 1217 AD patients (frequency <1%). No pathogenic expansions were observed in patients with MCI (n = 200) or control individuals (n = 1119). Nonpathogenic repeat length variability was not associated with AD, risk of conversion to AD in MCI individuals, or CSF biomarker levels. We conclude that pathogenic C9orf72 G4C2 repeat expansions can be detected in clinical AD patients and could act as a contributor to AD pathogenesis. Non-pathogenic repeat length variability did not affect risk of AD or MCI, nor AD biomarker levels in CSF, indicating that C9orf72 is not a direct AD risk factor.


Alzheimers & Dementia | 2015

A 22-single nucleotide polymorphism Alzheimer's disease risk score correlates with family history, onset age, and cerebrospinal fluid Aβ42

Kristel Sleegers; Karolien Bettens; Arne De Roeck; Caroline Van Cauwenberghe; Elise Cuyvers; Jan Verheijen; Hanne Struyfs; Jasper Van Dongen; Steven Vermeulen; Sebastiaan Engelborghs; Mathieu Vandenbulcke; Rik Vandenberghe; Peter Paul De Deyn; Christine Van Broeckhoven

The ability to identify individuals at increased genetic risk for Alzheimers disease (AD) may streamline biomarker and drug trials and aid clinical and personal decision making.


Neurobiology of Aging | 2013

Complement receptor 1 coding variant p.Ser1610Thr in Alzheimer's disease and related endophenotypes

Caroline Van Cauwenberghe; Karolien Bettens; Sebastiaan Engelborghs; Mathieu Vandenbulcke; Jasper Van Dongen; Steven Vermeulen; Rik Vandenberghe; Peter Paul De Deyn; Christine Van Broeckhoven; Kristel Sleegers

We previously described an intragenic functional copy number variation (CNV) in complement receptor 1 (CR1) that is associated with Alzheimer disease (AD) risk. A recent study, however, reported a rare CR1 coding variant p.Ser1610Thr (rs4844609) associated with AD susceptibility, explaining the effect of genome wide association (GWA) top single nucleotide polymorphism rs6656401. We assessed the role of the Ser1610Thr variant in AD pathogenesis and the effect on AD-related endophenotypes in a Flanders-Belgian cohort. We evaluated whether this rare variant rather than the CR1 CNV could explain the association of CR1 in our population. The Ser1610Thr variant was not associated with AD, memory impairment, total tau, amyloid β(1-42) or tau phosphorylated at threonine 181 levels. It did not explain (part of) the association of genome wide association top single-nucleotide polymorphisms rs3818361/rs6656401, nor of the CR1 CNV, with AD in our cohort, whereas the CR1 CNV and rs3818361/rs6656401 represented the same association signal. These findings question a role for the Ser1610Thr variant in AD risk and related endophenotypes, and reaffirm our previous observation that the CR1 CNV could be the true functional risk factor explaining the association between CR1 and AD.


Molecular Neurodegeneration | 2015

Reduced secreted clusterin as a mechanism for Alzheimer-associated CLU mutations

Karolien Bettens; Steven Vermeulen; Caroline Van Cauwenberghe; Bavo Heeman; Bob Asselbergh; Caroline Robberecht; Sebastiaan Engelborghs; Mathieu Vandenbulcke; Rik Vandenberghe; Peter Paul De Deyn; Marc Cruts; Christine Van Broeckhoven; Kristel Sleegers

BackgroundThe clusterin (CLU) gene has been identified as an important risk locus for Alzheimer’s disease (AD). Although the actual risk–increasing polymorphisms at this locus remain to be identified, we previously observed an increased frequency of rare non-synonymous mutations and small insertion-deletions of CLU in AD patients, which specifically clustered in the β-chain domain of CLU. Nonetheless the pathogenic nature of these variants remained unclear.Here we report a novel non-synonymous CLU mutation (p.I360N) in a Belgian Alzheimer patient and have explored the pathogenic nature of this and 10 additional CLU mutations on protein localization and secretion in vitro using immunocytochemistry, immunodetection and ELISAs.ResultsThree patient-specific CLU mutations in the β-chain (p.I303NfsX13, p.R338W and p.I360N) caused an alteration of the subcellular CLU localization and diminished CLU transport through the secretory pathway, indicative of possible degradation mechanisms. For these mutations, significantly reduced CLU intensity was observed in the Golgi while almost all CLU protein was exclusively present in the endoplasmic reticulum. This was further confirmed by diminished CLU secretion in HEK293T and HEK293 FLp-In cell lines.ConclusionsOur data lend further support to the contribution of rare coding CLU mutations in the pathogenesis of neurodegenerative diseases. Functional analyses suggest reduced secretion of the CLU protein as the mode of action for three of the examined CLU mutations. One of those is a frameshift mutation leading to a loss of secreted protein, and the other two mutations are amino acid substitutions in the disulfide bridge region, possibly interfering with heterodimerization of the α- and β-chain of CLU.


Neurology | 2016

Phenotypic characteristics of Alzheimer patients carrying an ABCA7 mutation.

Tobi Van den Bossche; Kristel Sleegers; Elise Cuyvers; Sebastiaan Engelborghs; Anne Sieben; Arne De Roeck; Caroline Van Cauwenberghe; Steven Vermeulen; Marleen Van den Broeck; Annelies Laureys; Karin Peeters; Maria Mattheijssens; Mathieu Vandenbulcke; Rik Vandenberghe; Jean-Jacques Martin; Peter Paul De Deyn; Patrick Cras; Christine Van Broeckhoven

Objective: To generate a clinical and pathologic phenotype of patients carrying rare loss-of-function mutations in ABCA7, identified in a Belgian Alzheimer patient cohort and in an autosomal dominant family. Methods: We performed a retrospective review of available data records, medical records, results of CSF analyses and neuroimaging studies, and neuropathology data. Results: The mean onset age of the mutation carriers (n = 22) was 73.4 ± 8.4 years with a wide age range of 36 (54–90) years, which was independent of APOE genotype and cerebrovascular disease. The mean disease duration was 5.7 ± 3.0 years (range 2–12 years). A positive family history was recorded for 10 carriers (45.5%). All patient carriers except one presented with memory complaints. The 4 autopsied brains showed typical immunohistochemical changes of late-onset Alzheimer disease. Conclusions: All patients carrying a loss-of-function mutation in ABCA7 exhibited a classical Alzheimer disease phenotype, though with a striking wide onset age range, suggesting the influence of unknown modifying factors.


Molecular Oncology | 2015

Decreased expression of ABAT and STC2 hallmarks ER‐positive inflammatory breast cancer and endocrine therapy resistance in advanced disease

Maurice P.H.M. Jansen; Leen Sas; Anieta M. Sieuwerts; Caroline Van Cauwenberghe; Diana Ramirez-Ardila; Maxime P. Look; Kirsten Ruigrok-Ritstier; Pascal Finetti; François Bertucci; Mieke M. Timmermans; Carolien H.M. van Deurzen; John W. M. Martens; Iris Simon; Paul Roepman; Sabine C. Linn; Peter van Dam; Marleen Kok; Filip Lardon; Peter B. Vermeulen; John A. Foekens; Luc Dirix; Els M. J. J. Berns; Steven Van Laere

Patients with Estrogen Receptor α‐positive (ER+) Inflammatory Breast Cancer (IBC) are less responsive to endocrine therapy compared with ER+ non‐IBC (nIBC) patients. The study of ER+ IBC samples might reveal biomarkers for endocrine resistant breast cancer.

Collaboration


Dive into the Caroline Van Cauwenberghe's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rik Vandenberghe

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mathieu Vandenbulcke

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge