Network


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

Hotspot


Dive into the research topics where Chantal Bazenet is active.

Publication


Featured researches published by Chantal Bazenet.


Alzheimers & Dementia | 2014

The future of blood-based biomarkers for Alzheimer's disease

Kim Henriksen; Sid E. O’Bryant; Harald Hampel; John Q. Trojanowski; Thomas J. Montine; Andreas Jeromin; Kaj Blennow; Anders Lönneborg; Tony Wyss-Coray; Holly Soares; Chantal Bazenet; Magnus Sjögren; William T. Hu; Simon Lovestone; Morten A. Karsdal; Michael W. Weiner

Treatment of Alzheimers disease (AD) is significantly hampered by the lack of easily accessible biomarkers that can detect disease presence and predict disease risk reliably. Fluid biomarkers of AD currently provide indications of disease stage; however, they are not robust predictors of disease progression or treatment response, and most are measured in cerebrospinal fluid, which limits their applicability. With these aspects in mind, the aim of this article is to underscore the concerted efforts of the Blood‐Based Biomarker Interest Group, an international working group of experts in the field. The points addressed include: (1) the major challenges in the development of blood‐based biomarkers of AD, including patient heterogeneity, inclusion of the “right” control population, and the blood–brain barrier; (2) the need for a clear definition of the purpose of the individual markers (e.g., prognostic, diagnostic, or monitoring therapeutic efficacy); (3) a critical evaluation of the ongoing biomarker approaches; and (4) highlighting the need for standardization of preanalytical variables and analytical methodologies used by the field.


Alzheimers & Dementia | 2014

Plasma proteins predict conversion to dementia from prodromal disease

Abdul Hye; Alison L. Baird; Nicholas J. Ashton; Chantal Bazenet; Rufina Leung; Eric Westman; Andrew Simmons; Richard Dobson; Martina Sattlecker; Michelle K. Lupton; Katie Lunnon; Aoife Keohane; Malcolm Ward; Hans Dieter Zucht; Danielle Pepin; Wei Zheng; Alan Tunnicliffe; Jill C. Richardson; Serge Gauthier; Hilkka Soininen; Iwona Kloszewska; Patrizia Mecocci; Magda Tsolaki; Bruno Vellas; Simon Lovestone

The study aimed to validate previously discovered plasma biomarkers associated with AD, using a design based on imaging measures as surrogate for disease severity and assess their prognostic value in predicting conversion to dementia.


Journal of Alzheimer's Disease | 2013

Candidate Blood Proteome Markers of Alzheimer's Disease Onset and Progression: A Systematic Review and Replication Study

Steven John Kiddle; Martina Sattlecker; Petroula Proitsi; Andrew Simmons; Eric Westman; Chantal Bazenet; Sally K. Nelson; Stephen E. Williams; Angela Hodges; Caroline Johnston; Hilkka Soininen; Iwona Kloszewska; Patrizia Mecocci; Magda Tsolaki; Bruno Vellas; Stephen Newhouse; Simon Lovestone; Richard Dobson

A blood-based protein biomarker, or set of protein biomarkers, that could predict onset and progression of Alzheimers disease (AD) would have great utility; potentially clinically, but also for clinical trials and especially in the selection of subjects for preventative trials. We reviewed a comprehensive list of 21 published discovery or panel-based (> 100 proteins) blood proteomics studies of AD, which had identified a total of 163 candidate biomarkers. Few putative blood-based protein biomarkers replicate in independent studies but we found that some proteins do appear in multiple studies; for example, four candidate biomarkers are found to associate with AD-related phenotypes in five independent research cohorts in these 21 studies: α-1-antitrypsin, α-2-macroglobulin, apolipoprotein E, and complement C3. Using SomaLogics SOMAscan proteomics technology, we were able to conduct a large-scale replication study for 94 of the 163 candidate biomarkers from these 21 published studies in plasma samples from 677 subjects from the AddNeuroMed (ANM) and the Alzheimers Research UK/Maudsley BRC Dementia Case Registry at Kings Health Partners (ARUK/DCR) research cohorts. Nine of the 94 previously reported candidates were found to associate with AD-related phenotypes (False Discovery Rate (FDR) q-value < 0.1). These proteins show sufficient replication to be considered for further investigation as a biomarker set. Overall, we show that there are some signs of a replicable signal in the range of proteins identified in previous studies and we are able to further replicate some of these. This suggests that AD pathology does affect the blood proteome with some consistency.


Molecular Psychiatry | 2014

Clusterin regulates β-amyloid toxicity via Dickkopf-1-driven induction of the wnt–PCP–JNK pathway

Richard Killick; Elena M. Ribe; Raya Al-Shawi; Bilal Malik; Claudie Hooper; Cathy Fernandes; Richard Dobson; Nolan Pm; Anbarasu Lourdusamy; Simon J. Furney; Kuang Lin; Gerome Breen; Richard Wroe; Alvina W.M. To; Karelle Leroy; Mirsada Causevic; Alessia Usardi; Robinson M; Wendy Noble; Richard Williamson; Katie Lunnon; Stuart Kellie; Christopher Hugh Reynolds; Chantal Bazenet; Angela Hodges; Jean Pierre Brion; John Stephenson; Simons Jp; Simon Lovestone

Although the mechanism of Aβ action in the pathogenesis of Alzheimer’s disease (AD) has remained elusive, it is known to increase the expression of the antagonist of canonical wnt signalling, Dickkopf-1 (Dkk1), whereas the silencing of Dkk1 blocks Aβ neurotoxicity. We asked if clusterin, known to be regulated by wnt, is part of an Aβ/Dkk1 neurotoxic pathway. Knockdown of clusterin in primary neurons reduced Aβ toxicity and DKK1 upregulation and, conversely, Aβ increased intracellular clusterin and decreased clusterin protein secretion, resulting in the p53-dependent induction of DKK1. To further elucidate how the clusterin-dependent induction of Dkk1 by Aβ mediates neurotoxicity, we measured the effects of Aβ and Dkk1 protein on whole-genome expression in primary neurons, finding a common pathway suggestive of activation of wnt–planar cell polarity (PCP)–c-Jun N-terminal kinase (JNK) signalling leading to the induction of genes including EGR1 (early growth response-1), NAB2 (Ngfi-A-binding protein-2) and KLF10 (Krüppel-like factor-10) that, when individually silenced, protected against Aβ neurotoxicity and/or tau phosphorylation. Neuronal overexpression of Dkk1 in transgenic mice mimicked this Aβ-induced pathway and resulted in age-dependent increases in tau phosphorylation in hippocampus and cognitive impairment. Furthermore, we show that this Dkk1/wnt–PCP–JNK pathway is active in an Aβ-based mouse model of AD and in AD brain, but not in a tau-based mouse model or in frontotemporal dementia brain. Thus, we have identified a pathway whereby Aβ induces a clusterin/p53/Dkk1/wnt–PCP–JNK pathway, which drives the upregulation of several genes that mediate the development of AD-like neuropathologies, thereby providing new mechanistic insights into the action of Aβ in neurodegenerative diseases.


Biomarkers in Medicine | 2012

Plasma biomarkers for Alzheimer’s disease: much needed but tough to find

Chantal Bazenet; Simon Lovestone

Alzheimers disease is a complex age-dependent neurodegenerative disease where definitive diagnosis is only possible after autopsy and where there is a long prodromal or preclinical phase. Biomarkers for both early diagnosis and prediction of disease progression are needed and extensive efforts to discover them have been undertaken. In this article, we have attempted to summarize the findings of current studies using proteomics and metabolomics approaches. We are also discussing how the use of emerging technologies and better study designs can support the identification of the much-needed Alzheimers disease plasma biomarkers.


Alzheimer's & Dementia: Diagnosis, Assessment & Disease Monitoring | 2015

Blood protein predictors of brain amyloid for enrichment in clinical trials

Nicholas J. Ashton; Steven John Kiddle; John Frederick Graf; Malcolm Ward; Alison L. Baird; Abdul Hye; Sarah Westwood; Karyuan Vivian Wong; Richard Dobson; Gil D. Rabinovici; Bruce L. Miller; Howard J. Rosen; Andrew Soliz Torres; Zhanpan Zhang; Lennart Thurfjell; Antonia Covin; Cristina Tan Hehir; David Baker; Chantal Bazenet; Simon Lovestone

Measures of neocortical amyloid burden (NAB) identify individuals who are at substantially greater risk of developing Alzheimers disease (AD). Blood‐based biomarkers predicting NAB would have great utility for the enrichment of AD clinical trials, including large‐scale prevention trials.


Journal of Alzheimer's Disease | 2015

Blood Protein Markers of Neocortical Amyloid-β Burden: A Candidate Study Using SOMAscan Technology.

Nicola Voyle; David Baker; Samantha Burnham; Antonia Covin; Zhanpan Zhang; Dipen P. Sangurdekar; Cristina Tan Hehir; Chantal Bazenet; Simon Lovestone; Steven John Kiddle

Abstract Background: Four previously reported studies have tested for association of blood proteins with neocortical amyloid-β burden (NAB). If shown to be robust, these proteins could have utility as a blood test for enrichment in clinical trials of Alzheimer’s disease (AD) therapeutics. Objective: This study aimed to investigate whether previously identified blood proteins also show evidence for association with NAB in serum samples from the Australian Imaging, Biomarker and Lifestyle Flagship Study of Ageing (AIBL). The study considers candidate proteins seen in cohorts other than AIBL and candidates previously discovered in the AIBL cohort. Methods: Our study used the SOMAscan platform for protein quantification in blood serum. Linear and logistic regressions were used to model continuous NAB and dichotomized NAB respectively using single proteins as a predictor. Multiple protein models were built using stepwise regression techniques and support vectors machines. Age and APOE ɛ4 carriage were used as covariates for all analysis. Results: Of the 41 proteins previously reported, 15 AIBL candidates and 20 non-AIBL candidates were available for testing. Of these candidates, pancreatic polypeptide (PPY) and IgM showed a significant association with NAB. Notably, IgM was found to associate with continuous NAB across cognitively normal control subjects. Conclusions: We have further demonstrated the association of PPY and IgM with NAB, despite technical differences between studies. There are several reasons for a lack of significance for the other candidates including platform differences and the use of serum rather than plasma samples. To investigate the possibility of technical differences causing lack of replication, further studies are required.


Journal of Alzheimer's Disease | 2016

Blood-Based Biomarker Candidates of Cerebral Amyloid Using PiB PET in Non-Demented Elderly

Sarah Westwood; Emanuela Leoni; Abdul Hye; Steven Lynham; Mizanur Khondoker; Nicholas J. Ashton; Steven John Kiddle; Alison L. Baird; Ricardo Sainz-Fuertes; Rufina Leung; John Frederick Graf; Cristina Tan Hehir; David Baker; Cristina Cereda; Chantal Bazenet; Malcolm Ward; Madhav Thambisetty; Simon Lovestone

Increasingly, clinical trials for Alzheimers disease (AD) are being conducted earlier in the disease phase and with biomarker confirmation using in vivo amyloid PET imaging or CSF tau and Aβ measures to quantify pathology. However, making such a pre-clinical AD diagnosis is relatively costly and the screening failure rate is likely to be high. Having a blood-based marker that would reduce such costs and accelerate clinical trials through identifying potential participants with likely pre-clinical AD would be a substantial advance. In order to seek such a candidate biomarker, discovery phase proteomic analyses using 2DGE and gel-free LC-MS/MS for high and low molecular weight analytes were conducted on longitudinal plasma samples collected over a 12-year period from non-demented older individuals who exhibited a range of 11C-PiB PET measures of amyloid load. We then sought to extend our discovery findings by investigating whether our candidate biomarkers were also associated with brain amyloid burden in disease, in an independent cohort. Seven plasma proteins, including A2M, Apo-A1, and multiple complement proteins, were identified as pre-clinical biomarkers of amyloid burden and were consistent across three time points (p <  0.05). Five of these proteins also correlated with brain amyloid measures at different stages of the disease (q <  0.1). Here we show that it is possible to detect a plasma based biomarker signature indicative of AD pathology at a stage long before the onset of clinical disease manifestation. As in previous studies, acute phase reactants and inflammatory markers dominate this signature.


Translational Psychiatry | 2015

Plasma protein biomarkers of Alzheimer’s disease endophenotypes in asymptomatic older twins: early cognitive decline and regional brain volumes

Steven John Kiddle; Claire J. Steves; Mitul A. Mehta; A Simmons; Xiaohui Xu; Stephen Newhouse; Martina Sattlecker; Nicholas J. Ashton; Chantal Bazenet; Richard Killick; Jihad Adnan; Eric Westman; Sally K. Nelson; Hilkka Soininen; Iwona Kloszewska; P. Mecocci; Magdalini Tsolaki; Bruno Vellas; Charles Curtis; Gerome Breen; Steven Williams; Simon Lovestone; Tim D. Spector; Richard Dobson

There is great interest in blood-based markers of Alzheimer’s disease (AD), especially in its pre-symptomatic stages. Therefore, we aimed to identify plasma proteins whose levels associate with potential markers of pre-symptomatic AD. We also aimed to characterise confounding by genetics and the effect of genetics on blood proteins in general. Panel-based proteomics was performed using SOMAscan on plasma samples from TwinsUK subjects who are asymptomatic for AD, measuring the level of 1129 proteins. Protein levels were compared with 10-year change in CANTAB-paired associates learning (PAL; n=195), and regional brain volumes (n=34). Replication of proteins associated with regional brain volumes was performed in 254 individuals from the AddNeuroMed cohort. Across all the proteins measured, genetic factors were found to explain ~26% of the variability in blood protein levels on average. The plasma level of the mitogen-activated protein kinase (MAPK) MAPKAPK5 protein was found to positively associate with the 10-year change in CANTAB-PAL in both the individual and twin difference context. The plasma level of protein MAP2K4 was found to suggestively associate negatively (Q<0.1) with the volume of the left entorhinal cortex. Future studies will be needed to assess the specificity of MAPKAPK5 and MAP2K4 to eventual conversion to AD.


Translational Psychiatry | 2016

Blood metabolite markers of neocortical amyloid-β burden: discovery and enrichment using candidate proteins.

Nicola Voyle; Min Kim; Petroula Proitsi; Nicholas J. Ashton; Alison L. Baird; Chantal Bazenet; Abdul Hye; Sarah Westwood; Raymond T. Chung; Malcolm Ward; Gil D. Rabinovici; Simon Lovestone; Gerome Breen; Cristina Legido-Quigley; Richard Dobson; Steven John Kiddle

We believe this is the first study to investigate associations between blood metabolites and neocortical amyloid burden (NAB) in the search for a blood-based biomarker for Alzheimers disease (AD). Further, we present the first multi-modal analysis of blood markers in this field. We used blood plasma samples from 91 subjects enrolled in the University of California, San Francisco Alzheimers Disease Research Centre. Non-targeted metabolomic analysis was used to look for associations with NAB using both single and multiple metabolic feature models. Five metabolic features identified subjects with high NAB, with 72% accuracy. We were able to putatively identify four metabolites from this panel and improve the model further by adding fibrinogen gamma chain protein measures (accuracy=79%). One of the five metabolic features was studied in the Alzheimers Disease Neuroimaging Initiative cohort, but results were inconclusive. If replicated in larger, independent studies, these metabolic features and proteins could form the basis of a blood test with potential for enrichment of amyloid pathology in anti-amyloid trials.

Collaboration


Dive into the Chantal Bazenet's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Abdul Hye

King's College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge