Network


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

Hotspot


Dive into the research topics where Maxime Caron is active.

Publication


Featured researches published by Maxime Caron.


American Journal of Human Genetics | 2013

Global Analysis of DNA Methylation Variation in Adipose Tissue from Twins Reveals Links to Disease-Associated Variants in Distal Regulatory Elements

Elin Grundberg; Eshwar Meduri; Johanna K. Sandling; Åsa K. Hedman; Sarah Keildson; Alfonso Buil; Stephan Busche; Wei Yuan; James Nisbet; Magdalena Sekowska; Alicja Wilk; Amy Barrett; Kerrin S. Small; Bing Ge; Maxime Caron; So-Youn Shin; Mark Lathrop; Emmanouil T. Dermitzakis; Mark I. McCarthy; Tim D. Spector; Jordana T. Bell; Panos Deloukas

Epigenetic modifications such as DNA methylation play a key role in gene regulation and disease susceptibility. However, little is known about the genome-wide frequency, localization, and function of methylation variation and how it is regulated by genetic and environmental factors. We utilized the Multiple Tissue Human Expression Resource (MuTHER) and generated Illumina 450K adipose methylome data from 648 twins. We found that individual CpGs had low variance and that variability was suppressed in promoters. We noted that DNA methylation variation was highly heritable (h(2)median = 0.34) and that shared environmental effects correlated with metabolic phenotype-associated CpGs. Analysis of methylation quantitative-trait loci (metQTL) revealed that 28% of CpGs were associated with nearby SNPs, and when overlapping them with adipose expression quantitative-trait loci (eQTL) from the same individuals, we found that 6% of the loci played a role in regulating both gene expression and DNA methylation. These associations were bidirectional, but there were pronounced negative associations for promoter CpGs. Integration of metQTL with adipose reference epigenomes and disease associations revealed significant enrichment of metQTL overlapping metabolic-trait or disease loci in enhancers (the strongest effects were for high-density lipoprotein cholesterol and body mass index [BMI]). We followed up with the BMI SNP rs713586, a cg01884057 metQTL that overlaps an enhancer upstream of ADCY3, and used bisulphite sequencing to refine this region. Our results showed widespread population invariability yet sequence dependence on adipose DNA methylation but that incorporating maps of regulatory elements aid in linking CpG variation to gene regulation and disease risk in a tissue-dependent manner.


Nature Genetics | 2014

Fusion of TTYH1 with the C19MC microRNA cluster drives expression of a brain-specific DNMT3B isoform in the embryonal brain tumor ETMR

Claudia L. Kleinman; Noha Gerges; Simon Papillon-Cavanagh; Patrick Sin-Chan; Albena Pramatarova; Dong Anh Khuong Quang; Véronique Adoue; Stephan Busche; Maxime Caron; Haig Djambazian; Amandine Bemmo; Adam M. Fontebasso; Tara Spence; Jeremy Schwartzentruber; Steffen Albrecht; Péter Hauser; Miklós Garami; Almos Klekner; László Bognár; Jose Luis Montes; Alfredo Staffa; Alexandre Montpetit; Pierre Bérubé; Magdalena Zakrzewska; Krzysztof Zakrzewski; Pawel P. Liberski; Zhifeng Dong; Peter M. Siegel; Thomas F. Duchaine; Christian Perotti

Embryonal tumors with multilayered rosettes (ETMRs) are rare, deadly pediatric brain tumors characterized by high-level amplification of the microRNA cluster C19MC. We performed integrated genetic and epigenetic analyses of 12 ETMR samples and identified, in all cases, C19MC fusions to TTYH1 driving expression of the microRNAs. ETMR tumors, cell lines and xenografts showed a specific DNA methylation pattern distinct from those of other tumors and normal tissues. We detected extreme overexpression of a previously uncharacterized isoform of DNMT3B originating at an alternative promoter that is active only in the first weeks of neural tube development. Transcriptional and immunohistochemical analyses suggest that C19MC-dependent DNMT3B deregulation is mediated by RBL2, a known repressor of DNMT3B. Transfection with individual C19MC microRNAs resulted in DNMT3B upregulation and RBL2 downregulation in cultured cells. Our data suggest a potential oncogenic re-engagement of an early developmental program in ETMR via epigenetic alteration mediated by an embryonic, brain-specific DNMT3B isoform.


Cell | 2016

Genetic Drivers of Epigenetic and Transcriptional Variation in Human Immune Cells

Lu Chen; Bing Ge; Francesco Paolo Casale; Louella Vasquez; Tony Kwan; Diego Garrido-Martín; Stephen Watt; Ying Yan; Kousik Kundu; Simone Ecker; Avik Datta; David C. Richardson; Frances Burden; Daniel Mead; Alice L. Mann; José María Fernández; Sophia Rowlston; Steven P. Wilder; Samantha Farrow; Xiaojian Shao; John J. Lambourne; Adriana Redensek; Cornelis A. Albers; Vyacheslav Amstislavskiy; Sofie Ashford; Kim Berentsen; Lorenzo Bomba; Guillaume Bourque; David Bujold; Stephan Busche

Summary Characterizing the multifaceted contribution of genetic and epigenetic factors to disease phenotypes is a major challenge in human genetics and medicine. We carried out high-resolution genetic, epigenetic, and transcriptomic profiling in three major human immune cell types (CD14+ monocytes, CD16+ neutrophils, and naive CD4+ T cells) from up to 197 individuals. We assess, quantitatively, the relative contribution of cis-genetic and epigenetic factors to transcription and evaluate their impact as potential sources of confounding in epigenome-wide association studies. Further, we characterize highly coordinated genetic effects on gene expression, methylation, and histone variation through quantitative trait locus (QTL) mapping and allele-specific (AS) analyses. Finally, we demonstrate colocalization of molecular trait QTLs at 345 unique immune disease loci. This expansive, high-resolution atlas of multi-omics changes yields insights into cell-type-specific correlation between diverse genomic inputs, more generalizable correlations between these inputs, and defines molecular events that may underpin complex disease risk.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Tissue factor expression provokes escape from tumor dormancy and leads to genomic alterations

Nathalie Magnus; Delphine Garnier; Brian Meehan; Serge McGraw; Tae Hoon Lee; Maxime Caron; Guillaume Bourque; Chloe Milsom; Nada Jabado; Jacquetta M. Trasler; Rafal Pawlinski; Nigel Mackman; Janusz Rak

Significance Our study shows that the clotting protein tissue factor (TF) controls the state of tumor dormancy and does so in conjunction with recruitment of inflammatory cells and blood vessels. We show that indolent glioma cells remain harmless in mice unless rendered TF positive. Our work also demonstrates the ability of TF to indirectly influence the DNA of cancer cells by facilitating gene mutations and silencing. This ability is important because injury, cardiovascular disease, or other conditions may activate the clotting system and contribute to the awakening of occult cancer cells. This understanding also may suggest a prophylactic use of blood thinners in cases where dormant cancer cells and clotting are suspected to coexist (e.g., after surgery). The coagulation system links immediate (hemostatic) and late (inflammatory, angiogenic) tissue responses to injury, a continuum that often is subverted in cancer. Here we provide evidence that tumor dormancy is influenced by tissue factor (TF), the cancer cell-associated initiator of the coagulation system and a signaling receptor. Thus, indolent human glioma cells deficient for TF remain viable but permanently dormant at the injection site for nearly a year, whereas the expression of TF leads to a step-wise transition to latent and overt tumor growth phases, a process that is preceded by recruitment of vascular (CD105+) and myeloid (CD11b+ and F4/80+) cells. Importantly, the microenvironment orchestrated by TF expression drives permanent changes in the phenotype, gene-expression profile, DNA copy number, and DNA methylation state of the tumor cells that escape from dormancy. We postulate that procoagulant events in the tissue microenvironment (niche) may affect the fate of occult tumor cells, including their biological and genetic progression to initiate a full-blown malignancy.


Nature Communications | 2015

Characterization of functional methylomes by next-generation capture sequencing identifies novel disease-associated variants.

Fiona Allum; Xiaojian Shao; Frédéric Guénard; Marie-Michelle Simon; Stephan Busche; Maxime Caron; John J. Lambourne; Julie Lessard; Karolina Tandre; Åsa K. Hedman; Tony Kwan; Bing Ge; Lars Rönnblom; Mark I. McCarthy; Panos Deloukas; Todd Richmond; Daniel Burgess; Tim D. Spector; André Tchernof; Simon Marceau; Mark Lathrop; Marie-Claude Vohl; Tomi Pastinen; Elin Grundberg

The scaling of observable properties of galaxy clusters with mass evolves with time. Assessing the role of the evolution is crucial to study the formation and evolution of massive halos and to avoid biases in the calibration. We present a general method to infer the mass and the redshift dependence, and the time-evolving intrinsic scatter of the mass–observable relations. The procedure self-calibrates the redshift dependent completeness function of the sample. The intrinsic scatter in the mass estimates used to calibrate the relation is considered too. We apply the method to the scaling of mass M∆ versus line of sight galaxy velocity dispersion σv, optical richness, X-ray luminosity, LX, and Sunyaev-Zel’dovich signal. Masses were calibrated with weak lensing measurements. The measured relations are in good agreement with time and mass dependencies predicted in the self-similar scenario of structure formation. The lone exception is the LX-M∆ relation whose time evolution is negative in agreement with formation scenarios with additional radiative cooling and uniform preheating at high redshift. The intrinsic scatter in the σv-M∆ relation is notably small, of order of 14 per cent. Robust predictions on the observed properties of the galaxy clusters in the CLASH sample are provided as cases of study. Catalogs and scripts are publicly available at http://pico.bo.astro.it/~sereno/CoMaLit/.Most genome-wide methylation studies (EWAS) of multifactorial disease traits use targeted arrays or enrichment methodologies preferentially covering CpG-dense regions, to characterize sufficiently large samples. To overcome this limitation, we present here a new customizable, cost-effective approach, methylC-capture sequencing (MCC-Seq), for sequencing functional methylomes, while simultaneously providing genetic variation information. To illustrate MCC-Seq, we use whole-genome bisulfite sequencing on adipose tissue (AT) samples and public databases to design AT-specific panels. We establish its efficiency for high-density interrogation of methylome variability by systematic comparisons with other approaches and demonstrate its applicability by identifying novel methylation variation within enhancers strongly correlated to plasma triglyceride and HDL-cholesterol, including at CD36. Our more comprehensive AT panel assesses tissue methylation and genotypes in parallel at ∼4 and ∼3 M sites, respectively. Our study demonstrates that MCC-Seq provides comparable accuracy to alternative approaches but enables more efficient cataloguing of functional and disease-relevant epigenetic and genetic variants for large-scale EWAS.


Cell Reports | 2016

The PGC-1α/ERRα Axis Represses One-Carbon Metabolism and Promotes Sensitivity to Anti-folate Therapy in Breast Cancer

Étienne Audet-Walsh; David J. Papadopoli; Simon-Pierre Gravel; Tracey Yee; Gaëlle Bridon; Maxime Caron; Guillaume Bourque; Vincent Giguère; Julie St-Pierre

Reprogramming of cellular metabolism plays a central role in fueling malignant transformation, and AMPK and the PGC-1α/ERRα axis are key regulators of this process. The intersection of gene-expression and binding-event datasets for breast cancer cells shows that activation of AMPK significantly increases the expression of PGC-1α/ERRα and promotes the binding of ERRα to its cognate sites. Unexpectedly, the data also reveal that ERRα, in concert with PGC-1α, negatively regulates the expression of several one-carbon metabolism genes, resulting in substantial perturbations in purine biosynthesis. This PGC-1α/ERRα-mediated repression of one-carbon metabolism promotes the sensitivity of breast cancer cells and tumors to the anti-folate drug methotrexate. These data implicate the PGC-1α/ERRα axis as a core regulatory node of folate cycle metabolism and further suggest that activators of AMPK could be used to modulate this pathway in cancer.


Human Molecular Genetics | 2015

High dose folic acid supplementation alters the human sperm methylome and is influenced by the MTHFR C677T polymorphism

Mahmoud Aarabi; Maria C. San Gabriel; Donovan Chan; Nathalie A. Behan; Maxime Caron; Tomi Pastinen; Guillaume Bourque; Amanda J. MacFarlane; A. Zini; Jacquetta M. Trasler

Dietary folate is a major source of methyl groups required for DNA methylation, an epigenetic modification that is actively maintained and remodeled during spermatogenesis. While high-dose folic acid supplementation (up to 10 times the daily recommended dose) has been shown to improve sperm parameters in infertile men, the effects of supplementation on the sperm epigenome are unknown. To assess the impact of 6 months of high-dose folic acid supplementation on the sperm epigenome, we studied 30 men with idiopathic infertility. Blood folate concentrations increased significantly after supplementation with no significant improvements in sperm parameters. Methylation levels of the differentially methylated regions of several imprinted loci (H19, DLK1/GTL2, MEST, SNRPN, PLAGL1, KCNQ1OT1) were normal both before and after supplementation. Reduced representation bisulfite sequencing (RRBS) revealed a significant global loss of methylation across different regions of the sperm genome. The most marked loss of DNA methylation was found in sperm from patients homozygous for the methylenetetrahydrofolate reductase (MTHFR) C677T polymorphism, a common polymorphism in a key enzyme required for folate metabolism. RRBS analysis also showed that most of the differentially methylated tiles were located in DNA repeats, low CpG-density and intergenic regions. Ingenuity Pathway Analysis revealed that methylation of promoter regions was altered in several genes involved in cancer and neurobehavioral disorders including CBFA2T3, PTPN6, COL18A1, ALDH2, UBE4B, ERBB2, GABRB3, CNTNAP4 and NIPA1. Our data reveal alterations of the human sperm epigenome associated with high-dose folic acid supplementation, effects that were exacerbated by a common polymorphism in MTHFR.


Nucleic Acids Research | 2015

Transient DNMT1 suppression reveals hidden heritable marks in the genome

Serge McGraw; Jacques X. Zhang; Mena Farag; Donovan Chan; Maxime Caron; Carolin Konermann; Christopher C. Oakes; K. Naga Mohan; Christoph Plass; Tomi Pastinen; Guillaume Bourque; J. Richard Chaillet; Jacquetta M. Trasler

Genome-wide demethylation and remethylation of DNA during early embryogenesis is essential for development. Imprinted germline differentially methylated domains (gDMDs) established by sex-specific methylation in either male or female germ cells, must escape these dynamic changes and sustain precise inheritance of both methylated and unmethylated parental alleles. To identify other, gDMD-like sequences with the same epigenetic inheritance properties, we used a modified embryonic stem (ES) cell line that emulates the early embryonic demethylation and remethylation waves. Transient DNMT1 suppression revealed gDMD-like sequences requiring continuous DNMT1 activity to sustain a highly methylated state. Remethylation of these sequences was also compromised in vivo in a mouse model of transient DNMT1 loss in the preimplantation embryo. These novel regions, possessing heritable epigenetic features similar to imprinted-gDMDs are required for normal physiological and developmental processes and when disrupted are associated with disorders such as cancer and autism spectrum disorders. This study presents new perspectives on DNA methylation heritability during early embryo development that extend beyond conventional imprinted-gDMDs.


Genome Biology | 2015

Epigenome data release: a participant-centered approach to privacy protection.

Stephanie O.M. Dyke; Warren Cheung; Yann Joly; Ole Ammerpohl; Pavlo Lutsik; Mark A. Rothstein; Maxime Caron; Stephan Busche; Guillaume Bourque; Lars Rönnblom; Paul Flicek; Stephan Beck; Martin Hirst; Henk Stunnenberg; Reiner Siebert; Jörn Walter; Tomi Pastinen

Large-scale epigenome mapping by the NIH Roadmap Epigenomics Project, the ENCODE Consortium and the International Human Epigenome Consortium (IHEC) produces genome-wide DNA methylation data at one base-pair resolution. We examine how such data can be made open-access while balancing appropriate interpretation and genomic privacy. We propose guidelines for data release that both reduce ambiguity in the interpretation of open-access data and limit immediate access to genetic variation data that are made available through controlled access.


Molecular Systems Biology | 2014

Allelic expression mapping across cellular lineages to establish impact of non-coding SNPs

Véronique Adoue; Alicia Schiavi; Nicholas Light; Jonas Carlsson Almlöf; Per Lundmark; Bing Ge; Tony Kwan; Maxime Caron; Lars Rönnblom; Chuan Wang; Shu-Huang Chen; Alison H. Goodall; François Cambien; Panos Deloukas; Willem H. Ouwehand; Ann-Christine Syvänen; Tomi Pastinen

Most complex disease‐associated genetic variants are located in non‐coding regions and are therefore thought to be regulatory in nature. Association mapping of differential allelic expression (AE) is a powerful method to identify SNPs with direct cis‐regulatory impact (cis‐rSNPs). We used AE mapping to identify cis‐rSNPs regulating gene expression in 55 and 63 HapMap lymphoblastoid cell lines from a Caucasian and an African population, respectively, 70 fibroblast cell lines, and 188 purified monocyte samples and found 40–60% of these cis‐rSNPs to be shared across cell types. We uncover a new class of cis‐rSNPs, which disrupt footprint‐derived de novo motifs that are predominantly bound by repressive factors and are implicated in disease susceptibility through overlaps with GWAS SNPs. Finally, we provide the proof‐of‐principle for a new approach for genome‐wide functional validation of transcription factor–SNP interactions. By perturbing NFκB action in lymphoblasts, we identified 489 cis‐regulated transcripts with altered AE after NFκB perturbation. Altogether, we perform a comprehensive analysis of cis‐variation in four cell populations and provide new tools for the identification of functional variants associated to complex diseases.

Collaboration


Dive into the Maxime Caron's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tony Kwan

University of Victoria

View shared research outputs
Top Co-Authors

Avatar

Serge McGraw

McGill University Health Centre

View shared research outputs
Top Co-Authors

Avatar

Panos Deloukas

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Daniel Sinnett

Université de Montréal

View shared research outputs
Top Co-Authors

Avatar

Mark Lathrop

Fondation Jean Dausset Centre d'Etude du Polymorphisme Humain

View shared research outputs
Top Co-Authors

Avatar

Chantal Richer

Université de Montréal

View shared research outputs
Researchain Logo
Decentralizing Knowledge