Network


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

Hotspot


Dive into the research topics where Annaick Carles is active.

Publication


Featured researches published by Annaick Carles.


Blood | 2013

Analysis of the clonal growth and differentiation dynamics of primitive barcoded human cord blood cells in NSG mice

Alice M.S. Cheung; Long V. Nguyen; Annaick Carles; Philip A. Beer; Paul H. Miller; David J.H.F. Knapp; Kiran Dhillon; Martin Hirst; Connie J. Eaves

Human cord blood (CB) offers an attractive source of cells for clinical transplants because of its rich content of cells with sustained repopulating ability in spite of an apparent deficiency of cells with rapid reconstituting ability. Nevertheless, the clonal dynamics of nonlimiting CB transplants remain poorly understood. To begin to address this question, we exposed CD34+ CB cells to a library of barcoded lentiviruses and used massively parallel sequencing to quantify the clonal distributions of lymphoid and myeloid cells subsequently detected in sequential marrow aspirates obtained from 2 primary NOD/SCID-IL2Rγ(-/-) mice, each transplanted with ∼10(5) of these cells, and for another 6 months in 2 secondary recipients. Of the 196 clones identified, 68 were detected at 4 weeks posttransplant and were often lympho-myeloid. The rest were detected later, after variable periods up to 13 months posttransplant, but with generally increasing stability throughout time, and they included clones in which different lineages were detected. However, definitive evidence of individual cells capable of generating T-, B-, and myeloid cells, for over a year, and self-renewal of this potential was also obtained. These findings highlight the caveats and utility of this model to analyze human hematopoietic stem cell control in vivo.


Cell Stem Cell | 2014

Clonal Analysis via Barcoding Reveals Diverse Growth and Differentiation of Transplanted Mouse and Human Mammary Stem Cells

Long V. Nguyen; Maisam Makarem; Annaick Carles; Michelle Moksa; Nagarajan Kannan; Pawan Pandoh; Peter Eirew; Tomo Osako; Melanie D. Kardel; Alice M.S. Cheung; William Kennedy; Kane Tse; Thomas Zeng; Yongjun Zhao; R. Keith Humphries; Samuel Aparicio; Connie J. Eaves; Martin Hirst

Cellular barcoding offers a powerful approach to characterize the growth and differentiation activity of large numbers of cotransplanted stem cells. Here, we describe a lentiviral genomic-barcoding and analysis strategy and its use to compare the clonal outputs of transplants of purified mouse and human basal mammary epithelial cells. We found that both sources of transplanted cells produced many bilineage mammary epithelial clones in primary recipients, although primary clones containing only one detectable mammary lineage were also common. Interestingly, regardless of the species of origin, many clones evident in secondary recipients were not detected in the primary hosts, and others that were changed from appearing luminal-restricted to appearing bilineage. This barcoding methodology has thus revealed conservation between mice and humans of a previously unknown diversity in the growth and differentiation activities of their basal mammary epithelial cells stimulated to grow in transplanted hosts.


Development | 2013

E-cadherin is required for the proper activation of the Lifr/Gp130 signaling pathway in mouse embryonic stem cells

Ignacio del Valle; Stefan Rudloff; Annaick Carles; Yong Li; Ewa Liszewska; Riana Vogt; Rolf Kemler

The leukemia inhibitory factor (Lif) signaling pathway is a crucial determinant for mouse embryonic stem (mES) cell self-renewal and pluripotency. One of the hallmarks of mES cells, their compact growth morphology, results from tight cell adhesion mediated through E-cadherin, β-catenin (Ctnnb1) and α-catenin with the actin cytoskeleton. β-catenin is also involved in canonical Wnt signaling, which has also been suggested to control mES cell stemness. Here, we analyze Ctnnb1-/- mES cells in which cell adhesion is preserved by an E-cadherin-α-catenin (Eα) fusion protein (Ctnnb1-/-Eα mES cells), and show that mimicking only the adhesive function of β-catenin is necessary and sufficient to maintain the mES cell state, making β-catenin/Wnt signaling obsolete in this process. Furthermore, we propose a role for E-cadherin in promoting the Lif signaling cascade, showing an association of E-cadherin with the Lifr-Gp130 receptor complex, which is most likely facilitated by the extracellular domain of E-cadherin. Without Eα, and thus without maintained cell adhesion, Ctnnb1-/- mES cells downregulate components of the Lif signaling pathway, such as Lifr, Gp130 and activated Stat3, as well as pluripotency-associated markers. From these observations, we hypothesize that the changes in gene expression accompanying the loss of pluripotency are a direct consequence of dysfunctional cell adhesion. Supporting this view, we find that the requirement for intact adhesion can be circumvented by the forced expression of constitutively active Stat3. In summary, we put forward a model in which mES cells can be propagated in culture in the absence of Ctnnb1, as long as E-cadherin-mediated cell adhesion is preserved.


Cancer Cell | 2016

Genome-Wide Profiles of Extra-cranial Malignant Rhabdoid Tumors Reveal Heterogeneity and Dysregulated Developmental Pathways

Hye Jung E Chun; Emilia L. Lim; Alireza Heravi-Moussavi; Saeed Saberi; Karen Mungall; Mikhail Bilenky; Annaick Carles; Kane Tse; Inna Shlafman; Kelsey Zhu; Jenny Q. Qian; Diana L. Palmquist; An He; William Long; Rodrigo Goya; Michelle Ng; Veronique G. LeBlanc; Erin Pleasance; Nina Thiessen; Tina Wong; Eric Chuah; Yong Jun Zhao; Jacquie Schein; Daniela S. Gerhard; Michael D. Taylor; Andrew J. Mungall; Richard A. Moore; Yussanne Ma; Steven J.M. Jones; Elizabeth J. Perlman

Malignant rhabdoid tumors (MRTs) are rare lethal tumors of childhood that most commonly occur in the kidney and brain. MRTs are driven by SMARCB1 loss, but the molecular consequences of SMARCB1 loss in extra-cranial tumors have not been comprehensively described and genomic resources for analyses of extra-cranial MRT are limited. To provide such data, we used whole-genome sequencing, whole-genome bisulfite sequencing, whole transcriptome (RNA-seq) and microRNA sequencing (miRNA-seq), and histone modification profiling to characterize extra-cranial MRTs. Our analyses revealed gene expression and methylation subgroups and focused on dysregulated pathways, including those involved in neural crest development.


Nature | 2017

Fate mapping of human glioblastoma reveals an invariant stem cell hierarchy

Xiaoyang Lan; David J. Jörg; Florence M.G. Cavalli; Laura M. Richards; Long V. Nguyen; Robert Vanner; Paul Guilhamon; Lilian Lee; Michelle Kushida; Davide Pellacani; Nicole I. Park; Fiona J. Coutinho; Heather Whetstone; Hayden Selvadurai; Clare Che; Betty Luu; Annaick Carles; Michelle Moksa; Naghmeh Rastegar; Renee Head; Sonam Dolma; Panagiotis Prinos; Michael D. Cusimano; Sunit Das; Mark Bernstein; C.H. Arrowsmith; Andrew J. Mungall; Richard A. Moore; Yussanne Ma; Marco Gallo

Human glioblastomas harbour a subpopulation of glioblastoma stem cells that drive tumorigenesis. However, the origin of intratumoural functional heterogeneity between glioblastoma cells remains poorly understood. Here we study the clonal evolution of barcoded glioblastoma cells in an unbiased way following serial xenotransplantation to define their individual fate behaviours. Independent of an evolving mutational signature, we show that the growth of glioblastoma clones in vivo is consistent with a remarkably neutral process involving a conserved proliferative hierarchy rooted in glioblastoma stem cells. In this model, slow-cycling stem-like cells give rise to a more rapidly cycling progenitor population with extensive self-maintenance capacity, which in turn generates non-proliferative cells. We also identify rare ‘outlier’ clones that deviate from these dynamics, and further show that chemotherapy facilitates the expansion of pre-existing drug-resistant glioblastoma stem cells. Finally, we show that functionally distinct glioblastoma stem cells can be separately targeted using epigenetic compounds, suggesting new avenues for glioblastoma-targeted therapy.


Nature Communications | 2014

DNA barcoding reveals diverse growth kinetics of human breast tumour subclones in serially passaged xenografts

Long V. Nguyen; Claire L. Cox; Peter Eirew; David J.H.F. Knapp; Davide Pellacani; Nagarajan Kannan; Annaick Carles; Michelle Moksa; Sneha Balani; Sohrab P. Shah; Martin Hirst; Samuel Aparicio; Connie J. Eaves

Genomic and phenotypic analyses indicate extensive intra- as well as intertumoral heterogeneity in primary human malignant cell populations despite their clonal origin. Cellular DNA barcoding offers a powerful and unbiased alternative to track the number and size of multiple subclones within a single human tumour xenograft and their response to continued in vivo passaging. Using this approach we find clone-initiating cell frequencies that vary from ~1/10 to ~1/10,000 cells transplanted for two human breast cancer cell lines and breast cancer xenografts derived from three different patients. For the cell lines, these frequencies are negatively affected in transplants of more than 20,000 cells. Serial transplants reveal five clonal growth patterns (unchanging, expanding, diminishing, fluctuating or of delayed onset), whose predominance is highly variable both between and within original samples. This study thus demonstrates the high growth potential and diverse growth properties of xenografted human breast cancer cells.


Nature Communications | 2015

Epigenetic and transcriptional determinants of the human breast

Philippe Gascard; Misha Bilenky; Mahvash Sigaroudinia; Jianxin Zhao; Luolan Li; Annaick Carles; Allen Delaney; Angela Tam; Baljit Kamoh; Stephanie Cho; Malachi Griffith; Andy Chu; Gordon Robertson; Dorothy Cheung; Irene Li; Alireza Heravi-Moussavi; Michelle Moksa; Matthew Mingay; Angela Hussainkhel; Brad H. Davis; Raman P. Nagarajan; Chibo Hong; Lorigail Echipare; Henriette O'Geen; Matthew J. Hangauer; Jeffrey B. Cheng; Dana S. Neel; Donglei Hu; Michael T. McManus; Richard A. Moore

While significant effort has been dedicated to the characterization of epigenetic changes associated with prenatal differentiation, relatively little is known about the epigenetic changes that accompany post-natal differentiation where fully functional differentiated cell types with limited lifespans arise. Here we sought to address this gap by generating epigenomic and transcriptional profiles from primary human breast cell types isolated from disease-free human subjects. From these data we define a comprehensive human breast transcriptional network, including a set of myoepithelial- and luminal epithelial-specific intronic retention events. Intersection of epigenetic states with RNA expression from distinct breast epithelium lineages demonstrates that mCpG provides a stable record of exonic and intronic usage, whereas H3K36me3 is dynamic. We find a striking asymmetry in epigenomic reprogramming between luminal and myoepithelial cell types, with the genomes of luminal cells harbouring more than twice the number of hypomethylated enhancer elements compared with myoepithelial cells.


Cell Reports | 2016

Analysis of Normal Human Mammary Epigenomes Reveals Cell-Specific Active Enhancer States and Associated Transcription Factor Networks

Davide Pellacani; Misha Bilenky; Nagarajan Kannan; Alireza Heravi-Moussavi; David J.H.F. Knapp; Sitanshu Gakkhar; Michelle Moksa; Annaick Carles; Richard A. Moore; Andrew J. Mungall; Marco A. Marra; Steven J.M. Jones; Samuel Aparicio; Martin Hirst; Connie J. Eaves

The normal adult human mammary gland is a continuous bilayered epithelial system. Bipotent and myoepithelial progenitors are prominent and unique components of the outer (basal) layer. The inner (luminal) layer includes both luminal-restricted progenitors and a phenotypically separable fraction that lacks progenitor activity. We now report an epigenomic comparison of these three subsets with one another, with their associated stromal cells, and with three immortalized, non-tumorigenic human mammary cell lines. Each genome-wide analysis contains profiles for six histone marks, methylated DNA, and RNA transcripts. Analysis of these datasets shows that each cell type has unique features, primarily within genomic regulatory regions, and that the cell lines group together. Analyses of the promoter and enhancer profiles place the luminal progenitors in between the basal cells and the non-progenitor luminal subset. Integrative analysis reveals networks of subset-specific transcription factors.


bioRxiv | 2018

MeCP2-E1 isoform is a dynamically expressed, weakly DNA-bound protein with different protein and DNA interactions compared to MeCP2-E2

Alexia Martinez de Paz; Leila Khajavi; Hélène Martin; Rafael Claveria-Gimeno; Susanne tom Dieck; Manjinder S. Cheema; Jose V. Sanchez-Mut; Malgorzata Moksa; Annaick Carles; Nicholas I. Brodie; Taimoor I. Sheikh; Melissa Freeman; Evgeniy V. Petrotchenko; Christoph H. Borchers; Erin M. Schuman; Matthias Zytnicki; Adrián Velázquez-Campoy; Olga Abian; Martin Hirst; Manel Esteller; John B. Vincent; Cécile E. Malnou; Juan Ausió

MeCP2 – a chromatin-binding protein associated with Rett syndrome – has two main isoforms, MeCP2-E1 and MeCP2-E2, with 96% amino acid identity differing in a few N-terminal amino acid residues. Previous studies have shown brain region-specific expression of these isoforms which, in addition to their different cellular localization and differential expression during brain development, suggest they may also have non-overlapping molecular mechanisms. However, differential functions of MeCP2-E1 and E2 remain largely unexplored. Here, we show that the N-terminal domains (NTD) of MeCP2-E1 and E2 modulate the ability of the methyl binding domain (MBD) to interact with DNA as well as influencing the turnover rates, binding dynamics, response to nuclear depolarization, and circadian oscillations of the two isoforms. Our proteomics data indicate that both isoforms exhibit unique interacting protein partners. Moreover, genome-wide analysis using ChIP-seq provide evidence for a shared as well as a specific regulation of different sets of genes. Our findings provide insight into the functional complexity of MeCP2 by dissecting differential aspects of its two isoforms. Significance Whether the two E1 and E2 isoforms of MeCP2 have different structural and/or functional implications has been highly controversial and is not well known. Here we show that the relatively short N-terminal sequence variation between the two isoforms impinges them with an important DNA binding difference. Moreover, MeCP2-E1 and E2 exhibit a different cellular dynamic behavior and have some distinctive interacting partners. In addition, while sharing genome occupancy they specifically bind to several distinctive genes.


Experimental Hematology | 2017

Whole-genome analysis reveals unexpected dynamics of mutant subclone development in a patient with JAK2-V617F-positive chronic myeloid leukemia

Ivan Sloma; Maria Teresa Mitjavila-Garcia; Olivier Feraud; Frank Griscelli; Noufissa Oudrhiri; Sanaa El Marsafy; Emilie Gobbo; Dominique Divers; Alexis Proust; David Smadja; Christophe Desterke; Annaick Carles; Yusanna Ma; Martin Hirst; Marco A. Marra; Connie J. Eaves; Annelise Bennaceur-Griscelli; Ali G. Turhan

We report here the first use of whole-genome sequencing (WGS) to examine the initial clonal dynamics in an unusual patient with chronic myeloid leukemia (CML), who presented in chronic phase (CP) with doubly marked BCR-ABL1+/JAK2V617F-mutant cells and, over a 9-year period, progressed into an accelerated phase (AP) and then terminal blast phase (BP). WGS revealed that the diagnostic cells also contained mutations in ASXL1, SEC23B, MAD1L1, and RREB1 as well as 12,000 additional uncommon DNA variants. WGS of endothelial cells generated from circulating precursors revealed many of these were shared with the CML clone. Surprisingly, WGS of induced pluripotent stem cells (iPSCs) derived from the AP cells revealed only six additional coding somatic mutations, despite retention by the hematopoietic progeny of the parental AP cell levels of BCR-ABL1 expression and sensitivity to imatinib and pimozide. Limited analysis of BP cells revealed independent subclonal progression to homozygosity of the MAD1L1 and RREB1 variants. MAD1L1 and SEC23B mutations were also identified in 2 of 101 cases of myeloproliferative neoplasms, but not in 42 healthy subjects. These findings challenge historic concepts of clonal evolution in CML.

Collaboration


Dive into the Annaick Carles's collaboration.

Top Co-Authors

Avatar

Martin Hirst

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Connie J. Eaves

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Michelle Moksa

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Alireza Heravi-Moussavi

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David J.H.F. Knapp

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marco A. Marra

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge