Network


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

Hotspot


Dive into the research topics where Danielle Dionne is active.

Publication


Featured researches published by Danielle Dionne.


Nucleic Acids Research | 2013

Discovery and characterization of artifactual mutations in deep coverage targeted capture sequencing data due to oxidative DNA damage during sample preparation

Maura Costello; Trevor J. Pugh; Timothy Fennell; Chip Stewart; Lee Lichtenstein; James C. Meldrim; Jennifer L. Fostel; Dennis Friedrich; Danielle Perrin; Danielle Dionne; Sharon Kim; Stacey Gabriel; Eric S. Lander; Sheila Fisher; Gad Getz

As researchers begin probing deep coverage sequencing data for increasingly rare mutations and subclonal events, the fidelity of next generation sequencing (NGS) laboratory methods will become increasingly critical. Although error rates for sequencing and polymerase chain reaction (PCR) are well documented, the effects that DNA extraction and other library preparation steps could have on downstream sequence integrity have not been thoroughly evaluated. Here, we describe the discovery of novel C > A/G > T transversion artifacts found at low allelic fractions in targeted capture data. Characteristics such as sequencer read orientation and presence in both tumor and normal samples strongly indicated a non-biological mechanism. We identified the source as oxidation of DNA during acoustic shearing in samples containing reactive contaminants from the extraction process. We show generation of 8-oxoguanine (8-oxoG) lesions during DNA shearing, present analysis tools to detect oxidation in sequencing data and suggest methods to reduce DNA oxidation through the introduction of antioxidants. Further, informatics methods are presented to confidently filter these artifacts from sequencing data sets. Though only seen in a low percentage of reads in affected samples, such artifacts could have profoundly deleterious effects on the ability to confidently call rare mutations, and eliminating other possible sources of artifacts should become a priority for the research community.


Science | 2017

Decoupling genetics, lineages, and microenvironment in IDH-mutant gliomas by single-cell RNA-seq.

Andrew S. Venteicher; Itay Tirosh; Christine Hebert; Keren Yizhak; Cyril Neftel; Mariella G. Filbin; Volker Hovestadt; Leah E. Escalante; McKenzie L. Shaw; Christopher Rodman; Shawn M. Gillespie; Danielle Dionne; Christina C. Luo; Hiranmayi Ravichandran; Ravindra Mylvaganam; Christopher Mount; Maristela L. Onozato; Brian V. Nahed; Hiroaki Wakimoto; William T. Curry; A. John Iafrate; Miguel Rivera; Matthew P. Frosch; Todd R. Golub; Priscilla K. Brastianos; Gad Getz; Anoop P. Patel; Michelle Monje; Daniel P. Cahill; Orit Rozenblatt-Rosen

Single-cell RNA sequencing identifies a common origin for specific types of human glioma brain tumors. Effects of the tumor microenvironment Glioma brain tumors that carry mutant copies of the IDH gene can be subdivided into two major classes. However, the development of and differences between these two classes are not well characterized. Venteicher et al. coupled bulk sequencing and publicly available data with single-cell RNA sequencing data on glioma patient tissue samples. They identified a common lineage program that is shared between glioma subtypes. This suggests that the observed differences between the two glioma classes originate from lineage-specific genetic changes and the tumor microenvironment. Science, this issue p. eaai8478 INTRODUCTION Tumor fitness, evolution, and resistance to therapy are governed by selection of malignant cells with specific genotypes, by expression programs related to cellular phenotypes, and by influences of the tumor microenvironment (TME). Although bulk tumor analysis can interrogate the genetic state of tumor cells with high precision, bulk expression profiles average the diverse cells within each tumor, thereby masking critical differences and providing limited insight into cancer cell programs and TME influences. Single-cell RNA sequencing (scRNA-seq) can help to address those challenges but incurs financial and logistic considerations, including the time required to accrue large cohorts of fresh tumor specimen for single-cell analysis. RATIONALE We reasoned that scRNA-seq of a limited number of representative tumors could be combined with bulk data from large cohorts to decipher differences between tumor subclasses. In this approach, bulk samples collected for large cohorts, such as from The Cancer Genome Atlas (TCGA), are first used to define the combined effects of differences in cancer cell genotypes, phenotypes, and the composition of the TME. Single-cell analysis of a limited set of representative tumors is then used to distinguish those effects. We applied this approach to understand the differences between two types of isocitrate dehydrogenase (IDH)–mutant gliomas: astrocytoma (IDH-A) and oligodendroglioma (IDH-O). IDH-A and IDH-O are distinguished by co-occurring signature genetic events and by histopathology and are thought to recapitulate distinct glial lineages. By combining 9879 scRNA-seq profiles from 10 IDH-A tumors, 4347 scRNA-seq profiles from six IDH-O tumors, and 165 TCGA bulk RNA profiles, we could decipher differences between these two tumor types at single-cell resolution. RESULTS We find that differences in bulk expression profiles between IDH-A and IDH-O are primarily explained by the impact of signature genetic events and TME composition, but not by distinct expression programs of glial lineages in the malignant cells. We infer that both IDH-A and IDH-O share the same developmental hierarchy, consisting in each case of three subpopulations of malignant cells: nonproliferating cells differentiated along the astrocytic and oligodendrocytic lineages, and proliferative undifferentiated cells that resemble neural stem/progenitor cells. By analyzing tumors of different clinical grades, we observe that higher-grade tumors present enhanced proliferation, larger pools of undifferentiated glioma cells, and an increase in macrophage over microglia programs in the TME. CONCLUSION Our approach provides a general framework to decipher differences between classes of human tumors by decoupling cancer cell genotypes, phenotypes, and the composition of the TME. The shared glial lineages and developmental hierarchies observed in IDH-A and IDH-O suggest a common progenitor for all IDH-mutant gliomas, shedding light on a long-standing debate in gliomagenesis. In contrast to the similarity in glial lineages, IDH-A and IDH-O differ significantly in their TME, and in particular in the abundance of microglia/macrophage cells. Microglia and macrophages also differ between IDH-A tumors of different grades. Our study redefines the cellular composition of human IDH-mutant gliomas, with important implications for disease management. Single-cell RNA-seq of IDH-mutant gliomas reveals tumor architecture. (Top) Human samples were dissociated and analyzed by scRNA-seq. (Bottom) IDH-O and IDH-A differ in genetics and TME but are both primarily composed of three main types of malignant cells: cycling stem-like cells and noncycling astrocyte-like and oligodendrocyte-like cells. Tumor progression is associated with increased proliferation, decreased differentiation, and increase in macrophages over microglia in the TME. Tumor subclasses differ according to the genotypes and phenotypes of malignant cells as well as the composition of the tumor microenvironment (TME). We dissected these influences in isocitrate dehydrogenase (IDH)–mutant gliomas by combining 14,226 single-cell RNA sequencing (RNA-seq) profiles from 16 patient samples with bulk RNA-seq profiles from 165 patient samples. Differences in bulk profiles between IDH-mutant astrocytoma and oligodendroglioma can be primarily explained by distinct TME and signature genetic events, whereas both tumor types share similar developmental hierarchies and lineages of glial differentiation. As tumor grade increases, we find enhanced proliferation of malignant cells, larger pools of undifferentiated glioma cells, and an increase in macrophage over microglia expression programs in TME. Our work provides a unifying model for IDH-mutant gliomas and a general framework for dissecting the differences among human tumor subclasses.


Nature | 2017

A single-cell survey of the small intestinal epithelium

Adam L. Haber; Moshe Biton; Noga Rogel; Rebecca H. Herbst; Karthik Shekhar; Christopher Smillie; Grace Burgin; Toni Delorey; Michael R. Howitt; Yarden Katz; Itay Tirosh; Semir Beyaz; Danielle Dionne; Mei Zhang; Raktima Raychowdhury; Wendy S. Garrett; Orit Rozenblatt-Rosen; Hai Ning Shi; Ömer H. Yilmaz; Ramnik J. Xavier; Aviv Regev

Intestinal epithelial cells absorb nutrients, respond to microbes, function as a barrier and help to coordinate immune responses. Here we report profiling of 53,193 individual epithelial cells from the small intestine and organoids of mice, which enabled the identification and characterization of previously unknown subtypes of intestinal epithelial cell and their gene signatures. We found unexpected diversity in hormone-secreting enteroendocrine cells and constructed the taxonomy of newly identified subtypes, and distinguished between two subtypes of tuft cell, one of which expresses the epithelial cytokine Tslp and the pan-immune marker CD45, which was not previously associated with non-haematopoietic cells. We also characterized the ways in which cell-intrinsic states and the proportions of different cell types respond to bacterial and helminth infections: Salmonella infection caused an increase in the abundance of Paneth cells and enterocytes, and broad activation of an antimicrobial program; Heligmosomoides polygyrus caused an increase in the abundance of goblet and tuft cells. Our survey highlights previously unidentified markers and programs, associates sensory molecules with cell types, and uncovers principles of gut homeostasis and response to pathogens.


Nature Biomedical Engineering | 2018

Effects of 3D culturing conditions on the transcriptomic profile of stem-cell-derived neurons

Halil Tekin; Sean Simmons; Beryl B. Cummings; Linyi Gao; Xian Adiconis; Cynthia Hession; Ayan Ghoshal; Danielle Dionne; Sourav R Choudhury; Volkan Yesilyurt; Neville E. Sanjana; Xi Shi; Congyi Lu; Matthias Heidenreich; Jen Q. Pan; Joshua Z. Levin; Feng Zhang

Understanding neurological diseases requires tractable genetic systems, and engineered three-dimensional (3D) neural tissues are an attractive choice. Yet how the cellular transcriptomic profiles in these tissues are affected by the encapsulating materials and are related to the human brain transcriptome is not well understood. Here, we report the characterization of the effects of different culturing conditions on the transcriptomic profiles of induced neuronal cells and developed a method for the rapid generation of 3D co-cultures of neuronal and astrocytic cells from the same pool of human embryonic stem cells. By comparing the gene-expression profiles of neuronal cells in culture conditions relevant to the developing human brain, we found that modifying the degree of crosslinking of composite hydrogels can tune expression patterns so that they correlate with those of specific brain regions and developmental stages. Moreover, single-cell-sequencing results showed that our engineered tissues recapitulate transcriptional patterns of cell types in the human brain. Analyses of culturing conditions will inform the development of 3D neural tissues for use as tractable models of brain diseases.Culturing conditions affect the transcriptomic profiles of induced neuronal cells, and 3D co-cultures of induced neuronal cells and astrocytic cells can be rapidly generated from the same pool of human embryonic stem cells.


bioRxiv | 2017

T helper cells modulate intestinal stem cell renewal and differentiation

Moshe Biton; Adam L. Haber; Semir Beyaz; Noga Rogel; Christopher Smillie; Karthik Shekhar; Alexandra Schnell; Zuojia Chen; Chuan Wu; Jose Ordovas-Montanes; David Alvarez; Rebecca H. Herbst; Itay Tirosh; Grace Burgin; Danielle Dionne; Michael E. Xifaras; Mei Zhang; Alex K. Shalek; Ulrich H. von Andrian; Daniel B. Graham; Orit Rozenblatt-Rosen; Hai Ning Shi; Vijay K. Kuchroo; Omer Yilmaz; Aviv Regev; Ramnik J. Xavier

In the small intestine, a cellular niche of diverse accessory cell types supports the rapid generation of mature epithelial cell types through self-renewal, proliferation, and differentiation of intestinal stem cells (ISCs). However, not much is known about interactions between immune cells and ISCs, and it is unclear if and how immune cell dynamics affect eventual ISC fate or the balance between self-renewal and differentiation. Here, we used single-cell RNA-seq (scRNA-Seq) of intestinal epithelial cells (IECs) to identify new mechanisms for ISC–immune cell interactions. Surprisingly, MHC class II (MHCII) is enriched in two distinct subsets of Lgr5+ crypt base columnar ISCs, which are also distinguished by higher proliferation rates. Using co-culture of T cells with intestinal organoids, cytokine stimulations, and in vivo mouse models, we confirm that CD4+ T helper (Th) cells communicate with ISCs and affect their differentiation, in a manner specific to the Th subtypes and their signature cytokines and dependent on MHCII expression by ISCs. Specific inducible knockout of MHCII in intestinal epithelial cells in mice in vivo results in expansion of the ISC pool. Mice lacking T cells have expanded ISC pools, whereas specific depletion of Treg cells in vivo results in substantial reduction of ISC numbers. Our findings show that interactions between Th cells and ISCs mediated via MHCII expressed in intestinal epithelial stem cells help orchestrate tissue-wide responses to external signals.


bioRxiv | 2018

Single-cell transcriptomics of the aged mouse brain reveals convergent, divergent and unique aging signatures

Methodios Ximerakis; Scott Lipnick; Sean Simmons; Xian Adiconis; Brendan T. Innes; Danielle Dionne; Lan Nguyen; Brittany A Mayweather; Ceren Ozek; Zachary Niziolek; Vincent Butty; Ruth Isserlin; Sean M. Buchanan; Stuart R Levine; Aviv Regev; Gary D. Bader; Joshua Z. Levin; Lee L. Rubin

The mammalian brain is complex, with multiple cell types performing a variety of diverse functions, but exactly how the brain is affected with aging remains largely unknown. Here we performed a single-cell transcriptomic analysis of young and old mouse brains. We provide a comprehensive dataset of aging-related genes, pathways and ligand-receptor interactions in nearly all brain cell types. Our analysis identified gene signatures that vary in a coordinated manner across cell types and gene sets that are regulated in a cell type specific manner, even at times in opposite directions. Thus, our data reveals that aging, rather than inducing a universal program drives a distinct transcriptional course in each cell population. These data provide an important resource for the aging community and highlight key molecular processes, including ribosomal biogenesis, underlying aging. We believe that this large-scale dataset, which is publicly accessible online (aging-mouse-brain), will facilitate additional discoveries directed towards understanding and modifying the aging process.


bioRxiv | 2018

A single cell-based atlas of human microglial states reveals associations with neurological disorders and histopathological features of the aging brain

Marta Olah; Vilas Menon; Naomi Habib; Mariko Taga; Christina Yung; Maria Cimpean; Anthony Khairalla; Danielle Dionne; Sarah C. Hopp; Matthew P. Frosch; Bradley T. Hyman; Thomas G. Beach; Rani A. Sarkis; Garth Rees Cosgrove; Jeffrey Helgager; Jeffrey A. Golden; Page B. Pennell; Julie A. Schneider; David A. Bennett; Aviv Regev; Wassim Elyaman; Elizabeth M. Bradshaw; Philip L. De Jager

Recent studies of bulk microglia have provided insights into the role of this immune cell type in central nervous system development, homeostasis and dysfunction. Nonetheless, our understanding of the diversity of human microglial cell states remains limited; microglia are highly plastic and have multiple different roles, making the extent of phenotypic heterogeneity a central question, especially in light of the development of therapies targeting this cell type. Here, we investigated the population structure of human microglia by single-cell RNA-sequencing. Using surgical- and autopsy-derived cortical brain samples, we identified 14 human microglial subpopulations and noted substantial intra- and inter-individual heterogeneity. These putative subpopulations display divergent associations with Alzheimer’s disease, multiple sclerosis, and other diseases. Several states show enrichment for genes found in disease-associated mouse microglial states, suggesting additional diversity among human microglia. Overall, human microglia appear to exist in different functional states with varying levels of involvement in different brain pathologies.


bioRxiv | 2018

Rewiring of the cellular and inter-cellular landscape of the human colon during ulcerative colitis

C. S. Smillie; M. Biton; J. Ordovas-Montanes; K. M. Sullivan; Grace Burgin; Daniel B. Graham; R. H. Herbst; Noga Rogel; M. Slyper; J. Waldman; M. Sud; E. Andrews; Adam L. Haber; S. Vickovic; Danielle Dionne; L. T. Nguyen; A. C. Villani; M. Hofree; E. A. Creasey; H. Huang; Orit Rozenblatt-Rosen; J. J. Garber; H. Khalili; A. N. Desch; M. J. Daly; A. N. Ananthakrishnan; A. K. Shalek; Ramnik J. Xavier; Aviv Regev

Abstract Genome-wide association studies (GWAS) have revealed risk alleles for ulcerative colitis (UC), but their cell type and pathway specificities are often unknown. Here, we generate an atlas of 115,517 cells from the colon mucosa of seven UC patients and ten healthy individuals, revealing 51 epithelial, stromal, and immune cell subsets. These included a subset of BEST4+ enterocytes, which may sense and respond to pH, and IL13RA2+IL-11+ inflammatory fibroblasts, which we associate with resistance to anti-TNF therapy. Inflammatory fibroblasts, inflammatory monocytes, microfold-like cells, and CD8+IL-17+ T cells expand during disease, and form intercellular interaction hubs that mediate cross-talk between diverse cellular lineages. We identify hundreds of putative autocrine and paracrine cell-cell interactions that may explain the migration, expansion, or inhibition of cell types with disease. Surprisingly, UC risk genes are often cell type specific and co-regulated in relatively few gene modules, suggesting convergence onto limited sets of cell types and pathways. Using this observation, we nominate and infer putative functions for UC risk genes across all GWAS loci. Our atlas thus provides a framework for interrogating complex human diseases and mapping risk variants onto their cell types and pathways of activity.


Nature | 2017

Erratum: The neuropeptide NMU amplifies ILC2-driven allergic lung inflammation

Antonia Wallrapp; Samantha J. Riesenfeld; Patrick R. Burkett; Raja-Elie E. Abdulnour; Jackson Nyman; Danielle Dionne; Matan Hofree; Michael S. Cuoco; Christopher Rodman; Daneyal Farouq; Brian J. Haas; Timothy L. Tickle; John J. Trombetta; Pankaj Baral; Christoph S. N. Klose; Tanel Mahlakõiv; David Artis; Orit Rozenblatt-Rosen; Isaac M. Chiu; Bruce D. Levy; Monika S. Kowalczyk; Aviv Regev; Vijay K. Kuchroo

This corrects the article DOI: 10.1038/nature24029


Cell | 2016

Perturb-Seq: Dissecting Molecular Circuits with Scalable Single-Cell RNA Profiling of Pooled Genetic Screens.

Atray Dixit; Oren Parnas; Biyu Li; Jenny Chen; Charles P. Fulco; Livnat Jerby-Arnon; Nemanja D. Marjanovic; Danielle Dionne; Tyler Burks; Raktima Raychowdhury; Britt Adamson; Thomas Norman; Eric S. Lander; Jonathan S. Weissman; Nir Friedman; Aviv Regev

Collaboration


Dive into the Danielle Dionne's collaboration.

Top Co-Authors

Avatar

Aviv Regev

Massachusetts Institute of Technology

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

Vijay K. Kuchroo

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Antonia Wallrapp

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge