Network


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

Hotspot


Dive into the research topics where Alicia N. Schep is active.

Publication


Featured researches published by Alicia N. Schep.


PLOS ONE | 2013

A Comparative Analysis of Transcription Factor Expression during Metazoan Embryonic Development

Alicia N. Schep; Boris Adryan

During embryonic development, a complex organism is formed from a single starting cell. These processes of growth and differentiation are driven by large transcriptional changes, which are following the expression and activity of transcription factors (TFs). This study sought to compare TF expression during embryonic development in a diverse group of metazoan animals: representatives of vertebrates (Danio rerio, Xenopus tropicalis), a chordate (Ciona intestinalis) and invertebrate phyla such as insects (Drosophila melanogaster, Anopheles gambiae) and nematodes (Caenorhabditis elegans) were sampled, The different species showed overall very similar TF expression patterns, with TF expression increasing during the initial stages of development. C2H2 zinc finger TFs were over-represented and Homeobox TFs were under-represented in the early stages in all species. We further clustered TFs for each species based on their quantitative temporal expression profiles. This showed very similar TF expression trends in development in vertebrate and insect species. However, analysis of the expression of orthologous pairs between more closely related species showed that expression of most individual TFs is not conserved, following the general model of duplication and diversification. The degree of similarity between TF expression between Xenopus tropicalis and Danio rerio followed the hourglass model, with the greatest similarity occuring during the early tailbud stage in Xenopus tropicalis and the late segmentation stage in Danio rerio. However, for Drosophila melanogaster and Anopheles gambiae there were two periods of high TF transcriptome similarity, one during the Arthropod phylotypic stage at 8–10 hours into Drosophila development and the other later at 16–18 hours into Drosophila development.


Nature Methods | 2017

chromVAR: inferring transcription-factor-associated accessibility from single-cell epigenomic data

Alicia N. Schep; Beijing Wu; Jason D. Buenrostro; William J. Greenleaf

Single-cell ATAC-seq (scATAC) yields sparse data that make conventional analysis challenging. We developed chromVAR (http://www.github.com/GreenleafLab/chromVAR), an R package for analyzing sparse chromatin-accessibility data by estimating gain or loss of accessibility within peaks sharing the same motif or annotation while controlling for technical biases. chromVAR enables accurate clustering of scATAC-seq profiles and characterization of known and de novo sequence motifs associated with variation in chromatin accessibility.


Cell | 2018

Integrated Single-Cell Analysis Maps the Continuous Regulatory Landscape of Human Hematopoietic Differentiation

Jason D. Buenrostro; M. Ryan Corces; Caleb Lareau; Beijing Wu; Alicia N. Schep; Martin J. Aryee; Ravindra Majeti; Howard Y. Chang; William J. Greenleaf

Human hematopoiesis involves cellular differentiation of multipotent cells into progressively more lineage-restricted states. While the chromatin accessibility landscape of this process has been explored in defined populations, single-cell regulatory variation has been hidden by ensemble averaging. We collected single-cell chromatin accessibility profiles across 10 populations of immunophenotypically defined human hematopoietic cell types and constructed a chromatin accessibility landscape of human hematopoiesis to characterize differentiation trajectories. We find variation consistent with lineage bias toward different developmental branches in multipotent cell types. We observe heterogeneity within common myeloid progenitors (CMPs) and granulocyte-macrophage progenitors (GMPs) and develop a strategy to partition GMPs along their differentiation trajectory. Furthermore, we integrated single-cell RNA sequencing (scRNA-seq) data to associate transcription factors to chromatin accessibility changes and regulatory elements to target genes through correlations of expression and regulatory element accessibility. Overall, this work provides a framework for integrative exploration of complex regulatory dynamics in a primary human tissue at single-cell resolution.


bioRxiv | 2017

chromVAR: Inferring transcription factor variation from single-cell epigenomic data

Alicia N. Schep; Beijing Wu; Jason D. Buenrostro; William J. Greenleaf

Single cell ATAC-seq (scATAC) yields sparse data that makes application of conventional computational approaches for data analysis challenging or impossible. We developed chromVAR, an R package for analyzing sparse chromatin accessibility data by estimating the gain or loss of accessibility within sets of peaks sharing the same motif or annotation while controlling for known technical biases. chromVAR enables accurate clustering of scATAC-seq profiles and enables characterization of known, or the de novo identification of novel, sequence motifs associated with variation in chromatin accessibility across single cells or other sparse epigenomic data sets.


DNA and Cell Biology | 2010

Characterization of an Ultra-Conserved Putative cis-Regulatory Module at the Mammalian Telomerase Reverse Transcriptase Gene

Diana A. Tran; Terence C. Wong; Alicia N. Schep; Robert A. Drewell

Telomeres are regions of repeated DNA sequence that cap the ends of eukaryotic chromosomes. They act as disposable safeguards to prevent the loss of important genetic information during DNA replication due to the inability of DNA polymerase to replicate DNA to the ends of linear chromosomes. The synthesis of new telomeric repeats using an RNA molecule as a template is catalyzed by the enzyme telomerase. In embryonic stem cells, the gene encoding the catalytic protein subunit of the telomerase complex (telomere reverse transcriptase [TERT]) is transcriptionally active and critical for telomere elongation, allowing for continued cellular differentiation during development. The TERT gene is down-regulated as embryogenesis progresses to limit the proliferative capacity of cells. As a result, in normal human adult somatic cells the TERT gene is silenced. However, in over 90% of cancers, the TERT gene is reactivated, allowing cells to bypass senescence and become immortalized. In this study, we explore the molecular mechanisms that regulate transcriptional expression of the TERT gene. Bioinformatic analysis of the noncoding genomic regions around the human TERT gene identified a TERT ultra-conserved (TUC) module located 5 kb upstream of the transcription start site. This 308 bp region is over 75% conserved between distantly related mammalian species and over 91% conserved among primate species. The cis-regulatory potential of the TUC region was tested in cell-based reporter gene assays. Transient transfections into HeLa and lung fibroblast cells demonstrated that the TUC module has transcriptional enhancer activity. Further bioinformatic analysis revealed that the TUC region is highly enriched in putative transcription factor binding sites for proteins involved during hematopoiesis, indicating that the TUC module may be an enhancer for the TERT gene in specific cell lineages.


Methods in Cell Biology | 2009

Identification and Characterization of Dynein Genes in Tetrahymena

David E. Wilkes; Nicole Bennardo; Clarence W.C. Chan; Yu-Loung Chang; Elizabeth O. Corpuz; Jennifer DuMond; Jordan A. Eboreime; Julianna Erickson; Jonathan Hetzel; Erin E. Heyer; Mark J. Hubenschmidt; Ekaterina Kniazeva; Hallie Kuhn; Michelle Lum; Andrea Sand; Alicia N. Schep; Oksana Sergeeva; Natt Supab; Caroline R. Townsend; Liesl Van Ryswyk; Hadley E. Watson; Alice E. Wiedeman; Vidyalakshmi Rajagopalan; David J. Asai

We describe the protocol through which we identify and characterize dynein subunit genes in the ciliated protozoan Tetrahymena thermophila. The gene(s) of interest is found by searching the Tetrahymena genome, and it is characterized in silico including the prediction of the open reading frame and identification of likely introns. The gene is then characterized experimentally, including the confirmation of the exon-intron organization of the gene and the measurement of the expression of the gene in nondeciliated and reciliating cells. In order to understand the function of the gene product, the gene is modified-for example, deleted, overexpressed, or epitope-tagged-using the straightforward gene replacement strategies available with Tetrahymena. The effect(s) of the dynein gene modification is evaluated by examining transformants for ciliary traits including cell motility, ciliogenesis, cell division, and the engulfment of particles through the oral apparatus. The multistepped protocol enables undergraduate students to engage in short- and long-term experiments. In our laboratory during the last 6 years, more than two dozen undergraduate students have used these methods to investigate dynein subunit genes.


Nature Communications | 2018

Unsupervised clustering and epigenetic classification of single cells

Mahdi Zamanighomi; Zhixiang Lin; Timothy Daley; Xi Chen; Zhana Duren; Alicia N. Schep; William J. Greenleaf; Wing Hung Wong

Characterizing epigenetic heterogeneity at the cellular level is a critical problem in the modern genomics era. Assays such as single cell ATAC-seq (scATAC-seq) offer an opportunity to interrogate cellular level epigenetic heterogeneity through patterns of variability in open chromatin. However, these assays exhibit technical variability that complicates clear classification and cell type identification in heterogeneous populations. We present scABC, an R package for the unsupervised clustering of single-cell epigenetic data, to classify scATAC-seq data and discover regions of open chromatin specific to cell identity.Single cell ATAC-seq (scATAC-seq) data reveals cellular level epigenetic heterogeneity but its application in delineating distinct subpopulations is still challenging. Here, the authors develop scABC, a statistical method for unsupervised clustering of scATAC-seq data and identification of open chromatin regions specific to cell identity.


Cell Reports | 2018

INO80 Chromatin Remodeling Coordinates Metabolic Homeostasis with Cell Division

Graeme J. Gowans; Alicia N. Schep; Ka Man Wong; Devin A. King; William J. Greenleaf; Ashby J. Morrison

SUMMARY Adaptive survival requires the coordination of nutrient availability with expenditure of cellular resources. For example, in nutrient-limited environments, 50% of all S. cerevisiae genes synchronize and exhibit periodic bursts of expression in coordination with respiration and cell division in the yeast metabolic cycle (YMC). Despite the importance of metabolic and proliferative synchrony, the majority of YMC regulators are currently unknown. Here, we demonstrate that the INO80 chromatin-remodeling complex is required to coordinate respiration and cell division with periodic gene expression. Specifically, INO80 mutants have severe defects in oxygen consumption and promiscuous cell division that is no longer coupled with metabolic status. In mutant cells, chromatin accessibility of periodic genes, including TORC1-responsive genes, is relatively static, concomitant with severely attenuated gene expression. Collectively, these results reveal that the INO80 complex mediates metabolic signaling to chromatin to restrict proliferation to metabolically optimal states.


Gene | 2011

Transcriptional repression by the proximal exonic region at the human TERT gene.

Terence C. Wong; Ethan S. Sokol; Alicia N. Schep; Meera Punjiya; Diana A. Tran; Divina Allan; Robert A. Drewell

In humans, the enzyme telomerase (hTERT) is responsible for the synthesis of new repeat sequences at the telomeres of chromosomes. Although active in early embryogenesis, the hTERT gene is transcriptionally silenced in almost all somatic cells in the adult, but is aberrantly re-activated in over 90% of human cancers. The molecular mechanisms responsible for repression of this gene are thought to involve the transcription factor CTCF. In this study, we bioinformatically identify putative CTCF binding sites in the hTERT proximal exonic region (PER) and determine their functional relevance in mediating transcriptional silencing at this gene. Tests using a reporter gene assay in HeLa cancer cells demonstrate that a sub-region of the PER exhibits strong transcriptional repressive activity. This repression is independent of the previously identified CTCF binding site near the transcriptional start site of the hTERT gene. In addition, site directed mutagenesis of three predicted CTCF binding sites, including a previously characterized in vivo site in exon 2, does not result in a loss of the repression mediated by the PER. The results from this study indicate that expression of the hTERT gene in HeLa cells is regulated by sequences in the PER. This transcriptional control is mediated through additional regulatory molecular mechanisms, independent of CTCF binding.


bioRxiv | 2018

Joint single-cell DNA accessibility and protein epitope profiling reveals environmental regulation of epigenomic heterogeneity

Xingqi Chen; Ulrike Litzenburger; Yuning Wei; Alicia N. Schep; Edward L. LaGory; Hani Choudhry; Amato J. Giaccia; William J. Greenleaf; Howard Y. Chang

Here we introduce Protein-indexed Assay of Transposase Accessible Chromatin with sequencing (Pi-ATAC) that combines single-cell chromatin and proteomic profiling. In conjunction with DNA transposition, the levels of multiple cell surface or intracellular protein epitopes are recorded by index flow cytometry and positions in arrayed microwells, and then subject to molecular barcoding for subsequent pooled analysis. Pi-ATAC simultaneously identifies the epigenomic and proteomic heterogeneity in individual cells. Pi-ATAC reveals a casual link between transcription factor abundance and DNA motif access, and deconvolute cell types and states in the tumor microenvironment in vivo. We identify a dominant role for hypoxia, marked by HIF1A protein, in the tumor microvenvironment for shaping the regulome in a subset of epithelial tumor cells.

Collaboration


Dive into the Alicia N. Schep's collaboration.

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge