87 results on '"Jesse Gillis"'
Search Results
2. Defining the extent of gene function using ROC curvature
- Author
-
Stephan Fischer, Jesse Gillis, Cold Spring Harbor Laboratory (CSHL), Hub Bioinformatique et Biostatistique - Bioinformatics and Biostatistics HUB, Institut Pasteur [Paris] (IP)-Université Paris Cité (UPCité), University of Toronto, and This work was supported by the National Institutes of Health [R01MH113005, R01LM012736, and U19MH114821 to J.G.].
- Subjects
Statistics and Probability ,Computational Mathematics ,Computational Theory and Mathematics ,[SDV.BIBS]Life Sciences [q-bio]/Quantitative Methods [q-bio.QM] ,Molecular Biology ,Biochemistry ,Computer Science Applications - Abstract
MotivationInteractions between proteins help us understand how genes are functionally related and how they contribute to phenotypes. Experiments provide imperfect ‘ground truth’ information about a small subset of potential interactions in a specific biological context, which can then be extended to the whole genome across different contexts, such as conditions, tissues or species, through machine learning methods. However, evaluating the performance of these methods remains a critical challenge. Here, we propose to evaluate the generalizability of gene characterizations through the shape of performance curves.ResultsWe identify Functional Equivalence Classes (FECs), subsets of annotated and unannotated genes that jointly drive performance, by assessing the presence of straight lines in ROC curves built from gene-centric prediction tasks, such as function or interaction predictions. FECs are widespread across data types and methods, they can be used to evaluate the extent and context-specificity of functional annotations in a data-driven manner. For example, FECs suggest that B cell markers can be decomposed into shared primary markers (10–50 genes), and tissue-specific secondary markers (100–500 genes). In addition, FECs suggest the existence of functional modules that span a wide range of the genome, with marker sets spanning at most 5% of the genome and data-driven extensions of Gene Ontology sets spanning up to 40% of the genome. Simple to assess visually and statistically, the identification of FECs in performance curves paves the way for novel functional characterization and increased robustness in the definition of functional gene sets.Availability and implementationCode for analyses and figures is available at https://github.com/yexilein/pyroc.Supplementary informationSupplementary data are available at Bioinformatics online.
- Published
- 2022
3. Supplementary Figure S1 from Intraductal Transplantation Models of Human Pancreatic Ductal Adenocarcinoma Reveal Progressive Transition of Molecular Subtypes
- Author
-
David A. Tuveson, Youngkyu Park, Steven Gallinger, Faiyaz Notta, Michael Wigler, Christopher R. Vakoc, Alexander Krasnitz, Jesse Gillis, Ralph H. Hruban, Laura D. Wood, Nicholas J. Roberts, Richard A. Burkhart, Chang-Il Hwang, Hervé Tiriac, Tim D.D. Somerville, Risa Karakida Kawaguchi, Gun Ho Jang, Jude Kendall, Siran Li, Pascal Belleau, Brinda Alagesan, Dennis Plenker, Giuseppina Caligiuri, Benno Traub, Astrid Deschênes, Lindsey A. Baker, and Koji Miyabayashi
- Abstract
Comparison of IGO- and OGO-derived lesions
- Published
- 2023
4. Supplementary Figure S2 from Intraductal Transplantation Models of Human Pancreatic Ductal Adenocarcinoma Reveal Progressive Transition of Molecular Subtypes
- Author
-
David A. Tuveson, Youngkyu Park, Steven Gallinger, Faiyaz Notta, Michael Wigler, Christopher R. Vakoc, Alexander Krasnitz, Jesse Gillis, Ralph H. Hruban, Laura D. Wood, Nicholas J. Roberts, Richard A. Burkhart, Chang-Il Hwang, Hervé Tiriac, Tim D.D. Somerville, Risa Karakida Kawaguchi, Gun Ho Jang, Jude Kendall, Siran Li, Pascal Belleau, Brinda Alagesan, Dennis Plenker, Giuseppina Caligiuri, Benno Traub, Astrid Deschênes, Lindsey A. Baker, and Koji Miyabayashi
- Abstract
Compared to OGO-derived tumors, IGO-derived tumors have less active TGF-β signaling and better resemble the Progenitor subtype of PDAC.
- Published
- 2023
5. Data from Intraductal Transplantation Models of Human Pancreatic Ductal Adenocarcinoma Reveal Progressive Transition of Molecular Subtypes
- Author
-
David A. Tuveson, Youngkyu Park, Steven Gallinger, Faiyaz Notta, Michael Wigler, Christopher R. Vakoc, Alexander Krasnitz, Jesse Gillis, Ralph H. Hruban, Laura D. Wood, Nicholas J. Roberts, Richard A. Burkhart, Chang-Il Hwang, Hervé Tiriac, Tim D.D. Somerville, Risa Karakida Kawaguchi, Gun Ho Jang, Jude Kendall, Siran Li, Pascal Belleau, Brinda Alagesan, Dennis Plenker, Giuseppina Caligiuri, Benno Traub, Astrid Deschênes, Lindsey A. Baker, and Koji Miyabayashi
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most lethal common malignancy, with little improvement in patient outcomes over the past decades. Recently, subtypes of pancreatic cancer with different prognoses have been elaborated; however, the inability to model these subtypes has precluded mechanistic investigation of their origins. Here, we present a xenotransplantation model of PDAC in which neoplasms originate from patient-derived organoids injected directly into murine pancreatic ducts. Our model enables distinction of the two main PDAC subtypes: intraepithelial neoplasms from this model progress in an indolent or invasive manner representing the classical or basal-like subtypes of PDAC, respectively. Parameters that influence PDAC subtype specification in this intraductal model include cell plasticity and hyperactivation of the RAS pathway. Finally, through intratumoral dissection and the direct manipulation of RAS gene dosage, we identify a suite of RAS-regulated secreted and membrane-bound proteins that may represent potential candidates for therapeutic intervention in patients with PDAC.Significance:Accurate modeling of the molecular subtypes of pancreatic cancer is crucial to facilitate the generation of effective therapies. We report the development of an intraductal organoid transplantation model of pancreatic cancer that models the progressive switching of subtypes, and identify stochastic and RAS-driven mechanisms that determine subtype specification.See related commentary by Pickering and Morton, p. 1448.This article is highlighted in the In This Issue feature, p. 1426
- Published
- 2023
6. Supplementary Video3 from Intraductal Transplantation Models of Human Pancreatic Ductal Adenocarcinoma Reveal Progressive Transition of Molecular Subtypes
- Author
-
David A. Tuveson, Youngkyu Park, Steven Gallinger, Faiyaz Notta, Michael Wigler, Christopher R. Vakoc, Alexander Krasnitz, Jesse Gillis, Ralph H. Hruban, Laura D. Wood, Nicholas J. Roberts, Richard A. Burkhart, Chang-Il Hwang, Hervé Tiriac, Tim D.D. Somerville, Risa Karakida Kawaguchi, Gun Ho Jang, Jude Kendall, Siran Li, Pascal Belleau, Brinda Alagesan, Dennis Plenker, Giuseppina Caligiuri, Benno Traub, Astrid Deschênes, Lindsey A. Baker, and Koji Miyabayashi
- Abstract
Confocal z-stack imaging of immunofluorescent (IF) images of mStrawberry-hT3 grafts 4 weeks after IGO transplantation
- Published
- 2023
7. Supplementary Figure S5 from Intraductal Transplantation Models of Human Pancreatic Ductal Adenocarcinoma Reveal Progressive Transition of Molecular Subtypes
- Author
-
David A. Tuveson, Youngkyu Park, Steven Gallinger, Faiyaz Notta, Michael Wigler, Christopher R. Vakoc, Alexander Krasnitz, Jesse Gillis, Ralph H. Hruban, Laura D. Wood, Nicholas J. Roberts, Richard A. Burkhart, Chang-Il Hwang, Hervé Tiriac, Tim D.D. Somerville, Risa Karakida Kawaguchi, Gun Ho Jang, Jude Kendall, Siran Li, Pascal Belleau, Brinda Alagesan, Dennis Plenker, Giuseppina Caligiuri, Benno Traub, Astrid Deschênes, Lindsey A. Baker, and Koji Miyabayashi
- Abstract
Copy number analysis of invasive and intraductal regions of tumors derived from IGO transplants of clonal and Autobow organoids
- Published
- 2023
8. Supplementary Video4 from Intraductal Transplantation Models of Human Pancreatic Ductal Adenocarcinoma Reveal Progressive Transition of Molecular Subtypes
- Author
-
David A. Tuveson, Youngkyu Park, Steven Gallinger, Faiyaz Notta, Michael Wigler, Christopher R. Vakoc, Alexander Krasnitz, Jesse Gillis, Ralph H. Hruban, Laura D. Wood, Nicholas J. Roberts, Richard A. Burkhart, Chang-Il Hwang, Hervé Tiriac, Tim D.D. Somerville, Risa Karakida Kawaguchi, Gun Ho Jang, Jude Kendall, Siran Li, Pascal Belleau, Brinda Alagesan, Dennis Plenker, Giuseppina Caligiuri, Benno Traub, Astrid Deschênes, Lindsey A. Baker, and Koji Miyabayashi
- Abstract
Confocal z-stack imaging of immunofluorescent (IF) images of mStrawberry-hM1A grafts 4 weeks after IGO transplantation
- Published
- 2023
9. Supplementary Figure S8 from Intraductal Transplantation Models of Human Pancreatic Ductal Adenocarcinoma Reveal Progressive Transition of Molecular Subtypes
- Author
-
David A. Tuveson, Youngkyu Park, Steven Gallinger, Faiyaz Notta, Michael Wigler, Christopher R. Vakoc, Alexander Krasnitz, Jesse Gillis, Ralph H. Hruban, Laura D. Wood, Nicholas J. Roberts, Richard A. Burkhart, Chang-Il Hwang, Hervé Tiriac, Tim D.D. Somerville, Risa Karakida Kawaguchi, Gun Ho Jang, Jude Kendall, Siran Li, Pascal Belleau, Brinda Alagesan, Dennis Plenker, Giuseppina Caligiuri, Benno Traub, Astrid Deschênes, Lindsey A. Baker, and Koji Miyabayashi
- Abstract
KRAS activation promotes a more basal-like phenotype in vivo.
- Published
- 2023
10. Supplementary Video2 from Intraductal Transplantation Models of Human Pancreatic Ductal Adenocarcinoma Reveal Progressive Transition of Molecular Subtypes
- Author
-
David A. Tuveson, Youngkyu Park, Steven Gallinger, Faiyaz Notta, Michael Wigler, Christopher R. Vakoc, Alexander Krasnitz, Jesse Gillis, Ralph H. Hruban, Laura D. Wood, Nicholas J. Roberts, Richard A. Burkhart, Chang-Il Hwang, Hervé Tiriac, Tim D.D. Somerville, Risa Karakida Kawaguchi, Gun Ho Jang, Jude Kendall, Siran Li, Pascal Belleau, Brinda Alagesan, Dennis Plenker, Giuseppina Caligiuri, Benno Traub, Astrid Deschênes, Lindsey A. Baker, and Koji Miyabayashi
- Abstract
Confocal z-stack imaging of immunofluorescent (IF) images of mStrawberry-hM1A grafts 2 weeks after IGO transplantation
- Published
- 2023
11. Supplementary Figure S6 from Intraductal Transplantation Models of Human Pancreatic Ductal Adenocarcinoma Reveal Progressive Transition of Molecular Subtypes
- Author
-
David A. Tuveson, Youngkyu Park, Steven Gallinger, Faiyaz Notta, Michael Wigler, Christopher R. Vakoc, Alexander Krasnitz, Jesse Gillis, Ralph H. Hruban, Laura D. Wood, Nicholas J. Roberts, Richard A. Burkhart, Chang-Il Hwang, Hervé Tiriac, Tim D.D. Somerville, Risa Karakida Kawaguchi, Gun Ho Jang, Jude Kendall, Siran Li, Pascal Belleau, Brinda Alagesan, Dennis Plenker, Giuseppina Caligiuri, Benno Traub, Astrid Deschênes, Lindsey A. Baker, and Koji Miyabayashi
- Abstract
Comparison of invasive and intraductal regions of tumors derived from IGO transplants of Slow-progressor organoids
- Published
- 2023
12. Supplementary Table S1-S4 from Intraductal Transplantation Models of Human Pancreatic Ductal Adenocarcinoma Reveal Progressive Transition of Molecular Subtypes
- Author
-
David A. Tuveson, Youngkyu Park, Steven Gallinger, Faiyaz Notta, Michael Wigler, Christopher R. Vakoc, Alexander Krasnitz, Jesse Gillis, Ralph H. Hruban, Laura D. Wood, Nicholas J. Roberts, Richard A. Burkhart, Chang-Il Hwang, Hervé Tiriac, Tim D.D. Somerville, Risa Karakida Kawaguchi, Gun Ho Jang, Jude Kendall, Siran Li, Pascal Belleau, Brinda Alagesan, Dennis Plenker, Giuseppina Caligiuri, Benno Traub, Astrid Deschênes, Lindsey A. Baker, and Koji Miyabayashi
- Abstract
Supplementary Table S1 Survival times of IGO and OGO mice Supplementary Table S2 Characteristics of patient-derived organoids, including KRAS, TP53, SMAD4, CDKN2A mutation status and patient stage Supplementary Table S3 Summary of Survival Data, including engraftment rate, mean survival, metastatic frequency Supplementary Table S4 Primers for quantitative PCR
- Published
- 2023
13. Supplementary Figure S3 from Intraductal Transplantation Models of Human Pancreatic Ductal Adenocarcinoma Reveal Progressive Transition of Molecular Subtypes
- Author
-
David A. Tuveson, Youngkyu Park, Steven Gallinger, Faiyaz Notta, Michael Wigler, Christopher R. Vakoc, Alexander Krasnitz, Jesse Gillis, Ralph H. Hruban, Laura D. Wood, Nicholas J. Roberts, Richard A. Burkhart, Chang-Il Hwang, Hervé Tiriac, Tim D.D. Somerville, Risa Karakida Kawaguchi, Gun Ho Jang, Jude Kendall, Siran Li, Pascal Belleau, Brinda Alagesan, Dennis Plenker, Giuseppina Caligiuri, Benno Traub, Astrid Deschênes, Lindsey A. Baker, and Koji Miyabayashi
- Abstract
When used for IGO transplants, distinct organoid lines generate either Fast- or Slow-progressors.
- Published
- 2023
14. Supplementary Method SM1 from Intraductal Transplantation Models of Human Pancreatic Ductal Adenocarcinoma Reveal Progressive Transition of Molecular Subtypes
- Author
-
David A. Tuveson, Youngkyu Park, Steven Gallinger, Faiyaz Notta, Michael Wigler, Christopher R. Vakoc, Alexander Krasnitz, Jesse Gillis, Ralph H. Hruban, Laura D. Wood, Nicholas J. Roberts, Richard A. Burkhart, Chang-Il Hwang, Hervé Tiriac, Tim D.D. Somerville, Risa Karakida Kawaguchi, Gun Ho Jang, Jude Kendall, Siran Li, Pascal Belleau, Brinda Alagesan, Dennis Plenker, Giuseppina Caligiuri, Benno Traub, Astrid Deschênes, Lindsey A. Baker, and Koji Miyabayashi
- Abstract
Method Used to Define the Thresholds for Fast- and Slow-Progressing IGO-Derived Tumors
- Published
- 2023
15. Supplementary Figure S7 from Intraductal Transplantation Models of Human Pancreatic Ductal Adenocarcinoma Reveal Progressive Transition of Molecular Subtypes
- Author
-
David A. Tuveson, Youngkyu Park, Steven Gallinger, Faiyaz Notta, Michael Wigler, Christopher R. Vakoc, Alexander Krasnitz, Jesse Gillis, Ralph H. Hruban, Laura D. Wood, Nicholas J. Roberts, Richard A. Burkhart, Chang-Il Hwang, Hervé Tiriac, Tim D.D. Somerville, Risa Karakida Kawaguchi, Gun Ho Jang, Jude Kendall, Siran Li, Pascal Belleau, Brinda Alagesan, Dennis Plenker, Giuseppina Caligiuri, Benno Traub, Astrid Deschênes, Lindsey A. Baker, and Koji Miyabayashi
- Abstract
KRAS activation promotes a more basal-like phenotype in vitro.
- Published
- 2023
16. Supplementary Figure S4 from Intraductal Transplantation Models of Human Pancreatic Ductal Adenocarcinoma Reveal Progressive Transition of Molecular Subtypes
- Author
-
David A. Tuveson, Youngkyu Park, Steven Gallinger, Faiyaz Notta, Michael Wigler, Christopher R. Vakoc, Alexander Krasnitz, Jesse Gillis, Ralph H. Hruban, Laura D. Wood, Nicholas J. Roberts, Richard A. Burkhart, Chang-Il Hwang, Hervé Tiriac, Tim D.D. Somerville, Risa Karakida Kawaguchi, Gun Ho Jang, Jude Kendall, Siran Li, Pascal Belleau, Brinda Alagesan, Dennis Plenker, Giuseppina Caligiuri, Benno Traub, Astrid Deschênes, Lindsey A. Baker, and Koji Miyabayashi
- Abstract
PDAC molecular subtype is a plastic phenotype that can be influenced by stromal interactions.
- Published
- 2023
17. Pan-human consensus genome significantly improves the accuracy of RNA-seq analyses
- Author
-
Benjamin Kaminow, Sara Ballouz, Jesse Gillis, and Alexander Dobin
- Subjects
Consensus ,Genome, Human ,Exome Sequencing ,Genetics ,Humans ,Genomics ,RNA-Seq ,Genetics (clinical) - Abstract
The Human Reference Genome serves as the foundation for modern genomic analyses. However, in its present form, it does not adequately represent the vast genetic diversity of the human population. In this study, we explored the consensus genome as a potential successor of the current reference genome and assessed its effect on the accuracy of RNA-seq read alignment. To find the best haploid genome representation, we constructed consensus genomes at the pan-human, superpopulation, and population levels, using variant information from The 1000 Genomes Project Consortium. Using personal haploid genomes as the ground truth, we compared mapping errors for real RNA-seq reads aligned to the consensus genomes versus the reference genome. For reads overlapping homozygous variants, we found that the mapping error decreased by a factor of approximately two to three when the reference was replaced with the pan-human consensus genome. We also found that using more population-specific consensuses resulted in little to no increase over using the pan-human consensus, suggesting a limit in the utility of incorporating a more specific genomic variation. Replacing the reference with consensus genomes impacts functional analyses, such as differential expressions of isoforms, genes, and splice junctions.
- Published
- 2022
18. Cellular anatomy of the mouse primary motor cortex
- Author
-
Judith Mizrachi, Partha P. Mitra, Arun Narasimhan, Philip R. Nicovich, Sarojini M. Attili, Hideki Kondo, Pavel Osten, Muye Zhu, Brian Zingg, Anthony M. Zador, Stephan Fischer, William Galbavy, Uree Chon, Liya Ding, Stephanie Mok, Kwanghun Chung, Florence D’Orazi, Xu An, Shenqin Yao, Philip Lesnar, Wayne Wakemen, James C. Gee, Darrick Lo, Kathleen Kelly, Ian Bowman, Lydia Ng, Peng Xie, Quanxin Wang, Karla E. Hirokawa, X. William Yang, Julie A. Harris, Xiuli Kuang, Huizhong W. Tao, Samik Bannerjee, Elise Shen, Xu Li, Z. Josh Huang, Ali Cetin, Young Gyun Park, Lijuan Liu, Corey Elowsky, Xiangning Li, Lin Gou, Hong-Wei Dong, Laura Korobkova, Joshua T. Hatfield, Junxiang Jason Huang, Hui Gong, Yun Wang, Houri Hintiryan, Nicholas N. Foster, Peter A. Groblewski, Michael S. Bienkowski, Diek W. Wheeler, Xiaoyin Chen, Yu-Chi Sun, Anastasiia Bludova, Maitham Naeemi, Rodrigo Muñoz-Castañeda, Joel D. Hahn, Jing Yuan, Hanchuan Peng, Katherine Matho, Jason Palmer, Huiqing Zhan, Yimin Wang, Hongkui Zeng, Michael Hawrylycz, Chris Sin Park, Li I. Zhang, Rhonda Drewes, Ramesh Palaniswamy, Bing-Xing Huo, Anan Li, Yongsoo Kim, Jesse Gillis, Byung Kook Lim, Lei Gao, Giorgio A. Ascoli, Xiaoli Qi, Meng Kuan Lin, Yaoyao Li, and Qingming Luo
- Subjects
Male ,Computer science ,Neuroimaging ,Neural circuits ,Article ,Mice ,Atlases as Topic ,Glutamates ,medicine ,Biological neural network ,Animals ,GABAergic Neurons ,Neurons ,Multidisciplinary ,Sequence Analysis, RNA ,Brain atlas ,Motor Cortex ,Motor control ,Neuroinformatics ,Cellular Anatomy ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Cellular resolution ,Organ Specificity ,Female ,Single-Cell Analysis ,Primary motor cortex ,Neuroscience ,Motor cortex - Abstract
An essential step toward understanding brain function is to establish a structural framework with cellular resolution on which multi-scale datasets spanning molecules, cells, circuits and systems can be integrated and interpreted1. Here, as part of the collaborative Brain Initiative Cell Census Network (BICCN), we derive a comprehensive cell type-based anatomical description of one exemplar brain structure, the mouse primary motor cortex, upper limb area (MOp-ul). Using genetic and viral labelling, barcoded anatomy resolved by sequencing, single-neuron reconstruction, whole-brain imaging and cloud-based neuroinformatics tools, we delineated the MOp-ul in 3D and refined its sublaminar organization. We defined around two dozen projection neuron types in the MOp-ul and derived an input–output wiring diagram, which will facilitate future analyses of motor control circuitry across molecular, cellular and system levels. This work provides a roadmap towards a comprehensive cellular-resolution description of mammalian brain architecture., Multi-modal analysis is used to generate a 3D atlas of the upper limb area of the mouse primary motor cortex, providing a framework for future studies of motor control circuitry.
- Published
- 2021
19. Author response: Prevalent and dynamic binding of the cell cycle checkpoint kinase Rad53 to gene promoters
- Author
-
Yi-Jun Sheu, Risa Karakida Kawaguchi, Jesse Gillis, and Bruce Stillman
- Published
- 2022
20. Modular cell type organization of cortical areas revealed by in situ sequencing
- Author
-
Xiaoyin Chen, Stephan Fischer, Aixin Zhang, Jesse Gillis, and Anthony M Zador
- Abstract
The cortex is composed of neuronal types with diverse gene expression that are organized into specialized cortical areas. These areas, each with characteristic cytoarchitecture1–3, connectivity4,5, and neuronal activity6–10, are wired into modular networks4,5,11. However, it remains unclear whether cortical areas and their modular organization can be similarly defined by their transcriptomic signatures. Here we used BARseq, a high-throughput in situ sequencing technique, to interrogate the expression of 107 cell type marker genes in 1.2 million cells over a mouse forebrain hemisphere at cellular resolution.De novoclustering of gene expression in single neurons revealed transcriptomic types that were consistent with previous single-cell RNAseq studies12,13. Within medium-grained cell types that are shared across all cortical areas, gene expression and the distribution of fine-grained cell types vary along the contours of cortical areas. The compositions of transcriptomic types are highly predictive of cortical area identity. We grouped cortical areas into modules so that areas within a module, but not across modules, had similar compositions of transcriptomic types. Strikingly, these modules match cortical subnetworks that are highly interconnected4,5,11, suggesting that cortical areas that are similar in cell types are also wired together. This “wire-by-similarity” rule reflects a novel organizing principle for the connectivity of cortical areas. Our BARseq-based strategy is high-throughput and low-cost, and scaling up this approach to many animals can potentially reveal the brain-wide molecular architecture across individuals, developmental times, and disease models.
- Published
- 2022
21. The BRAIN Initiative Cell Census Network Data Ecosystem: A User’s Guide
- Author
-
Maryann Martone, Satrajit Ghosh, Giorgio Ascoli, Patrick Hof, Rongxin Fang, David Osumi-Sutherland, Jesse Gillis, and Jan G. Bjaalie
- Abstract
Characterizing cellular diversity at different levels of biological organization across data modalities is a prerequisite to understanding the function of cell types in the brain. Classification of neurons is also required to manipulate cell types in controlled ways, and to understand their variation and vulnerability in brain disorders. TheBRAIN Initiative Cell Census Network (BICCN)is an integrated network of data generating centers, data archives and data standards developers, with the goal of systematic multimodal brain cell type profiling and characterization. Emphasis of the BICCN is on the whole mouse brain and demonstration of prototypes for human and non-human primate (NHP) brains. Here, we provide a guide to the cellular and spatial approaches employed, and to accessing and using the BICCN data and its extensive resources, including theBRAIN Cell Data Center (BCDC)which serves to manage and integrate data across the ecosystem. We illustrate the power of the BICCN data ecosystem through vignettes highlighting several BICCN analysis and visualization tools. Finally, we present emerging standards that have been developed or adopted by the BICCN toward FAIR (Wilkinson et al. 2016a) neuroscience. The combined BICCN ecosystem provides a comprehensive resource for the exploration and analysis of cell types in the brain.
- Published
- 2022
22. Prevalent and dynamic binding of the cell cycle checkpoint kinase Rad53 to gene promoters
- Author
-
Risa Karakida Kawaguchi, Yi-Jun Sheu, Bruce Stillman, and Jesse Gillis
- Subjects
DNA Replication ,Saccharomyces cerevisiae Proteins ,DNA replication initiation ,General Immunology and Microbiology ,General Neuroscience ,Cell Cycle ,Promoter ,Cell Cycle Proteins ,General Medicine ,Saccharomyces cerevisiae ,Cell Cycle Checkpoints ,Cell cycle ,Biology ,Protein Serine-Threonine Kinases ,General Biochemistry, Genetics and Molecular Biology ,Chromatin ,Cell biology ,Checkpoint Kinase 2 ,Transcription (biology) ,Gene expression ,Phosphorylation ,Gene ,Transcription factor ,DNA Damage - Abstract
Replication of the genome must be coordinated with gene transcription and cellular metabolism, especially following replication stress in the presence of limiting deoxyribonucleotides. TheS. cerevisiaeRad53 (CHEK2 in mammals) checkpoint kinase plays a major role in cellular responses to DNA replication stress. Cell cycle regulated, genome-wide binding of Rad53 to chromatin was examined. Under replication stress, the kinase bound to sites of active DNA replication initiation and fork progression, but unexpectedly to the promoters of about 20% of genes encoding proteins involved in multiple cellular functions. Rad53 promoter binding correlated with changes in expression of a subset of genes. Rad53 promoter binding to certain genes was influenced by sequence-specific transcription factors and less by checkpoint signaling. However, in checkpoint mutants, untimely activation of late-replicating origins reduces the transcription of nearby genes, with concomitant localization of Rad53 to their gene bodies. We suggest that the Rad53 checkpoint kinase coordinates genome-wide replication and transcription under replication stress conditions.
- Published
- 2022
23. Conserved coexpression at single cell resolution across primate brains
- Author
-
Hamsini Suresh, Megan Crow, Nikolas Jorstad, Rebecca Hodge, Ed Lein, Alexander Dobin, Trygve Bakken, and Jesse Gillis
- Abstract
Enhanced cognitive function in humans is hypothesized to result from cortical expansion and increased cellular diversity. However, the mechanisms that drive these phenotypic differences remain poorly understood, in part due to the lack of high-quality cellular resolution data in human and non-human primates. Here, we take advantage of single cell expression data from the middle temporal gyrus of five primates (human, chimp, gorilla, macaque and marmoset) to identify 57 homologous cell types and generate cell-type specific gene coexpression networks for comparative analysis. While ortholog expression patterns are generally well conserved, we find 24% of genes with extensive differences between human and non-human primates (3383/14,131), which are also associated with multiple brain disorders. To validate these observations, we perform a meta-analysis of coexpression networks across 19 animals, and find that a subset of these genes have deeply conserved coexpression across all non-human animals, and strongly divergent coexpression relationships in humans (139/3383, NHEJ1, GTF2H2, C2 and BBS5) typically evolve under relaxed selective constraints and may drive rapid evolutionary change in brain function.One Sentence SummaryCross-primate middle temporal gyrus single cell expression data reveals patterns of conservation and divergence that can be validated with population coexpression networks.
- Published
- 2022
24. Comparative transcriptomics reveals human-specific cortical features
- Author
-
Nikolas L. Jorstad, Janet H.T. Song, David Exposito-Alonso, Hamsini Suresh, Nathan Castro, Fenna M. Krienen, Anna Marie Yanny, Jennie Close, Emily Gelfand, Kyle J. Travaglini, Soumyadeep Basu, Marc Beaudin, Darren Bertagnolli, Megan Crow, Song-Lin Ding, Jeroen Eggermont, Alexandra Glandon, Jeff Goldy, Thomas Kroes, Brian Long, Delissa McMillen, Trangthanh Pham, Christine Rimorin, Kimberly Siletti, Saroja Somasundaram, Michael Tieu, Amy Torkelson, Katelyn Ward, Guoping Feng, William D. Hopkins, Thomas Höllt, C. Dirk Keene, Sten Linnarsson, Steven A. McCarroll, Boudewijn P. Lelieveldt, Chet C. Sherwood, Kimberly Smith, Christopher A. Walsh, Alexander Dobin, Jesse Gillis, Ed S. Lein, Rebecca D. Hodge, and Trygve E. Bakken
- Abstract
Humans have unique cognitive abilities among primates, including language, but their molecular, cellular, and circuit substrates are poorly understood. We used comparative single nucleus transcriptomics in adult humans, chimpanzees, gorillas, rhesus macaques, and common marmosets from the middle temporal gyrus (MTG) to understand human-specific features of cellular and molecular organization. Human, chimpanzee, and gorilla MTG showed highly similar cell type composition and laminar organization, and a large shift in proportions of deep layer intratelencephalic-projecting neurons compared to macaque and marmoset. Species differences in gene expression generally mirrored evolutionary distance and were seen in all cell types, although chimpanzees were more similar to gorillas than humans, consistent with faster divergence along the human lineage. Microglia, astrocytes, and oligodendrocytes showed accelerated gene expression changes compared to neurons or oligodendrocyte precursor cells, indicating either relaxed evolutionary constraints or positive selection in these cell types. Only a few hundred genes showed human-specific patterning in all or specific cell types, and were significantly enriched near human accelerated regions (HARs) and conserved deletions (hCONDELS) and in cell adhesion and intercellular signaling pathways. These results suggest that relatively few cellular and molecular changes uniquely define adult human cortical structure, particularly by affecting circuit connectivity and glial cell function.
- Published
- 2022
25. Scaling up reproducible research for single-cell transcriptomics using MetaNeighbor
- Author
-
Jesse Gillis, Stephan Fischer, Megan Crow, and Benjamin Harris
- Subjects
Computer science ,Datasets as Topic ,computer.software_genre ,Article ,General Biochemistry, Genetics and Molecular Biology ,Bioconductor ,Mice ,Software ,Animals ,Humans ,Generalizability theory ,Independence (probability theory) ,Protocol (science) ,business.industry ,Brain ,Reproducibility of Results ,Visualization ,Gene Expression Regulation ,Scalability ,Data mining ,Single-Cell Analysis ,Transcriptome ,business ,computer - Abstract
Single cell RNA-sequencing data have significantly advanced the characterization of cell type diversity and composition. However, cell type definitions vary across data and analysis pipelines, raising concerns about cell type validity and generalizability. With MetaNeighbor, we proposed an efficient and robust quantification of cell type replicability that preserves dataset independence and is highly scalable compared to dataset integration. In this protocol, we show how MetaNeighbor can be used to characterize cell type replicability by following a simple 3-step procedure: gene filtering, neighbor voting, and visualization. We show how these steps can be tailored to quantify cell type replicability, determine gene sets that contribute to cell type identity, and pretrain a model on a reference taxonomy to rapidly assess newly generated data. The protocol is based on an open-source R package available from Bioconductor and Github, requires basic familiarity with Rstudio or the R command line, and can typically be run in less than 5 minutes for millions of cells.
- Published
- 2021
26. Gene duplication and cellular divergence in crops
- Author
-
Kenneth Birnbaum, Bruno Guillotin, Ramin Rahni, Michael Passalacqua, Mohammed Mohammed, Xiaosa Xu, David Jackson, Simon Groen, and Jesse Gillis
- Abstract
Different plant species within the grasses were parallel targets of domestication, giving rise to crops with distinct evolutionary histories and traits. Key traits that distinguish these species are mediated by specialized cell types within organs. Here, we compare the transcriptomes of all cells within roots in three grasses—Zea mays (maize), Sorghum bicolor (sorghum), and outgroup Setaria viridis (Setaria). We first show that single-cell and single-nucleus RNA-seq provide complementary readouts of cell identity, warranting a combined analysis. Comparative cellular analysis shows that the transcriptomes of some cell types diverged more rapidly than others, in part by recruiting gene modules from other cell types. Furthermore, examining the whole genome duplication in maize, we detect extensive dosage compensation in surviving co-expressed homeologs, reinforcing genomic balance1. Homeolog pairs that underwent subfunctionalization2, partitioning their expression among cell types, represented a minor pattern but showed the highest rate of acquiring a novel (non-ancestral) domain. These results fit a conjecture in which mechanisms that maintain stoichiometric balance at the molecular level aid in homeolog retention for extended periods to allow new functions to arise. An unexpected synergy between spatial sub- and neo-functionalization then contributes to changes in transcriptional cell identity.
- Published
- 2022
27. Integrating barcoded neuroanatomy with spatial transcriptional profiling enables identification of gene correlates of projections
- Author
-
Yu-Chi Sun, Huiqing Zhan, Shaina Lu, Anthony M. Zador, Stephan Fischer, Jesse Gillis, and Xiaoyin Chen
- Subjects
Male ,0301 basic medicine ,Biology ,Auditory cortex ,Article ,Transcriptome ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Transcription (biology) ,Neural Pathways ,Gene expression ,medicine ,Animals ,Gene Regulatory Networks ,Gene ,Auditory Cortex ,Brain Mapping ,Electronic Data Processing ,Cadherin ,General Neuroscience ,Motor Cortex ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Neuroscience ,030217 neurology & neurosurgery ,Neuroanatomy ,Motor cortex - Abstract
Functional circuits consist of neurons with diverse axonal projections and gene expression. Understanding the molecular signature of projections requires high-throughput interrogation of both gene expression and projections to multiple targets in the same cells at cellular resolution, which is difficult to achieve using current technology. Here, we introduce BARseq2, a technique that simultaneously maps projections and detects multiplexed gene expression by in situ sequencing. We determined the expression of cadherins and cell-type markers in 29,933 cells, and the projections of 3,164 cells in both the mouse motor cortex and auditory cortex. Associating gene expression and projections in 1,349 neurons revealed shared cadherin signatures of homologous projections across the two cortical areas. These cadherins were enriched across multiple branches of the transcriptomic taxonomy. By correlating multi-gene expression and projections to many targets in single neurons with high throughput, BARseq2 provides a potential path to uncovering the molecular logic underlying neuronal circuits.
- Published
- 2021
28. Conserved cell-type specific signature of resilience to Alzheimer’s disease nominates role for excitatory intratelencephalic cortical neurons
- Author
-
Maria A. Telpoukhovskaia, Niran Hadad, Brianna Gurdon, Yanchao Dai, Andrew R. Ouellette, Sarah M. Neuner, Amy R. Dunn, Jon A. L. Willcox, Yiyang Wu, Logan Dumitrescu, Orhan Bellur, Ji-Gang Zhang, Kristen M.S. O’Connell, Eric B. Dammer, Nicholas T. Seyfried, Sukalp Muzumdar, Jesse Gillis, Paul Robson, Matthias Arnold, Timothy J. Hohman, Vivek M. Philip, Vilas Menon, and Catherine C. Kaczorowski
- Abstract
SummaryAlzheimer’s disease (AD), the leading cause of dementia, affects millions of people worldwide. With no disease-modifying medication currently available, the human toll and economic costs are rising rapidly. Under current standards, a patient is diagnosed with AD when both cognitive decline and pathology (amyloid plaques and neurofibrillary tangles) are present. Remarkably, some individuals who have AD pathology remain cognitively normal. Uncovering factors that lead to “cognitive resilience” to AD is a promising path to create new targets for therapies. However, technical challenges discovering novel human resilience factors limit testing, validation, and nomination of novel drugs for AD. In this study, we use single-nucleus transcriptional profiles of postmortem cortex from human individuals with high AD pathology who were either cognitively normal (resilient) or cognitively impaired (susceptible) at time of death, as well as mouse strains that parallel these differences in cognition with high amyloid load. Our cross-species discovery approach highlights a novel role for excitatory layer 4/5 cortical neurons in promoting cognitive resilience to AD, and nominates several resilience genes that includeATP1A1,GRIA3,KCNMA1, andSTXBP1. This putative cell type has been implicated in resilience in previous studies on bulk RNA-seq tissue, but our single-nucleus and cross-species approach identifies particular resilience-associated gene signatures in these cells. These novel resilience candidate genes were tested for replication in orthogonal data sets and confirmed to be correlated with cognitive resilience. Based on these gene signatures, we identified several potential mechanisms of resilience, including regulation of synaptic plasticity, axonal and dendritic development, and neurite vesicle transport along microtubules that are potentially targetable by available therapeutics. Because our discovery of resilience-associated genes in layer 4/5 cortical neurons originates from an integrated human and mouse transcriptomic space from susceptible and resilient individuals, we are positioned to test causality and perform mechanistic, validation, and pre-clinical studies in our human-relevant AD-BXD mouse panel.
- Published
- 2022
29. Coexpression reveals conserved gene programs that co-vary with cell type across kingdoms
- Author
-
Megan Crow, Hamsini Suresh, John Lee, and Jesse Gillis
- Subjects
Mice ,Gene Expression Regulation ,Organ Specificity ,Gene Expression Profiling ,Genetics ,Animals ,Gene Regulatory Networks - Abstract
What makes a mouse a mouse, and not a hamster? Differences in gene regulation between the two organisms play a critical role. Comparative analysis of gene coexpression networks provides a general framework for investigating the evolution of gene regulation across species. Here, we compare coexpression networks from 37 species and quantify the conservation of gene activity 1) as a function of evolutionary time, 2) across orthology prediction algorithms, and 3) with reference to cell- and tissue-specificity. We find that ancient genes are expressed in multiple cell types and have well conserved coexpression patterns, however they are expressed at different levels across cell types. Thus, differential regulation of ancient gene programs contributes to transcriptional cell identity. We propose that this differential regulation may play a role in cell diversification in both the animal and plant kingdoms.
- Published
- 2022
30. A global high-density chromatin interaction network reveals functional long-range and trans-chromosomal relationships
- Author
-
Ruchi Lohia, Nathan Fox, and Jesse Gillis
- Subjects
Mice ,Quantitative Trait Loci ,Humans ,Animals ,Chromosome Mapping ,Genomics ,Chromatin ,Chromosomes - Abstract
Background Chromatin contacts are essential for gene-expression regulation; however, obtaining a high-resolution genome-wide chromatin contact map is still prohibitively expensive owing to large genome sizes and the quadratic scale of pairwise data. Chromosome conformation capture (3C)-based methods such as Hi-C have been extensively used to obtain chromatin contacts. However, since the sparsity of these maps increases with an increase in genomic distance between contacts, long-range or trans-chromatin contacts are especially challenging to sample. Results Here, we create a high-density reference genome-wide chromatin contact map using a meta-analytic approach. We integrate 3600 human, 6700 mouse, and 500 fly Hi-C experiments to create species-specific meta-Hi-C chromatin contact maps with 304 billion, 193 billion, and 19 billion contacts in respective species. We validate that meta-Hi-C contact maps are uniquely powered to capture functional chromatin contacts in both cis and trans. We find that while individual dataset Hi-C networks are largely unable to predict any long-range coexpression (median 0.54 AUC), meta-Hi-C networks perform comparably in both cis and trans (0.65 AUC vs 0.64 AUC). Similarly, for long-range expression quantitative trait loci (eQTL), meta-Hi-C contacts outperform all individual Hi-C experiments, providing an improvement over the conventionally used linear genomic distance-based association. Assessing between species, we find patterns of chromatin contact conservation in both cis and trans and strong associations with coexpression even in species for which Hi-C data is lacking. Conclusions We have generated an integrated chromatin interaction network which complements a large number of methodological and analytic approaches focused on improved specificity or interpretation. This high-depth “super-experiment” is surprisingly powerful in capturing long-range functional relationships of chromatin interactions, which are now able to predict coexpression, eQTLs, and cross-species relationships. The meta-Hi-C networks are available at https://labshare.cshl.edu/shares/gillislab/resource/HiC/.
- Published
- 2022
31. Integrative analysis methods for spatial transcriptomics
- Author
-
Daniel Fürth, Shaina Lu, and Jesse Gillis
- Subjects
Computer science ,Spatially resolved ,genetic processes ,Computational Biology ,Cell Biology ,Computational biology ,Biochemistry ,Article ,Imaging ,Machine learning ,Transcriptome ,Transcriptomics ,Molecular Biology ,Analysis method ,Software ,Biotechnology ,Neuroscience - Abstract
Charting an organs’ biological atlas requires us to spatially resolve the entire single-cell transcriptome, and to relate such cellular features to the anatomical scale. Single-cell and single-nucleus RNA-seq (sc/snRNA-seq) can profile cells comprehensively, but lose spatial information. Spatial transcriptomics allows for spatial measurements, but at lower resolution and with limited sensitivity. Targeted in situ technologies solve both issues, but are limited in gene throughput. To overcome these limitations we present Tangram, a method that aligns sc/snRNA-seq data to various forms of spatial data collected from the same region, including MERFISH, STARmap, smFISH, Spatial Transcriptomics (Visium) and histological images. Tangram can map any type of sc/snRNA-seq data, including multimodal data such as those from SHARE-seq, which we used to reveal spatial patterns of chromatin accessibility. We demonstrate Tangram on healthy mouse brain tissue, by reconstructing a genome-wide anatomically integrated spatial map at single-cell resolution of the visual and somatomotor areas., Tangram is a versatile tool for aligning single-cell and single-nucleus RNA-seq data to spatially resolved transcriptomics data using deep learning.
- Published
- 2021
32. Variability of cross-tissue X-chromosome inactivation characterizes timing of human embryonic lineage specification events
- Author
-
Jonathan M. Werner, Sara Ballouz, John Hover, and Jesse Gillis
- Subjects
Adult ,Mammals ,Chromosomes, Human, X ,X Chromosome Inactivation ,Animals ,Humans ,Cell Biology ,Embryo, Mammalian ,Molecular Biology ,General Biochemistry, Genetics and Molecular Biology ,Developmental Biology - Abstract
X-chromosome inactivation (XCI) is a random, permanent, and developmentally early epigenetic event that occurs during mammalian embryogenesis. We harness these features to investigate characteristics of early lineage specification events during human development. We initially assess the consistency of X-inactivation and establish a robust set of XCI-escape genes. By analyzing variance in XCI ratios across tissues and individuals, we find that XCI is shared across all tissues, suggesting that XCI is completed in the epiblast (in at least 6-16 cells) prior to specification of the germ layers. Additionally, we exploit tissue-specific variability to characterize the number of cells present during tissue-lineage commitment, ranging from approximately 20 cells in liver and whole blood tissues to 80 cells in brain tissues. By investigating the variability of XCI ratios using adult tissue, we characterize embryonic features of human XCI and lineage specification that are otherwise difficult to ascertain experimentally.
- Published
- 2021
33. Cross-tissue analysis of allelic X-chromosome inactivation ratios resolves features of human development
- Author
-
Jesse Gillis, John Hover, Jonathan Werner, and Sara Ballouz
- Subjects
Lineage commitment ,Evolutionary biology ,Embryogenesis ,Tissue specific ,Chromosome ,Epigenetics ,Biology ,Allele ,Gene ,X-inactivation - Abstract
X-chromosome inactivation (XCI) is a random, permanent, and developmentally early epigenetic event that occurs during mammalian embryogenesis. We harness these features of XCI to investigate characteristics of early lineage specification events during human development. We initially assess the consistency of X-inactivation and establish a robust set of XCI-escape genes. By analyzing variance in XCI ratios across tissues and individuals, we find that XCI is completed prior to tissue specification and at a time when 6-16 cells are fated for all tissue lineages. Additionally, we exploit tissue specific variability to characterize the number of cells present at the time of each tissue’s lineage commitment, ranging from approximately 20 cells in liver and whole blood tissues to 80 cells in brain tissues. By investigating variance of XCI ratios using adult tissue, we resolve key features of human development otherwise difficult to ascertain experimentally and develop scalable methods easily applicable to future data.
- Published
- 2021
34. Defining the extent of gene function using ROC curvature
- Author
-
Stephan Fischer and Jesse Gillis
- Subjects
Identification (information) ,Range (mathematics) ,Receiver operating characteristic ,Computer science ,Robustness (evolution) ,Genomics ,Generalizability theory ,Computational biology ,Function (mathematics) ,Genome - Abstract
Machine learning in genomics plays a key role in leveraging high-throughput data, but assessing the generalizability of performance has been a persistent challenge. Here, we propose to evaluate the generalizability of gene characterizations through the shape of performance curves. We identify Functional Equivalence Classes (FECs), uniform subsets of annotated and unannotated genes that jointly drive performance, by assessing the presence of straight lines in ROC curves. FECs are widespread across modalities and methods, and can be used to evaluate the extent and context-specificity of functional annotations in a data-driven manner. For example, FECs suggest that B cell markers can be decomposed into shared primary markers (10 to 50 genes), and tissue-specific secondary markers (100 to 500□genes). In addition, FECs are compatible with a wide range of functional encodings, with marker sets spanning at most 5% of the genome and data-driven extensions of Gene Ontology sets spanning up to 40% of the genome. Simple to assess visually and statistically, the identification of FECs in performance curves paves the way for novel functional characterization and increased robustness in analysis.
- Published
- 2021
35. A Meta-Analytic Single-Cell Atlas of Mouse Bone Marrow Hematopoietic Development
- Author
-
Jesse Gillis, Benjamin Harris, and John Lee
- Subjects
Haematopoiesis ,Cell type ,medicine.anatomical_structure ,Lineage (genetic) ,In silico ,Cell ,medicine ,Computational biology ,Bone marrow ,Biology ,Progenitor cell ,biology.organism_classification ,Zebrafish - Abstract
The clinical importance of the hematopoietic system makes it one of the most heavily studied lineages in all of biology. A clear understanding of the cell types and functional programs during hematopoietic development is central to research in aging, cancer, and infectious diseases. Known cell types are traditionally identified by the expression of proteins on the surface of the cells. Stem and progenitor cells defined based on these markers are assigned functions based on their lineage potential. The rapid growth of single cell RNA sequencing technologies (scRNAseq) provides a new modality for evaluating the cellular and functional landscape of hematopoietic stem and progenitor cells. The popularity of this technology among hematopoiesis researchers enables us to conduct a robust meta-analysis of mouse bone marrow scRNAseq data. Using over 300,000 cells across 12 datasets, we evaluate the classification and function of cell types based on discrete clustering,in silicoFACS sorting, and a continuous trajectory. We identify replicable signatures that define cell types based on genes and known cellular functions. Additionally, we evaluate the conservation of signatures associated with erythroid and monocyte lineage development across species using co-expression networks. The co-expression networks predict the effectiveness of the signature at identifying erythroid and monocyte cells in zebrafish and human scRNAseq data. Together, this analysis provides a robust reference, particularly marker genes and functional annotations, for future experiments in hematopoietic development.Key PointsMeta-analysis of 9 mouse bone marrow scRNAseq identifies markers for cell types and hematopoietic developmentCharacterize a replicable functional landscape of cell types by exploiting co-expression
- Published
- 2021
36. Predictability of human differential gene expression
- Author
-
Paul Pavlidis, Megan Crow, Jesse Gillis, Sara Ballouz, and Nathaniel Lim
- Subjects
Graft Rejection ,0301 basic medicine ,Mutation rate ,Lung Neoplasms ,specificity ,Breast Neoplasms ,Genomics ,Computational biology ,Adenocarcinoma ,Biology ,Sensitivity and Specificity ,differential expression ,transcriptomics ,03 medical and health sciences ,0302 clinical medicine ,Recurrence ,Prior probability ,Biomarkers, Tumor ,replicability ,Humans ,Gene Regulatory Networks ,Predictability ,Probability ,Electronic Data Processing ,Genes, Essential ,Multidisciplinary ,Receiver operating characteristic ,Gene Expression Profiling ,Systems Biology ,Contrast (statistics) ,Human Genetics ,Biological Sciences ,Kidney Transplantation ,Phenotype ,Subtyping ,030104 developmental biology ,Gene Expression Regulation ,ROC Curve ,PNAS Plus ,030220 oncology & carcinogenesis ,Female ,Transcriptome ,metaanalysis - Abstract
Significance The identification of genes that are differentially expressed provides a molecular foothold onto biological questions of interest. Whether some genes are more likely to be differentially expressed than others, and to what degree, has never been assessed on a global scale. Here, we reanalyze more than 600 studies and find that knowledge of a gene’s prior probability of differential expression (DE) allows for accurate prediction of DE hit lists, regardless of the biological question. This result suggests redundancy in transcriptomics experiments that both informs gene set interpretation and highlights room for growth within the field., Differential expression (DE) is commonly used to explore molecular mechanisms of biological conditions. While many studies report significant results between their groups of interest, the degree to which results are specific to the question at hand is not generally assessed, potentially leading to inaccurate interpretation. This could be particularly problematic for metaanalysis where replicability across datasets is taken as strong evidence for the existence of a specific, biologically relevant signal, but which instead may arise from recurrence of generic processes. To address this, we developed an approach to predict DE based on an analysis of over 600 studies. A predictor based on empirical prior probability of DE performs very well at this task (mean area under the receiver operating characteristic curve, ∼0.8), indicating that a large fraction of DE hit lists are nonspecific. In contrast, predictors based on attributes such as gene function, mutation rates, or network features perform poorly. Genes associated with sex, the extracellular matrix, the immune system, and stress responses are prominent within the “DE prior.” In a series of control studies, we show that these patterns reflect shared biology rather than technical artifacts or ascertainment biases. Finally, we demonstrate the application of the DE prior to data interpretation in three use cases: (i) breast cancer subtyping, (ii) single-cell genomics of pancreatic islet cells, and (iii) metaanalysis of lung adenocarcinoma and renal transplant rejection transcriptomics. In all cases, we find hallmarks of generic DE, highlighting the need for nuanced interpretation of gene phenotypic associations.
- Published
- 2019
37. The EN-TEx resource of multi-tissue personal epigenomes & variant-impact models
- Author
-
Joel Rozowsky, Jorg Drenkow, Yucheng T Yang, Gamze Gursoy, Timur Galeev, Beatrice Borsari, Charles B Epstein, Kun Xiong, Jinrui Xu, Jiahao Gao, Keyang Yu, Ana Berthel, Zhanlin Chen, Fabio Navarro, Jason Liu, Maxwell S Sun, James Wright, Justin Chang, Christopher JF Cameron, Noam Shoresh, Elizabeth Gaskell, Jessika Adrian, Sergey Aganezov, François Aguet, Gabriela Balderrama-Gutierrez, Samridhi Banskota, Guillermo Barreto Corona, Sora Chee, Surya B Chhetri, Gabriel Conte Cortez Martins, Cassidy Danyko, Carrie A Davis, Daniel Farid, Nina P Farrell, Idan Gabdank, Yoel Gofin, David U Gorkin, Mengting Gu, Vivian Hecht, Benjamin C Hitz, Robbyn Issner, Melanie Kirsche, Xiangmeng Kong, Bonita R Lam, Shantao Li, Bian Li, Tianxiao Li, Xiqi Li, Khine Zin Lin, Ruibang Luo, Mark Mackiewicz, Jill E Moore, Jonathan Mudge, Nicholas Nelson, Chad Nusbaum, Ioann Popov, Henry E Pratt, Yunjiang Qiu, Srividya Ramakrishnan, Joe Raymond, Leonidas Salichos, Alexandra Scavelli, Jacob M Schreiber, Fritz J Sedlazeck, Lei Hoon See, Rachel M Sherman, Xu Shi, Minyi Shi, Cricket Alicia Sloan, J Seth Strattan, Zhen Tan, Forrest Y Tanaka, Anna Vlasova, Jun Wang, Jonathan Werner, Brian Williams, Min Xu, Chengfei Yan, Lu Yu, Christopher Zaleski, Jing Zhang, Kristin Ardlie, J Michael Cherry, Eric M Mendenhall, William S Noble, Zhiping Weng, Morgan E Levine, Alexander Dobin, Barbara Wold, Ali Mortazavi, Bing Ren, Jesse Gillis, Richard M Myers, Michael P Snyder, Jyoti Choudhary, Aleksandar Milosavljevic, Michael C Schatz, Roderic Guigó, Bradley E Bernstein, Thomas R Gingeras, and Mark Gerstein
- Subjects
Genetic variants ,Genomics ,Preprint ,Computational biology ,Biology ,Personal genomics - Abstract
Understanding how genetic variants impact molecular phenotypes is a key goal of functional genomics, currently hindered by reliance on a single haploid reference genome. Here, we present the EN-TEx resource of personal epigenomes, for ∼25 tissues and >10 assays in four donors (>1500 open-access functional genomic and proteomic datasets, in total). Each dataset is mapped to a matched, diploid personal genome, which has long-read phasing and structural variants. The mappings enable us to identify >1 million loci with allele-specific behavior. These loci exhibit coordinated epigenetic activity along haplotypes and less conservation than matched, non-allele-specific loci, in a fashion broadly paralleling tissue-specificity. Surprisingly, they can be accurately modelled just based on local nucleotide-sequence context. Combining EN-TEx with existing genome annotations reveals strong associations between allele-specific and GWAS loci and enables models for transferring known eQTLs to difficult-to-profile tissues. Overall, EN-TEx provides rich data and generalizable models for more accurate personal functional genomics.
- Published
- 2021
38. How many markers are needed to robustly determine a cell’s type?
- Author
-
Jesse Gillis and Stephan Fischer
- Subjects
Cell type ,Annotation ,medicine.anatomical_structure ,Gene panel ,Cell ,medicine ,Experimental validation ,Computational biology ,Pathway enrichment ,Biology ,Gene ,Selection (genetic algorithm) - Abstract
SummaryOur understanding of cell types has advanced considerably with the publication of single cell atlases. Marker genes play an essential role for experimental validation and computational analyses such as physiological characterization through pathway enrichment, annotation, and deconvolution. However, a framework for quantifying marker replicability and picking replicable markers is currently lacking. Here, using high quality data from the Brain Initiative Cell Census Network (BICCN), we systematically investigate marker replicability for 85 neuronal cell types. We show that, due to dataset-specific noise, we need to combine 5 datasets to obtain robust differentially expressed (DE) genes, particularly for rare populations and lowly expressed genes. We estimate that 10 to 200 meta-analytic markers provide optimal performance in downstream computational tasks. Replicable marker lists condense single cell atlases into interpretable and generalizable information about cell types, opening avenues for downstream applications, including cell type annotation, selection of gene panels and bulk data deconvolution.
- Published
- 2021
39. Learning single-cell chromatin accessibility profiles using meta-analytic marker genes
- Author
-
Risa Karakida Kawaguchi, Ziqi Tang, Stephan Fischer, Chandana Rajesh, Rohit Tripathy, Peter K. Koo, and Jesse Gillis
- Subjects
DNA binding site ,Regulation of gene expression ,Regulatory sequence ,Computer science ,Feature selection ,Computational biology ,Enhancer ,Molecular Biology ,Marker gene ,Gene ,Information Systems ,Chromatin - Abstract
Background Single-cell Assay for Transposase Accessible Chromatin using sequencing (scATAC-seq) measures genome-wide chromatin accessibility for the discovery of cell-type specific regulatory networks. ScATAC-seq combined with single-cell RNA sequencing (scRNA-seq) offers important avenues for ongoing research, such as novel cell-type specific activation of enhancer and transcription factor binding sites as well as chromatin changes specific to cell states. On the other hand, scATAC-seq data is known to be challenging to interpret due to its high number of zeros as well as the heterogeneity derived from different protocols. Because of the stochastic lack of marker gene activities, cell type identification by scATAC-seq remains difficult even at a cluster level. Results In this study, we exploit reference knowledge obtained from external scATAC-seq or scRNA-seq datasets to define existing cell types and uncover the genomic regions which drive cell-type specific gene regulation. To investigate the robustness of existing cell-typing methods, we collected 7 scATAC-seq datasets targeting mouse brain for a meta-analytic comparison of neuronal cell-type annotation, including a reference atlas generated by the BRAIN Initiative Cell Census Network (BICCN). By comparing the area under the receiver operating characteristics curves (AUROCs) for the three major cell types (inhibitory, excitatory, and non-neuronal cells), cell-typing performance by single markers is found to be highly variable even for known marker genes due to study-specific biases. How-ever, the signal aggregation of a large and redundant marker gene set, optimized via multiple scRNA-seq data, achieves the highest cell-typing performances among 5 existing marker gene sets, from the individual cell to cluster level. That gene set also shows a high consistency with the cluster-specific genes from inhibitory subtypes in two well-annotated datasets, suggesting applicability to rare cell types. Next, we demonstrate a comprehensive assessment of scATAC-seq cell typing using exhaustive combinations of the marker gene sets with supervised learning methods including machine learning classifiers and joint clustering methods. Our results show that the combinations using robust marker gene sets systematically ranked at the top, not only with model based prediction using a large reference data but also with a simple summation of expression strengths across markers. To demonstrate the utility of this robust cell typing approach, we trained a deep neural network to predict chromatin accessibility in each subtype using only DNA sequence. Through model interpretation methods, we identify key motifs enriched about robust gene sets for each neuronal subtype. Conclusions Through the meta-analytic evaluation of scATACseq cell-typing methods, we develop a novel method set to exploit the BICCN reference atlas. Our study strongly supports the value of robust marker gene selection as a feature selection tool and cross-dataset comparison between scATAC-seq datasets to improve alignment of scATAC-seq to known biology. With this novel, high quality epigenetic data, genomic analysis of regulatory regions can reveal sequence motifs that drive cell type-specific regulatory programs.
- Published
- 2021
40. Virtue as the mean: Pan-human consensus genome significantly improves the accuracy of RNA-seq analyses
- Author
-
Jesse Gillis, Alexander Dobin, Benjamin Kaminow, and Sara Ballouz
- Subjects
Genetic diversity ,education.field_of_study ,Population ,Functional impact ,RNA-Seq ,Computational biology ,Ploidy ,1000 Genomes Project ,Biology ,education ,Genome ,Reference genome - Abstract
The Human Reference Genome serves as the foundation for modern genomic analyses. However, in its present form, it does not adequately represent the vast genetic diversity of the human population. In this study, we explored the consensus genome as a potential successor of the current Reference genome and assessed its effect on the accuracy of RNA-seq read alignment. In order to find the best haploid genome representation, we constructed consensus genomes at the Pan-human, Super-population and Population levels, utilizing variant information from the 1000 Genomes project. Using personal haploid genomes as the ground truth, we compared mapping errors for real RNA-seq reads aligned to the consensus genomes versus the Reference genome. For reads overlapping homozygous variants, we found that the mapping error decreased by a factor of ~2-3 when the Reference was replaced with the Pan-human consensus genome. Interestingly, we also found that using more population-specific consensuses resulted in little to no increase over using the Pan-human consensus, suggesting a limit in the utility of incorporating more specific genomic variation. To assess the functional impact, we compared splice junction expression in the different genomes and found that the Pan-human consensus increases accuracy of splice junction quantification for hundreds of splice junctions.
- Published
- 2020
41. A granular view of X-linked chronic granulomatous disease exploiting single-cell transcriptomics
- Author
-
Sukalp Muzumdar, Sara Ballouz, Fung Lam, Maureen Degrange, Samantha Kreuzburg, Hey Chong, Christa Zerbe, Artemio Jongco, and Jesse Gillis
- Subjects
Immunology ,Immunology and Allergy - Abstract
X-linked chronic granulomatous disease (X-CGD) is a rare monogenetic immunodeficiency primarily affecting phagocytes. Precipitated by mutations in the CYBB gene, patients exhibit a compromised oxidative burst, leading to recurrent infections which can be life-threatening. Curiously, autoimmune manifestations are also common in patients and carriers. Here, exploiting the cell type-specific nature of this disorder, we characterize X-CGD on a transcriptional level using single-cell sequencing. Peripheral blood from 14 X-CGD probands and 10 carriers signed onto IRB approved protocol NCT00404560, as well as from 15 controls was sampled, and PBMCs and isolated monocytes were subjected to single-cell sequencing. Probands exhibited a strong differential expression signal relative to controls. This was composed of not only genes previously described to be up-regulated in X-CGD such as IFI27, and indeed an autoimmunity-associated broader type I interferon response, but also previously undescribed genes involved in monocyte function (ARG1), antimicrobial proteins (CAMP, SLPI), and inflammasome components (AIM2). Surprisingly, expression variability was not greater in carriers relative to probands or controls, indicating a lack of cell autonomous effects from the deletion of CYBB. Interestingly, aggregate expression of differentially expressed genes in the probands was able to classify carriers from sex-matched controls with high accuracy (AUROC = 0.92), indicating the presence of an X-CGD-specific gene signature. This gene signature was also strongly co-expressed across 17 chordate species, pointing towards the disruption of ancestral pathways important in antimicrobial immunity in X-CGD probands and carriers. This work was partially supported by a Swiss National Science Foundation fellowship to S.M.
- Published
- 2022
42. Author Correction: Comparative cellular analysis of motor cortex in human, marmoset and mouse
- Author
-
Trygve E. Bakken, Nikolas L. Jorstad, Qiwen Hu, Blue B. Lake, Wei Tian, Brian E. Kalmbach, Megan Crow, Rebecca D. Hodge, Fenna M. Krienen, Staci A. Sorensen, Jeroen Eggermont, Zizhen Yao, Brian D. Aevermann, Andrew I. Aldridge, Anna Bartlett, Darren Bertagnolli, Tamara Casper, Rosa G. Castanon, Kirsten Crichton, Tanya L. Daigle, Rachel Dalley, Nick Dee, Nikolai Dembrow, Dinh Diep, Song-Lin Ding, Weixiu Dong, Rongxin Fang, Stephan Fischer, Melissa Goldman, Jeff Goldy, Lucas T. Graybuck, Brian R. Herb, Xiaomeng Hou, Jayaram Kancherla, Matthew Kroll, Kanan Lathia, Baldur van Lew, Yang Eric Li, Christine S. Liu, Hanqing Liu, Jacinta D. Lucero, Anup Mahurkar, Delissa McMillen, Jeremy A. Miller, Marmar Moussa, Joseph R. Nery, Philip R. Nicovich, Sheng-Yong Niu, Joshua Orvis, Julia K. Osteen, Scott Owen, Carter R. Palmer, Thanh Pham, Nongluk Plongthongkum, Olivier Poirion, Nora M. Reed, Christine Rimorin, Angeline Rivkin, William J. Romanow, Adriana E. Sedeño-Cortés, Kimberly Siletti, Saroja Somasundaram, Josef Sulc, Michael Tieu, Amy Torkelson, Herman Tung, Xinxin Wang, Fangming Xie, Anna Marie Yanny, Renee Zhang, Seth A. Ament, M. Margarita Behrens, Hector Corrada Bravo, Jerold Chun, Alexander Dobin, Jesse Gillis, Ronna Hertzano, Patrick R. Hof, Thomas Höllt, Gregory D. Horwitz, C. Dirk Keene, Peter V. Kharchenko, Andrew L. Ko, Boudewijn P. Lelieveldt, Chongyuan Luo, Eran A. Mukamel, António Pinto-Duarte, Sebastian Preiss, Aviv Regev, Bing Ren, Richard H. Scheuermann, Kimberly Smith, William J. Spain, Owen R. White, Christof Koch, Michael Hawrylycz, Bosiljka Tasic, Evan Z. Macosko, Steven A. McCarroll, Jonathan T. Ting, Hongkui Zeng, Kun Zhang, Guoping Feng, Joseph R. Ecker, Sten Linnarsson, and Ed S. Lein
- Subjects
Multidisciplinary - Published
- 2022
43. Coexpression reveals conserved mechanisms of transcriptional cell identity
- Author
-
Megan Crow, Je-Hyuk Lee, Hamsini Suresh, and Jesse Gillis
- Subjects
Regulation of gene expression ,Gene duplication ,Transcriptional regulation ,Gene family ,Computational biology ,Biology ,Phenotype ,Functional genomics ,Gene ,Genome - Abstract
What makes a mouse a mouse, and not a hamster? The answer lies in the genome, and more specifically, in differences in gene regulation between the two organisms: where and when each gene is expressed. To quantify differences, a typical study will either compare functional genomics data from homologous tissues, limiting the approach to closely related species; or compare gene repertoires, limiting the resolution of the analysis to gross correlations between phenotypes and gene family size. As an alternative, gene coexpression networks provide a basis for studying the evolution of gene regulation without these constraints. By incorporating data from hundreds of independent experiments, meta-analytic coexpression networks reflect the convergent output of species-specific transcriptional regulation.In this work, we develop a measure of regulatory evolution based on gene coexpression. Comparing data from 14 species, we quantify the conservation of coexpression patterns 1) as a function of evolutionary time, 2) across orthology prediction algorithms, and 3) with reference to cell- and tissue-specificity. Strikingly, we uncover deeply conserved patterns of gradient-like expression across cell types from both the animal and plant kingdoms. These results suggest that ancient genes contribute to transcriptional cell identity through mechanisms that are independent of duplication and divergence.
- Published
- 2020
44. Replicability of spatial gene expression atlas data from the adult mouse brain
- Author
-
Anthony M. Zador, Stephan Fischer, Cantin Ortiz, Konstantinos Meletis, Jesse Gillis, Daniel Fürth, and Shaina Lu
- Subjects
Correlation ,Lasso (statistics) ,Linear regression ,Supervised learning ,Gene expression ,Feature selection ,Computational biology ,Biology ,Data type ,Spatial analysis - Abstract
BackgroundSpatial gene expression is particularly interesting in the mammalian brain, with the potential to serve as a link between many data types. However, as with any type of expression data, cross-dataset benchmarking of spatial data is a crucial first step. Here, we assess the replicability, with reference to canonical brain sub-divisions, between the Allen Institute’s in situ hybridization data from the adult mouse brain (ABA) and a similar dataset collected using Spatial Transcriptomics (ST). With the advent of tractable spatial techniques, for the first time we are able to benchmark the Allen Institute’s whole-brain, whole-transcriptome spatial expression dataset with a second independent dataset that similarly spans the whole brain and transcriptome.ResultsWe use LASSO, linear regression, and correlation-based feature selection in a supervised learning framework to classify expression samples relative to their assayed location. We show that Allen reference atlas labels are classifiable using transcription, but that performance is higher in the ABA than ST. Further, models trained in one dataset and tested in the opposite dataset do not reproduce classification performance bi-directionally. Finally, while an identifying expression profile can be found for a given brain area, it does not generalize to the opposite dataset.ConclusionsIn general, we found that canonical brain area labels are classifiable in gene expression space within dataset and that our observed performance is not merely reflecting physical distance in the brain. However, we also show that cross-platform classification is not robust. Emerging spatial datasets from the mouse brain will allow further characterization of cross-dataset replicability.
- Published
- 2020
45. Cellular Anatomy of the Mouse Primary Motor Cortex
- Author
-
Xu An, Xiangning Li, Corey Elowsky, Laura Korobkova, Lei Gao, Giorgio A. Ascoli, Judith Mizrachi, William Galbavy, Jesse Gillis, Byung Kook Lim, Hanchuan Peng, Katherine Matho, Jason Palmer, Liya Ding, Uree Chon, Michael Hawrylycz, Florence D’Orazi, Kathleen Kelly, James C. Gee, Anastasiia Bludova, Peter A. Groblewski, Xu Li, Z. Josh Huang, Wayne Wakemen, Yun Wang, Stephanie Mok, Xiuli Kuang, X. William Yang, Houri Hintiryan, Michael S. Bienkowski, Karla E. Hirokawa, Lydia Ng, Peng Xie, Diek W. Wheeler, Xiaoyin Chen, Jin Yuan, Julie A. Harris, Yaoyao Li, Ramesh Palaniswamy, Yongsoo Kim, Li I. Zhang, Qingming Luo, Darrick Lo, Samik Bannerjee, Huizhong Tao, Ian Bowman, Meng Kuan Lin, Yu-Chi Sun, Quanxin Wang, Elise Shen, Lijuan Liu, Joel D. Hahn, Joshua T. Hatfield, Anan Li, Maitham Naeemi, Hui Gong, Xiaoli Qi, Partha P. Mitra, Hideki Kondo, Philip R. Nicovich, Pavel Osten, Hongkui Zeng, Muye Zhu, Sarojini M. Attili, Brian Zingg, Anthony M. Zador, Stephan Fischer, Bing-Xing Huo, Shenqin Yao, Nicholas N. Foster, Philip Lesnar, Rodrigo Muñoz-Castañeda, Ali Cetin, Young Gyun Park, Arun Narasimhan, Kwanghun Chuang, Lin Gou, Hong-Wei Dong, Yimin Wang, Chris Sin Park, Rhonda Drewes, and Jason Huang
- Subjects
Synapse ,Cell type ,Laminar organization ,Computer science ,Motor control ,Neuroinformatics ,Cellular Anatomy ,Primary motor cortex ,Projection neuron ,Neuroscience - Abstract
An essential step toward understanding brain function is to establish a cellular-resolution structural framework upon which multi-scale and multi-modal information spanning molecules, cells, circuits and systems can be integrated and interpreted. Here, through a collaborative effort from the Brain Initiative Cell Census Network (BICCN), we derive a comprehensive cell type-based description of one brain structure - the primary motor cortex upper limb area (MOp-ul) of the mouse. Applying state-of-the-art labeling, imaging, computational, and neuroinformatics tools, we delineated the MOp-ul within the Mouse Brain 3D Common Coordinate Framework (CCF). We defined over two dozen MOp-ul projection neuron (PN) types by their anterograde targets; the spatial distribution of their somata defines 11 cortical sublayers, a significant refinement of the classic notion of cortical laminar organization. We further combine multiple complementary tracing methods (classic tract tracing, cell type-based anterograde, retrograde, and transsynaptic viral tracing, high-throughput BARseq, and complete single cell reconstruction) to systematically chart cell type-based MOp input-output streams. As PNs link distant brain regions at synapses as well as host cellular gene expression, our construction of a PN type resolution MOp-ul wiring diagram will facilitate an integrated analysis of motor control circuitry across the molecular, cellular, and systems levels. This work further provides a roadmap towards a cellular resolution description of mammalian brain architecture.
- Published
- 2020
46. Intraductal Transplantation Models of Human Pancreatic Ductal Adenocarcinoma Reveal Progressive Transition of Molecular Subtypes
- Author
-
David A. Tuveson, Jesse Gillis, Hervé Tiriac, Dennis Plenker, Laura D. Wood, Nicholas J. Roberts, Brinda Alagesan, Chang-Il Hwang, Benno Traub, Young-Kyu Park, Jude Kendall, Pascal Belleau, Tim D.D. Somerville, Risa Karakida Kawaguchi, Siran Li, Lindsey A. Baker, Steven Gallinger, Koji Miyabayashi, Faiyaz Notta, Richard A. Burkhart, Ralph H. Hruban, Michael Wigler, Alexander Krasnitz, Astrid Deschênes, Giuseppina Caligiuri, Gun Ho Jang, and Christopher R. Vakoc
- Subjects
0301 basic medicine ,Pancreatic ductal adenocarcinoma ,Xenotransplantation ,medicine.medical_treatment ,Oncology and Carcinogenesis ,Adenocarcinoma ,Malignancy ,Gene dosage ,03 medical and health sciences ,Mice ,Pancreatic Cancer ,0302 clinical medicine ,Rare Diseases ,Clinical Research ,Pancreatic cancer ,medicine ,Animals ,Humans ,2.1 Biological and endogenous factors ,In patient ,Aetiology ,Cancer ,Neoplastic ,Transition (genetics) ,business.industry ,Animal ,Carcinoma ,Pancreatic Ducts ,medicine.disease ,Prognosis ,Transplantation ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,030104 developmental biology ,Oncology ,Gene Expression Regulation ,Pancreatic Ductal ,030220 oncology & carcinogenesis ,Disease Models ,Cancer research ,business ,Digestive Diseases ,Carcinoma, Pancreatic Ductal - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most lethal common malignancy, with little improvement in patient outcomes over the past decades. Recently, subtypes of pancreatic cancer with different prognoses have been elaborated; however, the inability to model these subtypes has precluded mechanistic investigation of their origins. Here, we present a xenotransplantation model of PDAC in which neoplasms originate from patient-derived organoids injected directly into murine pancreatic ducts. Our model enables distinction of the two main PDAC subtypes: intraepithelial neoplasms from this model progress in an indolent or invasive manner representing the classical or basal-like subtypes of PDAC, respectively. Parameters that influence PDAC subtype specification in this intraductal model include cell plasticity and hyperactivation of the RAS pathway. Finally, through intratumoral dissection and the direct manipulation of RAS gene dosage, we identify a suite of RAS-regulated secreted and membrane-bound proteins that may represent potential candidates for therapeutic intervention in patients with PDAC. Significance: Accurate modeling of the molecular subtypes of pancreatic cancer is crucial to facilitate the generation of effective therapies. We report the development of an intraductal organoid transplantation model of pancreatic cancer that models the progressive switching of subtypes, and identify stochastic and RAS-driven mechanisms that determine subtype specification. See related commentary by Pickering and Morton, p. 1448. This article is highlighted in the In This Issue feature, p. 1426
- Published
- 2020
47. Single-cell RNA sequencing of developing maize ears facilitates functional analysis and trait candidate gene discovery
- Author
-
Zefu Lu, Brian R. Rice, Jorg Drenkow, Jesse Gillis, David A. Jackson, Bing Yang, Anding Luo, Anne W. Sylvester, Xiaosa Xu, Forrest Li, Lei Liu, Thomas R. Gingeras, Liya Wang, Megan Crow, Doreen Ware, Benjamin Harris, Xiaofei Wang, Robert J. Schmitz, Edgar Demesa-Arevalo, Nathan Fox, Alexander E. Lipka, and Si Nian Char
- Subjects
Candidate gene ,Quantitative Trait Loci ,Gene regulatory network ,Genome-wide association study ,Computational biology ,Biology ,Zea mays ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Pleiotropy ,Gene Expression Regulation, Plant ,Gene Regulatory Networks ,Molecular Biology ,Genetic Association Studies ,030304 developmental biology ,0303 health sciences ,Base Sequence ,Sequence Analysis, RNA ,Protoplasts ,RNA ,Gene Expression Regulation, Developmental ,Reproducibility of Results ,Cell Biology ,Cell sorting ,Single cell sequencing ,Genetic redundancy ,Single-Cell Analysis ,Transcriptome ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Crop productivity depends on activity of meristems that produce optimized plant architectures, including that of the maize ear. A comprehensive understanding of development requires insight into the full diversity of cell types and developmental domains and the gene networks required to specify them. Until now, these were identified primarily by morphology and insights from classical genetics, which are limited by genetic redundancy and pleiotropy. Here, we investigated the transcriptional profiles of 12,525 single cells from developing maize ears. The resulting developmental atlas provides a single-cell RNA sequencing (scRNA-seq) map of an inflorescence. We validated our results by mRNA in situ hybridization and by fluorescence-activated cell sorting (FACS) RNA-seq, and we show how these data may facilitate genetic studies by predicting genetic redundancy, integrating transcriptional networks, and identifying candidate genes associated with crop yield traits.
- Published
- 2020
48. Integrating barcoded neuroanatomy with spatial transcriptional profiling reveals cadherin correlates of projections shared across the cortex
- Author
-
Yu-Chi Sun, Xiaoyin Chen, Anthony M. Zador, Stephan Fischer, Shaina Lu, and Jesse Gillis
- Subjects
Transcriptome ,medicine.anatomical_structure ,Cellular resolution ,Neuronal circuits ,Cadherin ,Gene expression ,medicine ,Biology ,Auditory cortex ,Neuroscience ,Motor cortex ,Neuroanatomy - Abstract
Functional circuits consist of neurons with diverse axonal projections and gene expression. Understanding the molecular signature of projections requires high-throughput interrogation of both gene expression and projections to multiple targets in the same cells at cellular resolution, which is difficult to achieve using current technology. Here, we introduce BARseq2, a technique that simultaneously maps projections and detects multiplexed gene expression by in situ sequencing. We determined the expression of cadherins and cell-type markers in 29,933 cells, and the projections of 3,164 cells in both the mouse motor cortex and auditory cortex. Associating gene expression and projections in 1,349 neurons revealed shared cadherin signatures of homologous projections across the two cortical areas. These cadherins were enriched across multiple branches of the transcriptomic taxonomy. By correlating multi-gene expression and projections to many targets in single neurons with high throughput, BARseq2 provides a path to uncovering the molecular logic underlying neuronal circuits.
- Published
- 2020
49. CoCoCoNet: Conserved and Comparative Co-expression Across a Diverse Set of Species
- Author
-
Manthan Shah, Jesse Gillis, John Lee, Sara Ballouz, and Megan Crow
- Subjects
Saccharomyces cerevisiae Proteins ,Autism Spectrum Disorder ,AcademicSubjects/SCI00010 ,ved/biology.organism_classification_rank.species ,Gene Expression ,Computational biology ,Set (abstract data type) ,03 medical and health sciences ,0302 clinical medicine ,Gene Modules ,Arabidopsis ,Genetics ,Animals ,Humans ,Gene Regulatory Networks ,RNA-Seq ,Functional group (ecology) ,Model organism ,Gene ,Zebrafish ,030304 developmental biology ,0303 health sciences ,biology ,ved/biology ,biology.organism_classification ,Expression (mathematics) ,Web Server Issue ,Software ,030217 neurology & neurosurgery ,Function (biology) - Abstract
Co-expression analysis has provided insight into gene function in organisms from Arabidopsis to Zebrafish. Comparison across species has the potential to enrich these results, for example by prioritizing among candidate human disease genes based on their network properties, or by finding alternative model systems where their co-expression is conserved. Here, we present CoCoCoNet as a tool for identifyingconserved gene modules andcomparingco-expressionnetworks. CoCoCoNet is a resource for both data and methods, providing gold-standard networks and sophisticated tools for on-the-fly comparative analyses across 14 species. We show how CoCoCoNet can be used in two use cases. In the first, we demonstrate deep conservation of a nucleolus gene module across very divergent organisms, and in the second, we show how the heterogeneity of autism mechanisms in humans can be broken down by functional groups, and translated to model organisms. CoCoCoNet is free to use and available to all athttps://milton.cshl.edu/CoCoCoNet, with data and R scripts available atftp://milton.cshl.edu/data.
- Published
- 2020
50. Multiscale Co-Expression in the Brain
- Author
-
Jesse Gillis, Stephan Fischer, Megan Crow, and Benjamin Harris
- Subjects
0303 health sciences ,Cell type ,business.industry ,0206 medical engineering ,Cell ,Confounding ,Cell type specific ,02 engineering and technology ,Computational biology ,Biology ,Expression (mathematics) ,03 medical and health sciences ,Text mining ,medicine.anatomical_structure ,medicine ,Primary motor cortex ,business ,Gene ,020602 bioinformatics ,030304 developmental biology - Abstract
Single-cell RNA-sequencing (scRNAseq) data can reveal co-regulatory relationships between genes that may be hidden in bulk RNAseq due to cell type confounding. Using the primary motor cortex data from the Brain Initiative Cell Census Network (BICCN), we study cell type specific co-expression across 500,000 cells. Surprisingly, we find that the same gene-gene relationships that differentiate cell types are evident at finer and broader scales, suggesting a consistent multiscale regulatory landscape.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.