scholarly journals Decision letter: Gene regulatory network reconstruction using single-cell RNA sequencing of barcoded genotypes in diverse environments

2019 ◽  
2019 ◽  
Author(s):  
Christopher A Jackson ◽  
Dayanne M Castro ◽  
Giuseppe-Antonio Saldi ◽  
Richard Bonneau ◽  
David Gresham

AbstractUnderstanding how gene expression programs are controlled requires identifying regulatory relationships between transcription factors and target genes. Gene regulatory networks are typically constructed from gene expression data acquired following genetic perturbation or environmental stimulus. Single-cell RNA sequencing (scRNAseq) captures the gene expression state of thousands of individual cells in a single experiment, offering advantages in combinatorial experimental design, large numbers of independent measurements, and accessing the interaction between the cell cycle and environmental responses that is hidden by population-level analysis of gene expression. To leverage these advantages, we developed a method for transcriptionally barcoding gene deletion mutants and performing scRNAseq in budding yeast (Saccharomyces cerevisiae). We pooled diverse genotypes in 11 different environmental conditions and determined their expression state by sequencing 38,285 individual cells. We developed, and benchmarked, a framework for learning gene regulatory networks from scRNAseq data that incorporates multitask learning and constructed a global gene regulatory network comprising 12,018 interactions. Our study establishes a general approach to gene regulatory network reconstruction from scRNAseq data that can be employed in any organism.


eLife ◽  
2020 ◽  
Vol 9 ◽  
Author(s):  
Christopher A Jackson ◽  
Dayanne M Castro ◽  
Giuseppe-Antonio Saldi ◽  
Richard Bonneau ◽  
David Gresham

Understanding how gene expression programs are controlled requires identifying regulatory relationships between transcription factors and target genes. Gene regulatory networks are typically constructed from gene expression data acquired following genetic perturbation or environmental stimulus. Single-cell RNA sequencing (scRNAseq) captures the gene expression state of thousands of individual cells in a single experiment, offering advantages in combinatorial experimental design, large numbers of independent measurements, and accessing the interaction between the cell cycle and environmental responses that is hidden by population-level analysis of gene expression. To leverage these advantages, we developed a method for scRNAseq in budding yeast (Saccharomyces cerevisiae). We pooled diverse transcriptionally barcoded gene deletion mutants in 11 different environmental conditions and determined their expression state by sequencing 38,285 individual cells. We benchmarked a framework for learning gene regulatory networks from scRNAseq data that incorporates multitask learning and constructed a global gene regulatory network comprising 12,228 interactions.


Development ◽  
2020 ◽  
Vol 147 (17) ◽  
pp. dev191528 ◽  
Author(s):  
Stephany Foster ◽  
Nathalie Oulhen ◽  
Gary Wessel

ABSTRACTIdentifying cell states during development from their mRNA profiles provides insight into their gene regulatory network. Here, we leverage the sea urchin embryo for its well-established gene regulatory network to interrogate the embryo using single cell RNA sequencing. We tested eight developmental stages in Strongylocentrotus purpuratus, from the eight-cell stage to late in gastrulation. We used these datasets to parse out 22 major cell states of the embryo, focusing on key transition stages for cell type specification of each germ layer. Subclustering of these major embryonic domains revealed over 50 cell states with distinct transcript profiles. Furthermore, we identified the transcript profile of two cell states expressing germ cell factors, one we conclude represents the primordial germ cells and the other state is transiently present during gastrulation. We hypothesize that these cells of the Veg2 tier of the early embryo represent a lineage that converts to the germ line when the primordial germ cells are deleted. This broad resource will hopefully enable the community to identify other cell states and genes of interest to expose the underpinning of developmental mechanisms.


2021 ◽  
Author(s):  
Matthew Stone ◽  
Sunnie Grace McCalla ◽  
Alireza Fotuhi Siahpirani ◽  
Viswesh Periyasamy ◽  
Junha Shin ◽  
...  

Single-cell RNA-sequencing (scRNA-seq) offers unparalleled insight into the transcriptional pro- grams of different cellular states by measuring the transcriptome of thousands individual cells. An emerging problem in the analysis of scRNA-seq is the inference of transcriptional gene regulatory net- works and a number of methods with different learning frameworks have been developed. Here we present a expanded benchmarking study of eleven recent network inference methods on six published single-cell RNA-sequencing datasets in human, mouse, and yeast considering different types of gold standard networks and evaluation metrics. We evaluate methods based on their computing requirements as well as on their ability to recover the network structure. We find that while no method is a universal winner and most methods have a modest recovery of experimentally derived interactions based on global metrics such as AUPR, methods are able to capture targets of regulators that are relevant to the system under study. Based on overall performance we grouped the methods into three main categories and found a combination of information-theoretic and regression-based methods to have a generally high perfor- mance. We also evaluate the utility of imputation for gene regulatory network inference and find that a small number of methods benefit from imputation, which further depends upon the dataset. Finally, comparisons to inferred networks for comparable bulk conditions showed that networks inferred from scRNA-seq datasets are often better or at par to those from bulk suggesting that scRNA-seq datasets can be a cost-effective way for gene regulatory network inference. Our analysis should be beneficial in selecting algorithms for performing network inference but also argues for improved methods and better gold standards for accurate assessment of regulatory network inference methods for mammalian systems.


2021 ◽  
Author(s):  
Roozbeh Manshaei

Understanding and interpreting the inherently uncertain nature of complex biological systems, as well as the time to an event in these systems, are notable challenges in the field of bioinformatics. Overcoming these challenges could potentially lead to scientific discoveries, for example paving the path for the design of new drugs to target specific diseases such as cancer, or helping to apply more effective treatment for these diseases. In general, reverse engineering of these types of biological systems using online datasets is difficult. In particular, finding a unique solution to these systems is hard due to their complexity and the small sample size of datasets. This remains an unsolved problem due to such uncertainty, and the often intractable solution space of these systems. The term"uncertainty" describes the application-based margin of significance, validity, and efficiency of inferred or predictive models in their ability to extract characteristic properties and features describing the observed state of a given biological system. In this work, uncertainties within two specific bioinformatics domains are considered, namely "gene regulatory network reconstruction" (in which gene interactions/relationships within a biological entity are inferred from gene expression data); and "cancer survivorship prediction" (in which patient survival rates are predicted based on clinical factors and treatment outcomes). One approach to reduce uncertainty is to apply different constraints that have particular relevance to each application domain. In gene network reconstruction for instance, the consideration of constraints such as sparsity, stability and modularity, can inform and reduce uncertainty in the inferred reconstructions. While in cancer survival prediction, there is uncertainty in determining which clinical features (or feature aggregates) can improve associated prediction models. The inherent lack of understanding of how, why and when such constraints should be applied, however, prompts the need for a radically new approach. In this dissertation, a new approach is thus considered to aid human expert users in understanding and exploring inherent uncertainties associated with these two bioinformatics domains. Specifically, a novel set of tools is introduced and developed to assist in evidence gathering, constraint definition, and refinement of models toward the discovery of better solutions. This dissertation employs computational approaches, including convex optimization and feature selection/aggregation, in order to increase the chances of finding a unique solution. These approaches are realized through three novel interactive tools that employ tangible interaction in combination with graphical visualization to enable experts to query and manipulate the data. Tangible interaction provides physical embodiments of data and computational functions in support of learning and collaboration. Using these approaches, the dissertation demonstrates: (1) a modified stability constraint for reconstructing gene regulatory network that shows improvement in accuracy of predicted networks, (2) a novel modularity constraint (neighbor norm) for extracting available structures in the data which is validated with Laplacian eigenvalue spectrum, and (3) a hybrid method for estimating overall survival and inferring effective prognosis factors for patients with advanced prostate cancer that improves the accuracy of survival analysis.


2019 ◽  
Vol 17 (06) ◽  
pp. 1950035
Author(s):  
Huiqing Wang ◽  
Yuanyuan Lian ◽  
Chun Li ◽  
Yue Ma ◽  
Zhiliang Yan ◽  
...  

As a tool of interpreting and analyzing genetic data, gene regulatory network (GRN) could reveal regulatory relationships between genes, proteins, and small molecules, as well as understand physiological activities and functions within biological cells, interact in pathways, and how to make changes in the organism. Traditional GRN research focuses on the analysis of the regulatory relationships through the average of cellular gene expressions. These methods are difficult to identify the cell heterogeneity of gene expression. Existing methods for inferring GRN using single-cell transcriptional data lack expression information when genes reach steady state, and the high dimensionality of single-cell data leads to high temporal and spatial complexity of the algorithm. In order to solve the problem in traditional GRN inference methods, including the lack of cellular heterogeneity information, single-cell data complexity and lack of steady-state information, we propose a method for GRN inference using single-cell transcription and gene knockout data, called SINgle-cell transcription data-KNOckout data (SIN-KNO), which focuses on combining dynamic and steady-state information of regulatory relationship contained in gene expression. Capturing cell heterogeneity information could help understand the gene expression difference in different cells. So, we could observe gene expression changes more accurately. Gene knockout data could observe the gene expression levels at steady-state of all other genes when one gene is knockout. Classifying the genes before analyzing the single-cell data could determine a large number of non-existent regulation, greatly reducing the number of regulation required for inference. In order to show the efficiency, the proposed method has been compared with several typical methods in this area including GENIE3, JUMP3, and SINCERITIES. The results of the evaluation indicate that the proposed method can analyze the diversified information contained in the two types of data, establish a more accurate gene regulation network, and improve the computational efficiency. The method provides a new thinking for dealing with large datasets and high computational complexity of single-cell data in the GRN inference.


Sign in / Sign up

Export Citation Format

Share Document