scholarly journals scPADGRN: A preconditioned ADMM approach for reconstructing dynamic gene regulatory network using single-cell RNA sequencing data

2019 ◽  
Author(s):  
Xiao Zheng ◽  
Yuan Huang ◽  
Xiufen Zou

AbstractDisease development and cell differentiation both involve dynamic changes; therefore, the reconstruction of dynamic gene regulatory networks (DGRNs) is an important but difficult problem in systems biology. With recent technical advances in single-cell RNA sequencing (scRNA-seq), large volumes of scRNA-seq data are being obtained for various processes. However, most current methods of inferring DGRNs from bulk samples may not be suitable for scRNA-seq data. In this work, we present scPADGRN, a novel DGRN inference method using time-series scRNA-seq data. scPADGRN combines the preconditioned alternating direction method of multipliers with cell clustering for DGRN reconstruction. It exhibits advantages in accuracy, robustness and fast convergence. Moreover, a quantitative index called Differentiation Genes’ Interaction Enrichment (DGIE) is presented to quantify the interaction enrichment of genes related to differentiation. From the DGIE scores of relevant subnetworks, we infer that the functions of embryonic stem (ES) cells are most active initially and may gradually fade over time. The communication strength of known contributing genes that facilitate cell differentiation increases from ES cells to terminally differentiated cells. We also identify several genes responsible for the changes in the DGIE scores occurring during cell differentiation based on three real single-cell datasets. Our results demonstrate that single-cell analyses based on network inference coupled with quantitative computations can reveal key transcriptional regulators involved in cell differentiation and disease development.Author summarySingle-cell RNA sequencing (scRNA-seq) data are gaining popularity for providing access to cell-level measurements. Currently, time-series scRNA-seq data allow researchers to study dynamic changes during biological processes. This work proposes a novel method, scPADGRN, for application to time-series scRNA-seq data to construct dynamic gene regulatory networks, which are informative for investigating dynamic changes during disease development and cell differentiation. The proposed method shows satisfactory performance on both simulated data and three real datasets concerning cell differentiation. To quantify network dynamics, we present a quantitative index, DGIE, to measure the degree of activity of a certain set of genes in a regulatory network. Quantitative computations based on dynamic networks identify key regulators in cell differentiation and reveal the activity states of the identified regulators. Specifically, Bhlhe40, Msx2, Foxa2 and Dnmt3l might be important regulatory genes involved in differentiation from mouse ES cells to primitive endoderm (PrE) cells. For differentiation from mouse embryonic fibroblast cells to myocytes, Scx, Fos and Tcf12 are suggested to be key regulators. Sox5, Meis2, Hoxb3, Tcf7l1 and Plagl1 critically contribute during differentiation from human ES cells to definitive endoderm cells. These results may guide further theoretical and experimental efforts to understand cell differentiation processes and explore cell heterogeneity.

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.


Author(s):  
Rui-Qi Wang ◽  
Wei Zhao ◽  
Hai-Kui Yang ◽  
Jia-Mei Dong ◽  
Wei-Jie Lin ◽  
...  

Colorectal cancer (CRC) manifests as gastrointestinal tumors with high intratumoral heterogeneity. Recent studies have demonstrated that CRC may consist of tumor cells with different consensus molecular subtypes (CMS). The advancements in single-cell RNA sequencing have facilitated the development of gene regulatory networks to decode key regulators for specific cell types. Herein, we comprehensively analyzed the CMS of CRC patients by using single-cell RNA-sequencing data. CMS for all malignant cells were assigned using CMScaller. Gene set variation analysis showed pathway activity differences consistent with those reported in previous studies. Cell–cell communication analysis confirmed that CMS1 was more closely related to immune cells, and that monocytes and macrophages play dominant roles in the CRC tumor microenvironment. On the basis of the constructed gene regulation networks (GRNs) for each subtype, we identified that the critical transcription factor ERG is universally activated and upregulated in all CMS in comparison with normal cells, and that it performed diverse roles by regulating the expression of different downstream genes. In summary, molecular subtyping of single-cell RNA-sequencing data for colorectal cancer could elucidate the heterogeneity in gene regulatory networks and identify critical regulators of CRC.


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.


2021 ◽  
Author(s):  
Boris M. Brenerman ◽  
Benjamin D. Shapiro ◽  
Michael C. Schatz ◽  
Alexis Battle

AbstractSingle-cell RNA sequencing data contain patterns of correlation that are poorly captured by techniques that rely on linear estimation or assumptions of Gaussian behavior. We apply random forest regression to scRNAseq data from mouse brains, which identifies the co-regulation of genes within specific cellular contexts. By analyzing the estimators of the random forest, we identify several novel candidate gene regulatory networks and compare these networks in aged and young mice. We demonstrate that cell populations have cell-type specific phenotypes of aging that are not detected by other methods, including the collapse of differentiating oligodendrocytes but not precursors or mature oligodendrocytes.


2020 ◽  
Author(s):  
Harsh Shrivastava ◽  
Xiuwei Zhang ◽  
Srinivas Aluru ◽  
Le Song

AbstractMotivationGene regulatory networks (GRNs) are graphs that specify the interactions between transcription factors (TFs) and their target genes. Understanding these interactions is crucial for studying the mechanisms in cell differentiation, growth and development. Computational methods are needed to infer these networks from measured data. Although the availability of single cell RNA-Sequencing (scRNA-Seq) data provides unprecedented scale and resolution of gene-expression data, the inference of GRNs remains a challenge, mainly due to the complexity of the regulatory relationships and the noise in the data.ResultsWe propose GRNUlar, a novel deep learning architecture based on the unrolled algorithms idea for GRN inference from scRNA-Seq data. Like some existing methods which use prior information of which genes are TFs, GRNUlar also incorporates this TF information using a sparse multi-task deep learning architecture. We also demonstrate the application of a recently developed unrolled architecture GLAD to recover undirected GRNs in the absence of TF information. These unrolled architectures require supervision to train, for which we leverage the existing synthetic data simulators which generate scRNA-Seq data guided by a GRN. We show that unrolled algorithms outperform the state-of-the-art methods on synthetic data as well as real datasets in both the settings of TF information being absent or available.AvailabilityGithub link to GRNUlar - https://github.com/Harshs27/[email protected]


BMC Genomics ◽  
2020 ◽  
Vol 21 (S11) ◽  
Author(s):  
Shouguo Gao ◽  
Zhijie Wu ◽  
Xingmin Feng ◽  
Sachiko Kajigaya ◽  
Xujing Wang ◽  
...  

Abstract Background Presently, there is no comprehensive analysis of the transcription regulation network in hematopoiesis. Comparison of networks arising from gene co-expression across species can facilitate an understanding of the conservation of functional gene modules in hematopoiesis. Results We used single-cell RNA sequencing to profile bone marrow from human and mouse, and inferred transcription regulatory networks in each species in order to characterize transcriptional programs governing hematopoietic stem cell differentiation. We designed an algorithm for network reconstruction to conduct comparative transcriptomic analysis of hematopoietic gene co-expression and transcription regulation in human and mouse bone marrow cells. Co-expression network connectivity of hematopoiesis-related genes was found to be well conserved between mouse and human. The co-expression network showed “small-world” and “scale-free” architecture. The gene regulatory network formed a hierarchical structure, and hematopoiesis transcription factors localized to the hierarchy’s middle level. Conclusions Transcriptional regulatory networks are well conserved between human and mouse. The hierarchical organization of transcription factors may provide insights into hematopoietic cell lineage commitment, and to signal processing, cell survival and disease initiation.


eLife ◽  
2021 ◽  
Vol 10 ◽  
Author(s):  
Periklis Paganos ◽  
Danila Voronov ◽  
Jacob M Musser ◽  
Detlev Arendt ◽  
Maria Ina Arnone

Identifying the molecular fingerprint of organismal cell types is key for understanding their function and evolution. Here, we use single cell RNA sequencing (scRNA-seq) to survey the cell types of the sea urchin early pluteus larva, representing an important developmental transition from non-feeding to feeding larva. We identify 21 distinct cell clusters, representing cells of the digestive, skeletal, immune, and nervous systems. Further subclustering of these reveal a highly detailed portrait of cell diversity across the larva, including the identification of neuronal cell types. We then validate important gene regulatory networks driving sea urchin development and reveal new domains of activity within the larval body. Focusing on neurons that co-express Pdx-1 and Brn1/2/4, we identify an unprecedented number of genes shared by this population of neurons in sea urchin and vertebrate endocrine pancreatic cells. Using differential expression results from Pdx-1 knockdown experiments, we show that Pdx1 is necessary for the acquisition of the neuronal identity of these cells. We hypothesize that a network similar to the one orchestrated by Pdx1 in the sea urchin neurons was active in an ancestral cell type and then inherited by neuronal and pancreatic developmental lineages in sea urchins and vertebrates.


Sign in / Sign up

Export Citation Format

Share Document