skip to main content


Title: Inferring latent temporal progression and regulatory networks from cross-sectional transcriptomic data of cancer samples
Unraveling molecular regulatory networks underlying disease progression is critically important for understanding disease mechanisms and identifying drug targets. The existing methods for inferring gene regulatory networks (GRNs) rely mainly on time-course gene expression data. However, most available omics data from cross-sectional studies of cancer patients often lack sufficient temporal information, leading to a key challenge for GRN inference. Through quantifying the latent progression using random walks-based manifold distance, we propose a latent-temporal progression-based Bayesian method, PROB, for inferring GRNs from the cross-sectional transcriptomic data of tumor samples. The robustness of PROB to the measurement variabilities in the data is mathematically proved and numerically verified. Performance evaluation on real data indicates that PROB outperforms other methods in both pseudotime inference and GRN inference. Applications to bladder cancer and breast cancer demonstrate that our method is effective to identify key regulators of cancer progression or drug targets. The identified ACSS1 is experimentally validated to promote epithelial-to-mesenchymal transition of bladder cancer cells, and the predicted FOXM1-targets interactions are verified and are predictive of relapse in breast cancer. Our study suggests new effective ways to clinical transcriptomic data modeling for characterizing cancer progression and facilitates the translation of regulatory network-based approaches into precision medicine.  more » « less
Award ID(s):
1763272
NSF-PAR ID:
10222819
Author(s) / Creator(s):
; ;
Editor(s):
Roy, Sushmita
Date Published:
Journal Name:
PLOS Computational Biology
Volume:
17
Issue:
3
ISSN:
1553-7358
Page Range / eLocation ID:
e1008379
Format(s):
Medium: X
Sponsoring Org:
National Science Foundation
More Like this
  1. Inferring gene regulatory networks (GRNs) from single-cell gene expression datasets is a challenging task. Existing methods are often designed heuristically for specific datasets and lack the flexibility to incorporate additional information or compare against other algorithms. Further, current GRN inference methods do not provide uncertainty estimates with respect to the interactions that they predict, making inferred networks challenging to interpret. To overcome these challenges, we introduce Probabilistic Matrix Factorization for Gene Regulatory Network inference (PMF-GRN). PMF-GRN uses single-cell gene expression data to learn latent factors representing transcription factor activity as well as regulatory relationships between transcription factors and their target genes. This approach incorporates available experimental evidence into prior distributions over latent factors and scales well to single-cell gene expression datasets. By utilizing variational inference, we facilitate hyperparameter search for principled model selection and direct comparison to other generative models. To assess the accuracy of our method, we evaluate PMF-GRN using the model organisms Saccharomyces cerevisiae and Bacillus subtilis, benchmarking against database-derived gold standard interactions. We discover that, on average, PMF-GRN infers GRNs more accurately than current state-of-the-art single-cell GRN inference methods. Moreover, our PMF-GRN approach offers well-calibrated uncertainty estimates, as it performs gene regulatory network (GRN) inference in a probabilistic setting. These estimates are valuable for validation purposes, particularly when validated interactions are limited or a gold standard is incomplete. 
    more » « less
  2. Abstract

    Inferring gene regulatory networks (GRNs) from single-cell data is challenging due to heuristic limitations. Existing methods also lack estimates of uncertainty. Here we present Probabilistic Matrix Factorization for Gene Regulatory Network Inference (PMF-GRN). Using single-cell expression data, PMF-GRN infers latent factors capturing transcription factor activity and regulatory relationships. Using variational inference allows hyperparameter search for principled model selection and direct comparison to other generative models. We extensively test and benchmark our method using real single-cell datasets and synthetic data. We show that PMF-GRN infers GRNs more accurately than current state-of-the-art single-cell GRN inference methods, offering well-calibrated uncertainty estimates.

     
    more » « less
  3. Newman, Stuart A (Ed.)
    Cellular differentiation during hematopoiesis is guided by gene regulatory networks (GRNs) comprising transcription factors (TFs) and the effectors of cytokine signaling. Based largely on analyses conducted at steady state, these GRNs are thought to be organized as a hierarchy of bistable switches, with antagonism between Gata1 and PU.1 driving red- and white-blood cell differentiation. Here, we utilize transient gene expression patterns to infer the genetic architecture—the type and strength of regulatory interconnections—and dynamics of a twelve-gene GRN including key TFs and cytokine receptors. We trained gene circuits, dynamical models that learn genetic architecture, on high temporal-resolution gene-expression data from the differentiation of an inducible cell line into erythrocytes and neutrophils. The model is able to predict the consequences of gene knockout, knockdown, and overexpression experiments and the inferred interconnections are largely consistent with prior empirical evidence. The inferred genetic architecture is densely interconnected rather than hierarchical, featuring extensive cross-antagonism between genes from alternative lineages and positive feedback from cytokine receptors. The analysis of the dynamics of gene regulation in the model reveals that PU.1 is one of the last genes to be upregulated in neutrophil conditions and that the upregulation of PU.1 and other neutrophil genes is driven by Cebpa and Gfi1 instead. This model inference is confirmed in an independent single-cell RNA-Seq dataset from mouse bone marrow in which Cebpa and Gfi1 expression precedes the neutrophil-specific upregulation of PU.1 during differentiation. These results demonstrate that full PU.1 upregulation during neutrophil development involves regulatory influences extrinsic to the Gata1-PU.1 bistable switch. Furthermore, although there is extensive cross-antagonism between erythroid and neutrophil genes, it does not have a hierarchical structure. More generally, we show that the combination of high-resolution time series data and data-driven dynamical modeling can uncover the dynamics and causality of developmental events that might otherwise be obscured. 
    more » « less
  4. Kuijjer, Marieke (Ed.)
    Abstract Motivation Biological processes are regulated by underlying genes and their interactions that form gene regulatory networks (GRNs). Dysregulation of these GRNs can cause complex diseases such as cancer, Alzheimer’s and diabetes. Hence, accurate GRN inference is critical for elucidating gene function, allowing for the faster identification and prioritization of candidate genes for functional investigation. Several statistical and machine learning-based methods have been developed to infer GRNs based on biological and synthetic datasets. Here, we developed a method named AGRN that infers GRNs by employing an ensemble of machine learning algorithms. Results From the idea that a single method may not perform well on all datasets, we calculate the gene importance scores using three machine learning methods—random forest, extra tree and support vector regressors. We calculate the importance scores from Shapley Additive Explanations, a recently published method to explain machine learning models. We have found that the importance scores from Shapley values perform better than the traditional importance scoring methods based on almost all the benchmark datasets. We have analyzed the performance of AGRN using the datasets from the DREAM4 and DREAM5 challenges for GRN inference. The proposed method, AGRN—an ensemble machine learning method with Shapley values, outperforms the existing methods both in the DREAM4 and DREAM5 datasets. With improved accuracy, we believe that AGRN inferred GRNs would enhance our mechanistic understanding of biological processes in health and disease. Availabilityand implementation https://github.com/DuaaAlawad/AGRN. Supplementary information Supplementary data are available at Bioinformatics online. 
    more » « less
  5. Inferring gene regulatory networks (GRNs) from single-cell RNA-seq (scRNA-seq) data is an important computational question to find regulatory mechanisms involved in fundamental cellular processes. Although many computational methods have been designed to predict GRNs from scRNA-seq data, they usually have high false positive rates and none infer GRNs by directly using the paired datasets of case-versus-control experiments. Here we present a novel deep-learning-based method, named scTIGER, for GRN detection by using the co-differential relationships of gene expression profiles in paired scRNA-seq datasets. scTIGER employs cell-type-based pseudotiming, an attention-based convolutional neural network method and permutation-based significance testing for inferring GRNs among gene modules. As state-of-the-art applications, we first applied scTIGER to scRNA-seq datasets of prostate cancer cells, and successfully identified the dynamic regulatory networks of AR, ERG, PTEN and ATF3 for same-cell type between prostatic cancerous and normal conditions, and two-cell types within the prostatic cancerous environment. We then applied scTIGER to scRNA-seq data from neurons with and without fear memory and detected specific regulatory networks for BDNF, CREB1 and MAPK4. Additionally, scTIGER demonstrates robustness against high levels of dropout noise in scRNA-seq data.

     
    more » « less