The integration of single-cell multi-omics technologies is revolutionizing our ability to infer gene regulatory networks (GRNs) with unprecedented resolution.
The integration of single-cell multi-omics technologies is revolutionizing our ability to infer gene regulatory networks (GRNs) with unprecedented resolution. This article provides a comprehensive guide for researchers and drug development professionals, exploring the foundational principles of GRNs in evolutionary developmental biology. It details state-of-the-art computational methodsâfrom foundational models like scGPT to innovative tools like LINGER and cRegulonâfor inferring regulatory interactions from paired transcriptomic and epigenomic data. The content further addresses critical troubleshooting strategies for data sparsity and model interpretability, outlines rigorous validation frameworks against resources like ChIP-seq and eQTLs, and synthesizes how these advances are illuminating the regulatory underpinnings of cell fate decisions, disease mechanisms, and therapeutic target discovery.
A Gene Regulatory Network (GRN) is a collection of molecular regulators that interact with each other and with other substances in the cell to govern the gene expression levels of mRNA and proteins [1]. These networks represent the complex regulatory relationships that control cellular functions, development, and responses to environmental stimuli. GRNs provide a systems-level explanation of biological processes, enabling researchers to understand how genetic information flows through biological systems to determine cell identity, function, and behavior [1] [2]. In the context of single-cell multi-omics research, GRN inference has become crucial for understanding cellular heterogeneity and the mechanisms underlying development and disease [3] [4].
GRNs are mathematically represented as bipartite networks where nodes represent biological entities (genes and their regulators), and edges represent the regulatory interactions between them [1]. Molecular regulators include transcription factors (TFs), RNA-binding proteins, and non-coding RNAs that participate in the control of gene expression. The directionality of these networks is fundamental, as TFs regulate target genes and typically not vice versa [2]. The study of GRNs has been revolutionized by single-cell technologies, which enable the analysis of regulatory relationships at unprecedented resolution across diverse cell types and states [3] [5].
Nodes in GRNs represent distinct biological entities involved in gene regulation. The primary node types include:
Each node possesses specific properties, including expression levels, activation states, and spatial-temporal localization patterns that influence its regulatory potential.
Edges represent functional relationships between nodes and are characterized by:
GRN edges can represent different interaction types:
A regulon is a group of genes regulated as a unit, generally controlled by the same regulatory gene that expresses a protein acting as a repressor or activator [6]. In eukaryotes, this term refers to any group of non-contiguous genes controlled by the same regulatory gene [6]. The concept of core and extended regulons has emerged through comparative genomics, where core regulons contain functions directly related to the primary regulatory signal, while extended regulons reflect specific adaptations to environmental niches [7].
cRegulons (cellular regulons) represent the set of regulatory relationships active in specific cellular contexts, often inferred through computational approaches like SCENIC (Single-Cell Regulatory Network Inference and Clustering) [8]. These context-specific regulatory units enable the identification of transcription factors driving particular cell states in development and disease.
Table 1: Key Regulatory Units in Gene Regulatory Networks
| Regulatory Unit | Definition | Characteristics | Biological Significance |
|---|---|---|---|
| Regulon | Group of genes regulated by a common transcription factor | Genes may be non-contiguous; coordinated expression response | Enables coordinated cellular responses to stimuli [6] |
| Core Regulon | Subset of regulon conserved across species | Functions directly related to TF-inducing signal | Maintains essential, evolutionarily conserved functions [7] |
| Extended Regulon | Condition-specific portion of regulon | Contains adaptive functions for specific niches | Facilitates rapid adaptation to environmental changes [7] |
| cRegulon | Context-specific regulon activity | Inferred from single-cell data using tools like SCENIC | Identifies drivers of cell identity and state transitions [8] |
| Modulon | Set of regulons collectively regulated | Responds to overall conditions or stresses | Coordinates broader cellular programs beyond single TF control [6] |
GRN inference methods have evolved from correlation-based approaches to sophisticated machine learning models that integrate multiple data types:
Co-expression-based methods such as WGCNA, ARACNe, and GENIE3 infer TF-TG relationships from gene expression data by capturing covariation patterns [1] [3]. While useful, these methods typically produce undirected edges and cannot distinguish causal relationships from correlations [1] [3].
Integrated methods combine multiple data types to improve inference accuracy. For example, PANDA integrates protein-protein interactions, gene expression, and transcription factor binding motifs to reconstruct condition-specific regulatory networks [1]. MONSTER estimates phenotypic-specific regulatory networks using gene expression and TF binding motif data, then computes TF involvement in state transitions [1].
Single-cell multi-omics methods represent the cutting edge in GRN inference. LINGER uses lifelong neural networks to infer GRNs from single-cell multiome data, incorporating atlas-scale external bulk data and TF motif knowledge as manifold regularization [3]. PRISM-GRN employs a Bayesian model that incorporates known GRNs along with scRNA-seq and scATAC-seq data in a probabilistic framework [4]. GAEDGRN utilizes gravity-inspired graph autoencoders to capture directed network topology in GRN inference [9].
Table 2: Comparison of GRN Inference Methods and Their Applications
| Method | Approach | Data Requirements | Key Features | Reported Performance |
|---|---|---|---|---|
| LINGER [3] | Lifelong neural network | scMultiome data + external bulk atlas | Manifold regularization using TF motifs | 4-7x relative increase in accuracy over existing methods |
| PRISM-GRN [4] | Bayesian model | scRNA-seq + scATAC-seq ± prior GRN | Biologically interpretable architecture; works with unpaired data | Superior precision in sparse regulatory scenarios |
| GAEDGRN [9] | Graph autoencoder | scRNA-seq + prior GRN | Captures directed network topology; gene importance scoring | High accuracy and reduced training time on 7 cell types |
| MONSTER [1] | Regression-based | Gene expression + TF motifs | Identifies TF drivers of cell state transitions at network level | Reveals regulatory changes in phenotype transitions |
| SCENIC [8] | Regulatory inference | scRNA-seq | Identifies regulons and cellular activity states | Enables cell clustering based on regulatory states |
Validating inferred GRNs requires comparison with experimentally determined ground truth data. Common validation approaches include:
Performance metrics such as Area Under the Receiver Operating Characteristic Curve (AUC) and Area Under the Precision-Recall Curve (AUPR) ratio provide quantitative assessment of inference accuracy [3].
Principle: LINGER leverages lifelong learning to incorporate knowledge from external bulk data while refining predictions on single-cell multiome data, using neural networks with manifold regularization [3].
Workflow:
Input Data Preparation
External Data Integration
Model Pre-training
Model Refinement
GRN Extraction
Principle: SCENIC (Single-Cell Regulatory Network Inference and Clustering) identifies regulons and their activity in single-cell RNA-seq data by combining gene co-expression with transcription factor binding motif analysis [8].
Workflow:
Data Preprocessing
Co-expression Module Identification
Regulon Formation
Cellular Regulon Activity Scoring
Differential Regulon Analysis
Table 3: Key Research Reagent Solutions for GRN Analysis
| Category | Specific Items | Function/Application | Example Uses |
|---|---|---|---|
| Sequencing Technologies | 10x Genomics Single-Cell Multiome | Simultaneous measurement of gene expression and chromatin accessibility | LINGER input data generation [3] |
| Bulk Reference Data | ENCODE RNA-seq/ATAC-seq | Provides external regulatory context across diverse cellular contexts | Training data for LINGER pre-training [3] |
| Motif Databases | JASPAR, CIS-BP | Transcription factor binding specificity profiles | Regulon definition in SCENIC and PRISM-GRN [3] [8] |
| Validation Resources | ChIP-seq datasets | Ground truth for TF-binding events | Validation of trans-regulatory predictions [3] |
| Validation Resources | eQTL datasets (GTEx, eQTLGen) | Links genomic variants to expression changes | Cis-regulatory relationship validation [3] |
| Software Tools | LINGER, PRISM-GRN, SCENIC | GRN inference from single-cell data | Cell type-specific regulatory network reconstruction [3] [4] [8] |
| Perturbation Tools | CRISPR-based editors | Functional validation of regulatory relationships | Causal testing of TF-target relationships [10] |
GRN analysis extends beyond edge identification to network topology characterization. Key topological features include:
Network motifsârecurring regulatory patternsâprovide insights into functional circuit design. Common motifs include feed-forward loops, feedback loops, and single-input modules that generate specific dynamic behaviors.
GRNs evolve through changes in network architecture, with important implications for evolutionary developmental biology (Evo-Devo) [10]. Core regulonsâcontaining functions directly related to the TF-inducing signalâtend to be conserved across species, while extended regulons evolve rapidly to reflect species-specific adaptations [7]. Heterochrony (changes in timing) and homeosis (changes in identity) at the single-cell level can drive the emergence of novel cell types through alterations in GRN architecture [10].
In mammalian hematopoietic stem cells, sequence heterochronyâchanging the order in which transcription factors C/EBPα and GATA become activeâswitches daughter cell identity from eosinophils to basophils [10]. This demonstrates how temporal rewiring of GRNs can generate cellular diversity without changes in gene content.
GRN analysis provides insights into disease mechanisms by identifying dysregulated regulatory programs. In glioblastoma, SCENIC analysis identified IRF and STAT family transcription factors as specific regulators of a unique tumor cell subset that creates an immunomodulatory microenvironment [8]. In chlorine-induced acute lung injury, regulatory network analysis revealed Ets1, Elf1, and Elk3 as key upregulated transcription factors in T cells following exposure [8].
Hypertension research using GRN approaches has demonstrated that genes with differential expression have interactive effects, highlighting the importance of network-level understanding beyond single gene analyses [8]. These applications underscore the translational potential of GRN research in identifying therapeutic targets and biomarkers.
The emergence of single-cell multi-omics technologies represents a paradigm shift in our ability to decipher the complex regulatory logic governing evolutionary developmental biology (evo-devo). These technologies enable the simultaneous measurement of multiple molecular layersâtranscriptomics, epigenomics, and spatial contextâwithin individual cells, providing unprecedented resolution for deconstructing cellular heterogeneity and lineage relationships [11]. For the first time, researchers can systematically infer Gene Regulatory Networks (GRNs) that underlie cell fate decisions, tissue patterning, and phylogenetic constraints across species.
In evo-devo research, GRNs serve as the fundamental computational framework encoding the developmental program of an organism. Traditional bulk sequencing methods obscured cell-to-cell variation, masking rare transitional states and regulatory dynamics critical for understanding evolutionary processes. The integration of single-cell transcriptomics with epigenomic profiling and spatial information now permits the reconstruction of combinatorial regulatory modules that drive cellular differentiation and evolutionary adaptation [12] [11]. This technological revolution is powered by concurrent advances in both experimental methods and computational frameworks, particularly foundation models pretrained on millions of cells that enable zero-shot transfer learning across biological contexts and species [11].
Single-cell multi-omics technologies have evolved from single-modality measurements to integrated platforms that capture complementary molecular features from the same cell. The current landscape encompasses diverse methodological approaches targeting different combinations of omics layers.
Table 1: Single-Cell Multi-Omics Technologies and Applications
| Technology Type | Measured Modalities | Key Applications in Evo-Devo | References |
|---|---|---|---|
| scEpi2-seq | DNA methylation + Histone modifications (H3K9me3, H3K27me3, H3K36me3) | Epigenetic memory during cell type specification; Dynamics of heterochromatin formation | [13] |
| scGPT (Foundation Model) | Cross-modal integration (transcriptomics + epigenomics) | Zero-shot cell annotation; In silico perturbation modeling; Cross-species comparison | [11] |
| cRegulon Framework | TF combinatorics + chromatin accessibility + gene expression | Identification of conserved regulatory modules across species; Cell type attractor states | [12] |
| Spatial Molecular Imaging (CosMx SMI) | Whole transcriptome + protein markers + spatial context | Tissue organization principles; Stem cell niche characterization; Morphogenetic gradients | [14] |
The scEpi2-seq method exemplifies cutting-edge innovation in epigenetic profiling, achieving simultaneous detection of histone modifications and DNA methylation at single-cell resolution. This technique leverages TET-assisted pyridine borane sequencing (TAPS) instead of bisulfite conversion, preserving adapter sequences for more efficient library preparation. Application of scEpi2-seq to mouse intestinal development revealed how H3K27me3 and DNA methylation interact during cell type specification, demonstrating that differentially methylated regions can exhibit independent regulation alongside histone modification control [13].
The complexity of single-cell multi-omics data has driven the development of sophisticated machine learning approaches for GRN inference. These methods have evolved from classical statistical models to deep learning architectures capable of capturing nonlinear regulatory relationships.
Table 2: Machine Learning Methods for GRN Inference from Single-Cell Data
| Algorithm | Learning Type | Key Technology | Compatible Data Types |
|---|---|---|---|
| cRegulon | Unsupervised | Matrix factorization + mixture modeling | scRNA-seq + scATAC-seq |
| GRNFormer | Supervised | Graph Transformer | Single-cell transcriptomics |
| DeepMAPS | Unsupervised | Heterogeneous graph transformer | scATAC-seq + Multi-omic |
| GCLink | Contrastive learning | Graph contrastive link prediction | Single-cell multi-omics |
| scGPT | Foundation model | Transformer + masked gene modeling | Cross-modal integration |
The cRegulon algorithm represents a significant conceptual advance by modeling combinatorial regulation of transcription factors based on diverse GRNs derived from single-cell multi-omics data [12]. Unlike earlier methods that considered single TFs (Regulons) or TF-enhancer units (eRegulons), cRegulon identifies modules of cooperating TFs that collectively regulate common target genes. This approach more accurately reflects the biological reality of developmental regulation, where master transcription factors often act in complexes to determine cell fate. Through benchmarking and applications to real datasets, cRegulon has demonstrated superior performance in identifying TF combinatorial modules as fundamental regulatory units underlying Waddington's epigenetic landscape [12].
Principle: scEpi2-seq enables joint profiling of histone modifications and DNA methylation in single cells by combining antibody-directed chromatin tagging with TET-assisted pyridine borane sequencing [13].
Reagents and Equipment:
Step-by-Step Workflow:
Quality Control Metrics:
Principle: cRegulon infers regulatory modules by modeling combinatorial regulation of transcription factors across multiple cell types/states using single-cell multi-omics data [12].
Input Data Requirements:
Computational Steps:
Implementation Considerations:
Successful implementation of single-cell multi-omics approaches requires both wet-lab reagents and computational resources optimized for evo-devo GRN inference.
Table 3: Essential Research Reagent Solutions for Single-Cell Multi-Omics
| Resource Category | Specific Product/Platform | Function in Workflow |
|---|---|---|
| Spatial Profiling | CosMx Spatial Molecular Imager (Bruker) | High-plex single-cell imaging of RNA and protein in tissue context |
| Epigenetic Profiling | scEpi2-seq Reagent System | Simultaneous detection of histone modifications and DNA methylation |
| Single-Cell Partitioning | 10x Genomics Chromium System | High-throughput single-cell partitioning and barcoding |
| Multi-omic Assay | NEBNext Single-Cell Multi-Omics Kit | Simultaneous profiling of transcriptome and epigenome from same cell |
| Foundation Models | scGPT, scPlantFormer, Nicheformer | Pretrained models for cross-species annotation and perturbation modeling |
| GRN Inference | cRegulon Software Package | Identification of TF combinatorial modules from multi-omics data |
| Data Integration | BioLLM Framework | Standardized benchmarking and integration of single-cell foundation models |
Bruker's CosMx Spatial Molecular Imager enables whole transcriptome imaging at single-cell resolution within intact tissue sections, providing critical spatial context for GRN inference in developing organisms [14]. For computational analysis, foundation models like scGPTâpretrained on over 33 million cellsâdemonstrate exceptional performance in zero-shot cell type annotation and in silico perturbation modeling, significantly accelerating comparative analyses across species [11].
The interpretation of single-cell multi-omics data requires specialized visualization strategies that represent the interconnected nature of transcriptional and epigenetic regulation across spatial contexts.
This integrated framework highlights how different data modalities converge to reconstruct evolutionarily informed GRNs. The modularity of GRNs enables reduction of complexity while preserving biological meaning, with regulatory modules serving as fundamental units that can be compared across species to infer evolutionary trajectories [12] [11]. Tools like cRegulon specifically exploit this modularity to identify conserved and divergent regulatory programs underlying homologous developmental processes.
The single-cell multi-omics revolution has fundamentally transformed our approach to GRN inference in evolutionary developmental biology. By simultaneously profiling transcriptomic, epigenomic, and spatial information from individual cells, researchers can now reconstruct regulatory networks with unprecedented resolution and context specificity. The development of specialized technologies like scEpi2-seq for epigenetic profiling and computational frameworks like cRegulon for identifying combinatorial TF modules represents significant milestones in this rapidly advancing field.
Looking forward, several emerging trends promise to further accelerate discovery. Foundation models pretrained on massive single-cell datasets will enable increasingly accurate cross-species predictions and in silico perturbation studies [11]. The integration of spatial context with multi-omic measurements will illuminate how tissue microenvironment influences regulatory dynamics [14] [15]. Finally, the development of standardized benchmarking platforms like BioLLM will promote reproducibility and collaborative model development across the scientific community [11]. As these technologies mature, they will continue to provide deeper insights into the evolutionary principles that shape developmental gene regulatory networks across the tree of life.
The integration of single-cell multi-omics into evolutionary developmental biology (Evo-Devo) has revolutionized our capacity to decipher the gene regulatory networks (GRNs) that orchestrate cell fate, shape developmental trajectories, and underlie disease mechanisms. GRNs are mathematical representations of the complex interactions between transcription factors (TFs), cis-regulatory elements (CREs), and their target genes that collectively control cellular identity and function [16] [17]. The emerging field of ecological evolutionary developmental biology (Eco-Evo-Devo) emphasizes that these developmental processes are not isolated but are shaped by environmental cues and evolutionary pressures [18]. Rather than being a loose aggregation of research topics, Eco-Evo-Devo provides a coherent conceptual framework for exploring the causal relationships among developmental, ecological, and evolutionary levels, positioning GRN analysis as a central tool for understanding phenotypic diversification [18].
The advent of single-cell technologies has been pivotal, enabling researchers to move beyond bulk tissue analysis and capture the cellular heterogeneity that underpins developmental and evolutionary processes. While bulk sequencing averages signals across thousands of cells, single-cell sequencing (SCS) reveals cell-to-cell variability, identifies rare populations, and allows for the reconstruction of evolutionary relationships [19]. This is particularly powerful in Evo-Devo research, where understanding the emergence of novel cell types and the divergence of developmental pathways across species is paramount. Modern single-cell multi-omics platforms can now simultaneously profile multiple molecular layersâsuch as the transcriptome and epigenomeâfrom the same cell, providing an unprecedented, matched view of the regulatory state [3] [16]. This technological leap addresses the central biological question of how genomic sequence and environmental interaction give rise to organismal form and function through the mechanism of GRN activity.
Inferring GRNs from single-cell multiome data (e.g., simultaneously measured scRNA-seq and scATAC-seq) requires computational methods that can integrate these disparate data types to predict regulatory interactions. The underlying methodological foundations are diverse, each with distinct strengths and limitations for specific biological questions [16].
Table: Methodological Foundations for GRN Inference
| Approach | Underlying Principle | Key Advantages | Common Limitations |
|---|---|---|---|
| Correlation-based | Measures co-expression or association (e.g., Pearson's correlation, mutual information) between TFs and potential target genes [16]. | Simple, intuitive, and computationally efficient. | Cannot distinguish direct from indirect regulation; infers undirected networks [16]. |
| Regression models | Models a target gene's expression as a function of multiple predictor TFs and CREs, using techniques like linear or penalized regression (LASSO) [16]. | Provides interpretable coefficients indicating regulatory strength and direction. | Can be unstable with highly correlated predictors; may overfit without regularization [16]. |
| Probabilistic models | Uses graphical models to represent the probabilistic dependence between variables like TFs and their targets [16]. | Allows for filtering and prioritization of interactions based on probability. | Often relies on distributional assumptions that may not fit all gene expression data [16]. |
| Deep learning models | Employs neural networks (e.g., multi-layer perceptrons, autoencoders) to learn complex, non-linear regulatory relationships from the data [16]. | Highly versatile and can model complex, non-linear mechanisms. | Requires large amounts of data; less interpretable ("black box"); computationally intensive [16]. |
This protocol details the use of two state-of-the-art deep learning methods, LINGER and scTFBridge, which are designed to leverage single-cell multiome data and external knowledge for robust GRN inference.
I. Experimental Design and Data Preparation
II. Computational GRN Inference Procedure
Workflow Title: GRN Inference with Lifelong Learning & Disentanglement
LINGER Workflow (Lifelong neural network for gene regulation) [3]:
BulkNN) designed to predict target gene (TG) expression using TF expression and regulatory element (RE) accessibility as input. Pre-train this model on the large-scale external bulk data (e.g., from ENCODE) to learn a general regulatory profile.scTFBridge Workflow [20]:
scTFBridge) on the single-cell multiome data. The model is designed to separate (disentangle) the latent data representation into components that are shared across the transcriptomic and epigenomic layers and components that are specific to each layer.III. Downstream Analysis and Validation
Table: Essential Research Reagents and Platforms for Single-Cell Multi-omics GRN Studies
| Reagent / Platform | Function / Application | Key Characteristics |
|---|---|---|
| 10x Genomics Multiome ATAC + Gene Expression | Simultaneous profiling of chromatin accessibility (ATAC-seq) and gene expression (RNA-seq) in the same single cell [3] [19]. | Provides a paired view of the epigenomic and transcriptomic state; high-throughput; commercial standard. |
| Fluidigm C1 System | Plate-based microfluidics platform for single-cell isolation and library preparation [19]. | Suitable for processing hundreds of cells with high sensitivity; ideal for full-length transcriptome protocols like Smart-seq3. |
| Mission Bio Tapestri | Targeted scDNA-seq platform for high-throughput genotyping of thousands of cells [19]. | Enables linking of clonal genetic architecture to transcriptional phenotypes; focuses on pre-defined genomic regions. |
| Bioskryb ResolveDNA | Whole-genome amplification platform for single-cell whole-genome or whole-exome sequencing [19]. | Provides broader genomic coverage for analyzing single-nucleotide variants and copy number alterations at lower throughput. |
| JASPAR / Cistrome Databases | Curated databases of transcription factor binding motifs and chromatin profiles [20] [21]. | Provides essential prior knowledge for linking accessible chromatin regions to potential TF binding. |
| ChIP-seq / CUT&Tag | Genome-wide profiling of protein-DNA interactions for specific transcription factors [17]. | Gold-standard experimental method for validating TF binding sites inferred from GRN models. |
| CM-579 | CM-579, CAS:1846570-40-8, MF:C29H40N4O3, MW:492.7 g/mol | Chemical Reagent |
| COH000 | COH000, MF:C25H25NO5, MW:419.5 g/mol | Chemical Reagent |
The neural crest is a vertebrate-specific cell population that gives rise to diverse structures, including parts of the craniofacial skeleton, peripheral nervous system, and pigment cells. Its evolution is considered a key event in vertebrate diversification. A core objective in Evo-Devo is to reconstruct the GRN underlying neural crest development and to understand how it evolved.
Experimental Diagram Title: Reconstructing the Neural Crest GRN Across Species
This integrated approach moves beyond simple gene expression comparisons to reveal the precise architectural changes in the underlying GRN that facilitated the evolution of this key developmental cell type.
A central tenet of Eco-Evo-Devo is that environmental factors can influence development to generate phenotypic variation that is subject to evolutionary selection. This protocol outlines how to use single-cell multi-omics to study the interaction between environment and GRNs in shaping skeletal morphology, a classic Evo-Devo model [18] [24].
Protocol:
The emergence of foundation models represents a paradigm shift in single-cell multi-omics analysis, enabling the learning of universal cellular representations that transform our ability to decipher gene regulatory networks (GRNs). These models, pre-trained on massive datasets comprising millions of cells, capture fundamental biological principles that transfer across diverse downstream tasks. This application note examines two pioneering frameworksâscGPT and scPlantFormerâdetailing their architectural innovations, training methodologies, and experimental protocols for GRN inference in evolutionary developmental biology research. We provide comprehensive benchmarking data, implementation workflows, and reagent solutions to equip researchers with practical tools for leveraging these transformative technologies in drug development and basic research.
Foundation models are large-scale neural networks pre-trained on extensive datasets that learn generalizable representations transferable to various downstream tasks [11]. In single-cell biology, these models have emerged as powerful tools for deciphering cellular heterogeneity, gene regulation, and developmental processes. The conceptual parallel between natural language and cellular biologyâwhere genes correspond to words and cells to documentsâhas enabled the successful adaptation of transformer architectures to biological data [25] [26].
Universal representation learning in single-cell biology involves capturing fundamental principles of gene expression, regulation, and cellular identity that remain consistent across tissues, species, and experimental conditions. scGPT and scPlantFormer exemplify this approach, demonstrating exceptional capability in cross-species annotation, perturbation modeling, and gene regulatory network inference [11] [26]. Their performance stems from pre-training on datasets of unprecedented scaleâ33 million cells for scGPT and 1 million plant cell profiles for scPlantFormerâallowing them to distill critical biological insights concerning genes and cells [25] [26].
For evolutionary developmental biology (evo-devo), these models offer particular promise in uncovering conserved regulatory principles across species and tracing the evolutionary trajectories of cell types. By learning universal representations, foundation models can bridge the gap between model organisms and humans, accelerating the translation of basic research findings into therapeutic insights.
Both scGPT and scPlantFormer build upon the transformer architecture but incorporate distinct modifications tailored to biological data.
scGPT employs a standard transformer decoder architecture similar to GPT models in natural language processing. The model uses gene expressions as input tokens, embedding them into a continuous representation space before processing through multiple transformer blocks with masked self-attention [25] [27]. This design enables scGPT to learn contextual relationships between genes and predict expression values in a self-supervised manner through masked gene modeling.
scPlantFormer implements a transformer encoder architecture with CellMAE (Cell Masked Autoencoding) pretraining, specifically optimized for plant single-cell omics [26]. The model incorporates phylogenetic constraints into its attention mechanism, allowing it to capture evolutionary relationships between species [11]. This design excels at cross-species data integration and resolving batch effects common in plant single-cell datasets.
Table 1: Technical Specifications of Foundation Models
| Feature | scGPT | scPlantFormer |
|---|---|---|
| Architecture Type | Transformer Decoder | Transformer Encoder with CellMAE |
| Pre-training Data | 33+ million cells [25] | 1 million Arabidopsis thaliana cells [26] |
| Pre-training Objectives | Masked gene modeling, contrastive learning [11] | Mean square error loss, plant-specific optimization [26] |
| Key Innovation | Generative pre-training across diverse cell types | Phylogenetic constraints in attention mechanism |
| Handling Multi-omics | Native support for multi-omic integration [25] | Specialized for plant transcriptomics with cross-species capabilities |
Figure 1: Foundation Model Training Workflow. Both scGPT and scPlantFormer follow this general pipeline, transforming single-cell data into universal representations through self-supervised pre-training.
Protocol 1: Single-Cell Data Preprocessing for Foundation Models
Quality Control Metrics
Normalization Procedure
Batch Effect Considerations
Protocol 2: Transfer Learning for Downstream Tasks
Task Formulation
Fine-tuning Procedure for scGPT
Fine-tuning Procedure for scPlantFormer
Protocol 3: Gene Regulatory Network Inference
Data Requirements
Implementation with LINGER Framework
Validation Procedures
Table 2: Benchmarking Results Across Downstream Tasks
| Task | Model | Performance Metrics | Comparative Advantage |
|---|---|---|---|
| Cell Type Annotation | scGPT | 92% accuracy on PBMC datasets [25] | Zero-shot capability to novel cell types |
| scPlantFormer | 92% cross-species accuracy [11] | Effective across plant species | |
| GRN Inference | LINGER | 4-7x relative increase in accuracy vs. baselines [3] | Incorporates atlas-scale external data |
| scGPT | Superior AUPRC in perturbation prediction [25] | Generative modeling of network dynamics | |
| Multi-omic Integration | GLUE | Lowest FOSCTTM error in benchmark [29] | Graph-linked embedding for distinct feature spaces |
| scGPT | Effective integration of transcriptomics and epigenomics [25] | Unified representation space | |
| Cross-species Analysis | scPlantFormer | Accurate annotation between Arabidopsis and rice [26] | Phylogenetic attention mechanism |
Foundation models demonstrate particular utility in evo-devo research through their ability to:
Identify Conserved Regulatory Programs
Model Developmental Processes
Accelerate Translational Applications
Figure 2: GRN Inference Workflow for Evo-Devo Research. Foundation models integrate multi-omic data with external resources to infer regulatory networks applicable to evolutionary studies.
Table 3: Key Research Reagents and Computational Tools
| Resource | Type | Function | Access |
|---|---|---|---|
| scGPT Model Zoo | Pre-trained Models | Task-specific fine-tuning starting points | GitHub: bowang-lab/scGPT [27] |
| scPlantFormer | Plant Foundation Model | Cross-species analysis in plants | GitHub Repository [26] |
| LINGER | GRN Inference | Lifelong learning for regulatory networks | Nature Biotechnology [3] |
| GLUE | Multi-omic Integration | Graph-linked unified embedding | Nature Biotechnology [29] |
| CellXGene | Data Repository | 33+ million cells for reference mapping | CZ CELLxGENE Discover [11] |
| BioLLM | Benchmarking Framework | Standardized evaluation of foundation models | [11] |
The integration of foundation models into single-cell multi-omics analysis represents a transformative advancement with profound implications for evolutionary developmental biology and drug development. scGPT and scPlantFormer exemplify how universal representation learning can uncover fundamental principles of gene regulation that transcend species boundaries.
Future developments will likely focus on multimodal integrationâcombining transcriptomic, epigenomic, proteomic, and spatial data within unified foundation models [11]. Additionally, interpretability enhancements will be crucial for translating model predictions into mechanistic biological insights, particularly for identifying key regulatory drivers of development and disease [11] [31].
For drug development professionals, these models offer accelerating pathways from target identification to validation by predicting cell type-specific responses to perturbations and identifying conserved regulatory mechanisms across species. The ability to conduct in silico experiments on universal representations provides an efficient alternative to costly screening approaches while offering insights into fundamental biological processes.
As foundation models continue to evolve, they will increasingly serve as the computational scaffold for understanding the regulatory logic of development, evolution, and diseaseâultimately bridging the gap between cellular omics and actionable biological understanding.
The Waddington epigenetic landscape serves as a powerful metaphor for understanding cellular differentiation and fate decisions during development. Originally proposed by Conrad H. Waddington, this conceptual framework depicts a developing cell as a ball rolling down a sloping hillside characterized by valleys and ridges [32]. The path taken by the ball represents a specific developmental trajectory, with branching points corresponding to fate decisions, and the end points of the valleys (the basins) representing the terminally differentiated cell types [33] [32]. In modern systems biology, this metaphorical landscape has been formalized mathematically, where the various cell states are conceptualized as attractors within a high-dimensional state space defined by the activity levels of genes in a gene regulatory network (GRN) [32] [34]. The stability of a cell type is then understood in terms of the depth and size of these attractor basins, with the barriers between basins correlating with the difficulty of transitioning from one cell state to another [32].
This framework has gained renewed importance with the advent of single-cell multi-omics technologies, which enable the simultaneous measurement of gene expression and chromatin accessibility in individual cells [3] [35]. These rich datasets provide an unprecedented opportunity to infer the architecture of GRNs and computationally reconstruct the Waddington landscape for specific biological systems, from embryonic development to disease processes like cancer [33]. The ability to quantify this landscape offers profound insights into the mechanisms underlying cellular heterogeneity, phenotypic plasticity, and the reprogramming of cell identity [32]. This Application Note provides detailed protocols for applying this framework through GRN inference from single-cell multiome data, with specific examples from developmental biology and cancer research.
In contemporary quantitative biology, Waddington's metaphorical landscape is formalized through dynamical systems theory. The state of a cell is represented by a vector ( S(t) = (x1, x2, ..., xN) ), where each ( xi ) represents the expression value or cellular concentration of the product of gene ( i ) in an N-gene regulatory network [32]. The probability landscape ( P ) is defined over this N-dimensional state space, where states with locally highest probability (lowest potential, where ( U = -\ln P )) represent the attractor states of the underlying GRN [32]. These attractors correspond to distinct cell phenotypesâincluding stable differentiated states, progenitor states, and pathological states like cancer subtypes [33] [32].
The stability of these cellular states is quantitatively related to the barrier heights between attractor basins in the landscape. Higher barriers correspond to longer escape times, making transitions between cell types less likely and thus representing more stable cellular states [32]. This quantitative relationship provides crucial insights for understanding differentiation processes and designing cellular reprogramming strategies. The developmental progression from a pluripotent stem cell to differentiated lineages can be visualized as a ball moving from the broad, elevated basin of the undifferentiated state down to deeper, more confined basins representing terminally differentiated cells [32].
Several mathematical approaches exist for quantifying the Waddington landscape, with Hopfield networks providing one particularly powerful framework [34]. These auto-associative artificial neural networks store patterns as attractors of the network and can recall these patterns from noisy or incomplete inputs, effectively modeling the recall of stable gene expression patterns during differentiation [34]. For a network of N genes, the state of the system is defined by the expression levels of these genes, and the attractors represent the stable expression configurations corresponding to different cell types.
An alternative approach utilizes ordinary differential equations (ODEs) based on known regulatory interactions [33]. For a GRN model, the system can be described by equations of the form ( dx/dt = F(x) ), where ( x ) is a vector of gene expression levels and ( F ) represents the regulatory logic [33] [32]. The epigenetic landscape can then be constructed by performing numerous stochastic simulations from different initial conditions and calculating the probability distribution of the states [33]. For instance, in a study of breast cancer subtypes, 10,000 stochastic simulations were used to map the attractor landscape containing HER2+ and triple-negative breast cancer (TNBC) basins [33].
Figure 1: The Waddington Landscape Metaphor for Cell Fate Decisions. This diagram visualizes the progressive restriction of cell fate during development, from pluripotent stem cells to various differentiated states through branching valleys on an epigenetic landscape.
The LINGER (Lifelong neural network for gene regulation) framework represents a significant advance in GRN inference from single-cell multiome data, achieving a fourfold to sevenfold relative increase in accuracy over existing methods [3]. Unlike approaches that rely on gene expression data alone, LINGER integrates single-cell paired gene expression and chromatin accessibility data while incorporating atlas-scale external bulk data across diverse cellular contexts [3]. This approach addresses a fundamental challenge in GRN inference: learning complex regulatory mechanisms from limited independent data points. The method incorporates prior knowledge of transcription factor (TF) motifs as a manifold regularization, significantly enhancing prediction accuracy for both cis-regulatory and trans-regulatory interactions [3].
LINGER employs a lifelong learning strategy (also called continuous learning), where knowledge learned from past data (external bulk datasets) helps learn new tasks (single-cell data) with limited data more effectively [3]. The framework constructs GRNs containing three types of interactions: trans-regulation (TF-TG), cis-regulation (RE-TG), and TF-binding (TF-RE), providing a comprehensive view of gene regulatory mechanisms [3]. After initial GRN inference from reference single-cell multiome data, LINGER enables estimation of transcription factor activity solely from bulk or single-cell gene expression data, leveraging abundant gene expression datasets to identify driver regulators in case-control studies [3].
Table 1: LINGER Model Components and Their Functions
| Component | Architecture | Function | Key Parameters |
|---|---|---|---|
| BulkNN | Three-layer neural network | Pre-training using external bulk data | Layer weights and biases |
| scNN | Three-layer neural network | Single-cell data refinement | Regulatory module parameters |
| Manifold Regularization | Motif-based constraint | Enrichment of TF motifs binding to REs in same regulatory module | Regularization strength |
| EWC Loss | Elastic Weight Consolidation | Preservation of knowledge from bulk data during single-cell training | Fisher information matrix |
Pre-training on bulk data: Initialize the neural network model (BulkNN) to predict target gene (TG) expression using TF expression and regulatory element (RE) accessibility as input. Train this model on the external bulk data to learn initial regulatory relationships [3].
Refinement on single-cell data: Apply elastic weight consolidation (EWC) loss to refine the model on single-cell data while preserving knowledge gained from bulk data [3]. The EWC regularization uses the Fisher information to determine the magnitude of parameter deviation, encouraging the posterior distribution to remain close to the prior distribution derived from bulk data [3].
Regulatory strength inference: After training, infer the regulatory strength of TF-TG and RE-TG interactions using Shapley values, which estimate the contribution of each feature for each gene [3]. Calculate TF-RE binding strength from the correlation of TF and RE parameters learned in the second layer of the neural network [3].
GRN assembly: Construct cell population GRNs, cell type-specific GRNs, and cell-level GRNs based on the general GRN and cell type-specific profiles derived from the model [3].
Performance validation: Validate trans-regulatory inferences against ChIP-seq data by calculating the area under the receiver operating characteristic curve (AUC) and area under the precision-recall curve (AUPR) ratio [3]. For cis-regulatory inferences, assess consistency with eQTL studies by grouping RE-TG pairs by distance and comparing AUC and AUPR ratios across different distance groups [3].
Downstream analysis applications:
Figure 2: LINGER Workflow for GRN Inference. This diagram illustrates the lifelong learning approach that integrates external bulk data and TF motif information with single-cell multiome data to infer gene regulatory networks at multiple resolutions.
Table 2: Essential Research Reagents for Single-Cell Multiome GRN Inference
| Reagent/Resource | Function | Example Sources |
|---|---|---|
| 10x Genomics Multiome | Simultaneous profiling of gene expression and chromatin accessibility from the same single cell | 10x Genomics |
| ENCODE Consortium Data | Atlas-scale external bulk data for pre-training | ENCODE Project |
| ChIP-seq Validation Data | Ground truth for trans-regulatory interactions | Cistrome Data Browser |
| eQTL Data (GTEx/eQTLGen) | Validation of cis-regulatory predictions | GTEx Portal, eQTLGen |
| TF Motif Databases | Prior knowledge for TF-RE connections | JASPAR, CIS-BP |
The attractor landscape framework provides particular insight into cancer heterogeneity, where multiple cell states coexist within tumors and contribute to therapy resistance and cancer recurrence [33]. In a recent study of breast cancer subtypes, researchers investigated how NF-κB epigenetic variability contributes to progression of the HER2+ BC subtype toward a more aggressive triple-negative breast cancer (TNBC) phenotype [33]. The study focused on a GRN involving NF-κB and key epithelial-mesenchymal transition (EMT) transcription factors TWIST1, SIP1, and SLUG, which constitute a broader regulatory network active across diverse tumor contexts [33].
This GRN exhibits an epigenetic landscape with two attractor basins corresponding to the observed expression profiles of HER2+ and TNBC subtypes, separated by an unstable stationary state [33]. The research demonstrated that stochastic fluctuations in NF-κB levels induce spontaneous irreversible transitions from HER2+ to TNBC attractor basins at different times, contributing to subtype heterogeneity [33]. This system provides an excellent model for studying attractor dynamics in a pathological context and illustrates how the Waddington landscape framework can explain cellular plasticity and fate transitions in cancer.
Network definition: Construct a GRN model that includes NF-κB (p65:p50 heterodimer) as a key regulator that activates expression of TWIST1, SNAIL, and SLUG, as well as its own subunits p50 and p65 through positive feedback [33].
Ordinary Differential Equations: Implement ODEs based on the regulatory interactions. For a general GRN with genes ( x1, x2, ..., xn ), the system can be described by: [ \frac{dxi}{dt} = Fi(x1, x2, ..., xn) - \gammai xi ] where ( Fi ) represents the regulatory logic governing production and ( \gammai ) represents degradation [33].
Parameter calibration: Calibrate model parameters using experimentally measured RNA levels of all network components across the cell types of interest (e.g., HER2+ and TNBC subtypes) simultaneously [33]. Fine-tune parameters to reproduce temporal dynamics, such as recovery times after perturbation with NF-κB inhibitor DHMEQ [33].
Stationary state identification: Solve the algebraic equations derived from the ODE system (set ( dx/dt = 0 )) to find all stationary states [33]. Characterize the stability of each state by analyzing the eigenvalues of the Jacobian matrix.
Stochastic simulations: Perform numerous stochastic simulations (e.g., 10,000 simulations) starting from different uniformly distributed initial conditions in the phase space [33]. Use methods like the Gillespie algorithm or Langevin dynamics to incorporate intrinsic noise.
Landscape visualization: Construct the epigenetic landscape by calculating the probability distribution of states from the stochastic simulations [33]. Visualize the landscape in 2D or 3D projections using key genes or factors as axes (e.g., p50 vs p65) [33].
Transition analysis: Simulate long-term single-cell trajectories to observe spontaneous transitions between attractor basins [33]. Analyze the directionality and timing of these transitions, noting that irreversible transitions indicate asymmetry in the landscape topography.
Perturbation experiments: Treat cells with pathway-specific inhibitors (e.g., DHMEQ for NF-κB inhibition) and measure recovery dynamics of network components [33]. Compare experimental recovery times with model predictions.
Single-cell analysis: Use single-cell RNAseq to characterize heterogeneity in patient samples and cell lines [33]. Analyze correlation patterns between network components (e.g., p65 and p50) across different subtypes, as more symmetric attractor basins may exhibit lower correlation [33].
Attractor stability assessment: Evaluate how mutations or drugs alter attractor basin sizes and transition probabilities by measuring their effects on gene expression patterns and subtype proportions [33].
Figure 3: GRN Model for Breast Cancer Subtypes. This network diagram shows the key regulatory interactions in the NF-κB-centered GRN that generates two attractor states corresponding to HER2+ and TNBC breast cancer subtypes.
Table 3: Essential Reagents for Breast Cancer Attractor Landscape Study
| Reagent/Resource | Function | Example Sources |
|---|---|---|
| Breast Cancer Cell Lines | Model systems for different subtypes (HCC-1954 for HER2+, MDA-MB-231 for TNBC) | ATCC |
| NF-κB Inhibitor (DHMEQ) | Perturbation agent to test network dynamics and recovery | Commercial suppliers |
| qPCR Reagents | Quantification of NF-κB, TWIST1, SNAIL, and SLUG expression | Various manufacturers |
| Single-cell RNAseq Kits | Characterization of heterogeneity in cell populations | 10x Genomics, Parse Biosciences |
Boolean networks (BNs) provide a powerful, coarse-grained approach to modeling GRNs, where genes are represented as binary agents (ON/OFF) and their interactions are captured through Boolean functions [36]. In this framework, attractorsâstable states or cycles of states of the systemâare associated with biological phenotypes, making their identification crucial for understanding cellular differentiation and fate decisions [36]. However, traditional computing approaches struggle with the exponential growth of the state space in such models (with ( 2^n ) possible states for n genes), limiting exhaustive simulations to relatively small networks (typically ( n \leq 29 )) [36].
A novel quantum search algorithm has been developed for identifying attractors in synchronous Boolean networks, offering potential advantages for analyzing larger GRNs [36]. This algorithm uses iterative quantum amplitude suppression to systematically identify all attractors of a Boolean network, with each run guaranteed to discover a new attractor [36]. The approach demonstrates strong resilience to noise on current NISQ (noisy intermediate-scale quantum) devices, making it a promising advance toward practical quantum-enhanced biological modeling [36].
Gene binarization: Convert gene expression data to binary values (ON/OFF) using appropriate thresholding methods. For single-cell data, this can be based on expression levels relative to cell population distributions.
Boolean function definition: For each gene, define a Boolean function that determines its state based on the states of its regulators. These functions can be derived from known regulatory interactions or inferred from data.
Update scheme specification: Implement a synchronous update scheme where all genes are updated simultaneously at each time step based on the current state of the system [36].
Qubit initialization: Initialize the qubit system to a superposition of all possible states using Hadamard gates applied to each qubit [36]. For n genes, this creates a superposition of all ( 2^n ) possible states.
Boolean function application: Implement the Boolean functions as quantum gates that evolve the superposition state [36]. Each possible network state evolves into its corresponding time-evolved state according to the synchronous update rules.
Iterative amplitude suppression:
Noise mitigation: Implement error mitigation strategies tailored to the specific quantum hardware being used, as the algorithm has shown robustness to noise on current NISQ devices like the ibm_brisbane processor [36].
Classical verification: For small networks, verify quantum algorithm results using classical methods such as the BoolNet package in R [36].
Biological interpretation: Map identified attractors to known or potential cellular phenotypes based on their gene expression patterns.
Basin analysis: Analyze the size and structure of attractor basins to assess the relative stability of different cellular states.
Table 4: Computational Resources for Quantum-Enhanced GRN Analysis
| Resource | Function | Access Method |
|---|---|---|
| Quantum Computing Simulators | Algorithm development and testing without quantum hardware | Qiskit, Cirq, Forest |
| IBM Quantum Processors | Implementation and execution of quantum algorithms | IBM Quantum Experience |
| BoolNet R Package | Classical verification of attractor identification | CRAN Repository |
| Quantum Algorithm Libraries | Pre-built functions for quantum amplitude suppression | Open-source quantum computing libraries |
The Waddington landscape framework provides a powerful conceptual and quantitative approach for understanding cellular differentiation and fate decisions in the context of gene regulatory networks. The protocols outlined hereâranging from GRN inference using cutting-edge machine learning methods like LINGER to attractor landscape construction in specific biological systems and the emerging application of quantum algorithms for attractor identificationâprovide researchers with practical tools for applying this framework to their specific research questions.
The integration of single-cell multi-omics data with increasingly sophisticated computational methods is transforming the Waddington landscape from a qualitative metaphor to a quantitative, predictive framework [3] [35]. This enables not only deeper understanding of developmental processes and disease mechanisms but also practical applications in drug development and cellular reprogramming. As these methods continue to evolve, particularly with advances in quantum computing and AI-driven modeling, we anticipate increasingly accurate and comprehensive reconstructions of epigenetic landscapes that will further illuminate the fundamental principles governing cellular identity and fate decisions.
In evolutionary developmental biology (evo-devo), understanding the gene regulatory networks (GRNs) that control cellular identity and morphological diversity is a fundamental pursuit. The advent of single-cell multi-omics technologies, which allow for the simultaneous measurement of a cell's transcriptome and epigenome (e.g., via paired scRNA-seq and scATAC-seq), has provided the data necessary to reconstruct these networks with unprecedented resolution [3] [37]. The computational challenge lies in accurately inferring the causal regulatory relationships between transcription factors (TFs), cis-regulatory elements (CREs), and their target genes from these sparse and noisy data. This has led to the development of diverse computational approaches, each with distinct foundations and assumptions, which are critically assessed in this application note.
Computational methods for GRN inference from single-cell multi-omics data can be broadly categorized into several paradigms. The table below summarizes the core principles, key tools, and specific applications of these foundational approaches.
Table 1: Foundational Computational Approaches for GRN Inference from Single-Cell Multi-omics Data
| Methodological Foundation | Core Principle | Representative Tools | Application in GRN Inference |
|---|---|---|---|
| Correlation & Association | Identifies co-variation or spatial association between TF expression/activity and target gene expression, or between chromatin accessibility and gene expression. | scSAGRN [37], PCC [3] | Infers TF-gene and peak-gene linkages based on statistical dependencies, often using neighborhood information from integrated data. |
| Regression Models | Models the expression of a target gene as a linear or non-linear function of the expression levels of TFs and the accessibility of CREs. | Elastic Net, LINGER (initial layer) [3] | Directly predicts gene expression from regulator activities, with coefficients interpreted as regulatory strengths. |
| Probabilistic Models | Uses a Bayesian framework to integrate multi-omics data and prior knowledge (e.g., known GRNs, motifs) to infer a posterior probability of regulatory interactions. | PRISM-GRN [4], BREM-SC [38] | Provides robust, interpretable GRN inference with inherent uncertainty quantification, valuable for sparse single-cell data. |
| Deep Learning Models | Employs multi-layer neural networks (e.g., VAEs, GANs) to learn complex, non-linear relationships between input modalities and infer latent regulatory structures. | LINGER [3], scMODAL [39], BABEL [38] | Captures high-order interactions; can be enhanced with lifelong learning from external atlas-scale data. |
| Collismycin A | Collismycin A | Bench Chemicals | |
| Conteltinib | Conteltinib, CAS:1384860-29-0, MF:C32H45N9O3S, MW:635.8 g/mol | Chemical Reagent | Bench Chemicals |
The performance of GRN inference methods is typically benchmarked using ground truth data from independent experimental sources, such as ChIP-seq for TF-target gene interactions and eQTL studies for cis-regulatory linkages [3] [37]. Key evaluation metrics include the Area Under the Receiver Operating Characteristic Curve (AUC) and the Area Under the Precision-Recall Curve (AUPR), with the latter being particularly informative for imbalanced datasets where true edges are sparse.
Table 2: Performance Benchmarking of Selected GRN Inference Methods
| Method | Foundation | Reported Performance | Key Advantage |
|---|---|---|---|
| LINGER [3] | Deep Learning (Lifelong) | 4x to 7x relative increase in AUC over existing methods; superior AUPR ratio across blood cell ChIP-seq ground truths. | Leverages atlas-scale external bulk data for highly accurate inference from limited single-cell data. |
| scSAGRN [37] | Spatial Association | Outperforms FigR, GLUE, SEASTAR, and SCENIC+ in TF recovery, peak-gene, and TF-gene linkage prediction on human bone marrow and cell line data. | Effectively identifies key activating and repressive TFs by incorporating spatial correlation. |
| PRISM-GRN [4] | Probabilistic (Bayesian) | Superior precision in GRN reconstruction, especially for sparse true interactions; performs well with unpaired data and limited prior information. | Biologically interpretable architecture; robust and flexible across various biological contexts. |
| Elastic Net [3] | Regression | Lower prediction accuracy for gene expression compared to non-linear neural network models, particularly for genes with negative correlations. | Provides a simple, linear baseline model. |
This section outlines a generalized workflow for inferring and validating GRNs from single-cell multiome data, synthesizing protocols from the cited methodologies.
Goal: Prepare scRNA-seq and scATAC-seq count matrices for GRN inference. Reagents & Materials:
Procedure:
Goal: Infer a high-confidence GRN by leveraging a deep learning model pre-trained on external bulk data. Reagents & Materials:
Procedure:
The following diagram illustrates the core LINGER workflow:
Goal: Assess the biological accuracy of the inferred GRN and perform evolutionary or developmental analysis. Reagents & Materials:
Procedure:
The following table details key computational and data resources essential for conducting GRN inference studies.
Table 3: Essential Research Reagents and Resources for GRN Inference
| Resource Name | Type | Function in GRN Research |
|---|---|---|
| 10x Genomics Multiome Kit | Wet-lab Reagent | Generates paired scRNA-seq and scATAC-seq libraries from the same single cell, providing the foundational data for GRN inference. |
| ENCODE Consortium Data | Reference Data | Provides a large-scale atlas of bulk RNA-seq, ChIP-seq, and ATAC-seq across cell types, used for pre-training models like LINGER's BulkNN [3]. |
| JASPAR/CIS-BP | Database | Curated databases of TF binding motifs, used to connect accessible chromatin regions to potential binding TFs [3]. |
| GTEx/eQTLGen | Validation Data | Provide genotype-gene expression association data, serving as ground truth for validating predicted cis-regulatory interactions (RE-TG links) [3]. |
| LINGER Software | Computational Tool | A deep learning framework that uses lifelong learning to achieve high-accuracy GRN inference from single-cell multiome data [3]. |
| PRISM-GRN Software | Computational Tool | A Bayesian model that incorporates prior knowledge and multi-omics data for precise, robust, and interpretable GRN reconstruction [4]. |
| scMODAL Software | Computational Tool | A deep learning framework for integrating diverse single-cell omics data, which can support downstream GRN analysis [39]. |
| TRRUST | Database | A curated database of human and mouse transcriptional regulatory networks, useful as prior knowledge or for validation [37]. |
| CPI-4203 | CPI-4203, MF:C16H14N4O, MW:278.31 g/mol | Chemical Reagent |
| CPI-455 |
The following diagram summarizes the logical flow of information and analysis steps in a standard GRN inference pipeline, from raw data to biological insight.
Inference of Gene Regulatory Networks (GRNs) is a central problem in computational biology, essential for understanding how cells alter gene expression in response to different environments and how noncoding genetic variants cause disease [3]. GRNs are composed of transcription factors (TFs) that bind DNA regulatory elements to activate or repress the expression of target genes [3]. Despite extensive efforts, GRN inference accuracy has remained disappointingly low, marginally exceeding random predictions [3]. The LINGER (Lifelong neural network for gene regulation) framework represents a paradigm shift in this field, achieving a fourfold to sevenfold relative increase in accuracy over existing methods by incorporating atlas-scale external bulk data across diverse cellular contexts and prior knowledge of transcription factor motifs as a manifold regularization [3].
LINGER is a computational framework designed to infer GRNs from single-cell multiome data using count matrices of gene expression and chromatin accessibility along with cell type annotation as input [3]. It outputs a cell population GRN, cell type-specific GRNs, and cell-level GRNs, each containing three types of interactions: trans-regulation (TFâTG), cis-regulation (REâTG), and TF-binding (TFâRE) [3]. The framework integrates comprehensive gene regulatory profiles from external bulk data through lifelong machine learning (also called continuous learning), where knowledge learned in the past helps learn new tasks with little data or effort [3].
The LINGER framework implements a three-layer neural network model to fit the expression of target genes (TGs), taking as input TF expression and the accessibility of regulatory elements (REs) [3]. The second layer consists of weighted sums of TFs and REs, forming regulatory modules guided by TFâRE motif matching through manifold regularization [3]. This leads to the enrichment of TF motifs binding to REs that belong to the same regulatory module [3].
LINGER's approach addresses three major challenges in GRN inference:
Input Data Requirements:
Preprocessing Steps:
Phase 1: Pre-training on External Bulk Data
Phase 2: Refinement on Single-cell Data
Phase 3: Regulatory Network Extraction
Performance Assessment:
Table 1: Quantitative Performance Comparison of LINGER Against Baseline Methods
| Method | AUC (Trans-regulation) | AUPR Ratio (Trans-regulation) | AUC (Cis-regulation, eQTLGen) | AUPR Ratio (Cis-regulation, eQTLGen) |
|---|---|---|---|---|
| LINGER | Significantly higher | Significantly higher | Higher across all distance groups | Higher across all distance groups |
| scNN | Moderate | Moderate | Moderate | Moderate |
| BulkNN | Low | Low | Low | Low |
| PCC | Lowest | Lowest | Lowest | Lowest |
The following diagram illustrates the complete LINGER workflow, from data input through network inference and downstream applications:
Table 2: Essential Research Reagents and Computational Tools for GRN Inference
| Resource Type | Specific Examples | Function in GRN Inference |
|---|---|---|
| Single-cell Multiome Platforms | 10x Genomics Multiome ATAC-seq + Gene Expression | Provides paired measurements of chromatin accessibility and gene expression from the same single cell [3] |
| External Reference Data | ENCODE project bulk data [3], GTEx eQTLs [3], eQTLGen [3] | Provides atlas-scale regulatory information across diverse cellular contexts for training and validation |
| Prior Knowledge Databases | Transcription factor motif databases (JASPAR, CIS-BP) | Informs TF-RE binding predictions through motif matching and manifold regularization [3] |
| Validation Data Sources | ChIP-seq datasets [3], eQTL studies [3] | Serves as ground truth for benchmarking trans-regulatory and cis-regulatory predictions |
| Computational Frameworks | LINGER implementation [3], BEAST2 for phylogenetic inference [40] | Provides algorithms for network inference and evolutionary analysis |
| Experimental Model Systems | PBMCs from 10x Genomics [3], CAMA-1 breast cancer cell line [40], Pristionchus pacificus [41] | Offer biologically relevant systems for method development and validation |
The integration of GRN inference with evolutionary developmental biology (evo-devo) provides powerful insights into the regulatory mechanisms underlying phenotypic diversity. LINGER enables the reconstruction of evolutionary relationships among cells by linking genotypic and phenotypic variation, which is particularly valuable for understanding developmental decisions in evolutionary model systems [40] [41]. For example, in the nematode Pristionchus pacificus, which exhibits developmental plasticity of mouth form and feeding behavior, GRN inference can reveal the regulatory network underlying evolutionary novelties [41]. The mouth-dimorphism (predator vs. bacterivore) in P. pacificus is determined by a complex regulatory network involving key regulators eud-1, sult-1, and nhr-40, where subtle changes in regulatory structure can influence evolutionary trajectories [41].
A significant challenge in evolutionary GRN inference is structural uncertaintyâmultiple competing model structures that could explain the same biological data [41]. This is particularly relevant in evolutionary contexts where regulatory networks may have diverged between species or populations. Comprehensive model comparison approaches, such as fitting thousands of distinct regulatory network models to gene expression data across experimental conditions, can identify conserved regulatory features supported by data across multiple contexts [41]. LINGER's ability to integrate diverse data sources and prior knowledge provides a robust framework for addressing this structural uncertainty in evolutionary studies.
Table 3: LINGER Performance in Evolutionary Developmental Biology Contexts
| Application Domain | Key Regulatory Components Identified | Biological Insight Gained |
|---|---|---|
| Mouth-form Evolution in Nematodes | eud-1, sult-1, nhr-40 regulatory network [41] | High degree of positive regulation and interconnectivity between key developmental regulators [41] |
| Cancer Evolution and Drug Resistance | K-Ras, β-catenin pathways in breast cancer [40] | Multiple genetic lineages independently acquired resistance mechanisms, suggesting need for multi-lineage targeting strategies [40] |
| Developmental Plasticity | Environmental influence on phenotypic ratios [41] | Regulatory networks that enable flexible responses to environmental cues while maintaining developmental robustness |
A critical challenge in GRN inference is practical indistinguishabilityâthe inability to uniquely infer network structure given typical experimental data constraints [41]. This arises from several factors characteristic of biological data: intrinsic and extrinsic noise, limited temporal sampling, and the impossibility of measuring all relevant system states [41]. LINGER addresses these challenges through several mechanisms:
The maximalist approach of comparing thousands of potential network structures, as demonstrated in evolutionary studies of P. pacificus, represents a powerful strategy for robust GRN inference [41]. However, this approach is computationally intensive and may not be feasible for all research questions. LINGER provides a more efficient alternative through its neural network architecture and regularization strategies, while still accommodating structural uncertainty through its flexible framework.
LINGER enables enhanced interpretation of disease-associated variants and genes through several mechanisms:
For drug development professionals, LINGER offers several valuable capabilities:
The framework's ability to reconstruct cancer cell phylogenetic relationships and link them to phenotypic outcomes, such as drug resistance in breast cancer and multiple myeloma tumors, demonstrates its utility across biological contexts and disease stages [40]. By revealing evidence of genetic evolution across disease progression and convergent evolution where multiple lineages evolve toward a common mechanism of resistance, LINGER provides insights essential for developing durable therapeutic strategies [40].
Gene regulatory networks (GRNs) represent the complex collections of molecular regulators that interact to govern cellular gene expression, ultimately determining cell function, fate, and phenotypic outcomes [42]. In evolutionary developmental biology (evo-devo), understanding how these networks evolve to produce diverse body plans and structures represents a central challenge [43] [44]. While traditional GRN analysis has focused on linear regulator-target relationships, emerging evidence indicates that combinatorial regulation - where multiple transcription factors (TFs) collaborate to control common target genes - plays a pivotal role in developmental processes and evolutionary innovation [12].
Current regulatory units like Regulons (target genes controlled by a single TF) or eRegulons (incorporating regulatory elements) fail to capture the cooperative nature of TF interactions observed in key developmental processes [12]. For instance, in mammalian embryonic stem cells, Sox2, Nanog, and Pou5f1 cooperatively govern pluripotency, while MYOD, MYOG, MEF2A, and MYF6 collectively dictate skeletal muscle specification [12]. Similarly, studies of deuterostome body axis formation have revealed conserved GRNs orchestrated by Nodal, Gdf1/3, and Lefty that exhibit evolutionary rewiring through enhancer hijacking events [44].
The cRegulon framework addresses this gap by modeling combinatorial regulation from single-cell multi-omics data, providing regulatory units that underpin the cell type landscape and offering new insights into the modular architecture of GRNs across evolutionary timescales [12] [45].
cRegulon represents a computational framework for identifying combinatorial regulatory modules from single-cell multi-omics data. Specifically, a cRegulon comprises:
This framework extends beyond existing methods by simultaneously modeling TF combinatorial interactions while accounting for cell-type specificity in regulation. The GRN of a cell type may be composed of several distinct cRegulons that regulate gene expression in distinct pathways or processes, while a single cRegulon may be shared by GRNs of multiple cell types, reflecting evolutionary conservation of regulatory modules [12].
Table 1: Core Components of a cRegulon
| Component | Description | Biological Significance |
|---|---|---|
| TF Module | Set of transcription factor pairs that cooperatively regulate targets | Determines specificity of regulatory control; often evolutionarily conserved |
| Regulatory Elements | DNA regions (enhancers, promoters) where TFs bind | Provides context-specific accessibility and modulation |
| Target Genes | Genes whose expression is controlled by the TF module | Executes functional programs defining cell identity |
The cRegulon pipeline begins with single-cell multi-omics data, typically paired scRNA-seq and scATAC-seq data, which can be aligned at either the cell level or context level [12]. The initial processing stages include:
This foundation enables subsequent identification of combinatorial regulation patterns across different cellular contexts.
The core innovation of cRegulon lies in its approach to detecting TF combinatorial interactions:
For each cluster-specific GRN, cRegulon combines co-regulation effects and activity specificities to define a combinatorial effect for each TF pair. This generates a matrix (C) containing pairwise combinatorial effects across all cell clusters [12].
To model the modularity of GRNs, the method assumes (C) can be approximated by a mixture of rank-1 matrices, each corresponding to a module of co-regulating TFs. The optimization model is designed to identify these TF modules across all cell clusters simultaneously with cell type annotation [12].
The mathematical formulation involves solving:
[ \min \|C - \sum{k=1}^{K} \lambdak uk uk^\top\|_F^2 ]
where (uk) represents the k-th TF module, (\lambdak) represents its contribution, and (K) is the number of modules [12].
For successful cRegulon analysis, researchers should prepare:
Data Integration and Normalization
GRN Inference for Cell Clusters
cRegulon Identification
Validation and Biological Interpretation
cRegulon has demonstrated superior performance in identifying TF combinatorial modules compared to existing approaches. Validation using simulated datasets and real biological systems shows enhanced accuracy in both module identification and cell type annotation [12] [45]. When applied to a mixture of four cell lines, cRegulon successfully captured biologically relevant regulatory units specific to each cellular context.
Application of cRegulon to human fetal development data identified 25 distinct cRegulons associated with major developmental pathways [46]. These modules capture the coordinated action of TFs driving cell fate decisions during embryogenesis.
Table 2: Representative cRegulons from Human Fetal Atlas Analysis
| cRegulon | Top Transcription Factors | Biological Functions |
|---|---|---|
| M1 | FOXG1, RFX4, NEUROD2, NEUROG2, ASCL1, ESRRB | Neural progenitor state maintenance |
| M9 | HNF4A, HES1, PROX1, HNF1A, ONECUT1, GATA6, PDX1 | Pancreatic exocrine development |
| M12 | STAT5B, TAL1, GATA2, GATA1 | Hemopoietic lineage commitment |
| M15 | MYOG, TBX1, MYOD1, MYF6, MYF5 | Muscle structure development |
| M18 | CEBPA, XBP1, ONECUT2, HNF4A, ONECUT1, HNF1A | Lipid-glucose homeostasis |
The cRegulon framework provides unique insights into evo-devo by enabling comparative analysis of regulatory modules across species. For example, studies of Nodal signaling in amphioxus revealed how GRN rewiring through enhancer hijacking events led to the co-option of Gdf1/3-like for body axis patterning, replacing the ancestral Gdf1/3 function [44]. This evolutionary innovation was facilitated by the translocation of Gdf1/3-like to the Lefty genomic region, enabling shared regulatory control.
cRegulon analysis can detect such conserved combinatorial modules despite genomic rearrangements, revealing the evolutionary constraints on TF cooperation. The framework supports the hypothesis that developmental system drift often occurs through reorganization of regulatory connections while preserving core functional modules [44].
Table 3: Essential Research Reagents for cRegulon-based Studies
| Reagent/Method | Function in Analysis | Considerations |
|---|---|---|
| 10x Genomics Multiome | Provides paired scRNA-seq + scATAC-seq data | Enables direct correlation of chromatin accessibility and gene expression |
| SCENIC+ | Infers enhancer-driven gene regulatory networks | Provides input GRNs for cRegulon analysis [12] |
| LINGER | Lifelong learning approach for GRN inference | Incorporates atlas-scale external data as regularization [3] |
| cisTarget Databases | Species-specific motif collections | Essential for linking TFs to regulatory elements |
| CRISPR Perturbation | Functional validation of predicted TF modules | Establishes causal relationships in regulatory networks [47] |
For evolutionary developmental biologists, cRegulon offers a powerful approach to investigate how changes in combinatorial regulation drive phenotypic evolution. The methodology enables:
The framework is particularly valuable for studying developmental system drift, where similar developmental outcomes are achieved through different genetic pathways [44]. By decomposing GRNs into reusable cRegulon modules, researchers can trace how evolution tinkers with combinatorial partnerships to generate diversity while preserving essential functions.
cRegulon represents a significant advancement in GRN inference by explicitly modeling the combinatorial nature of transcriptional regulation. By identifying reusable regulatory modules that underpin cell type landscapes, the framework provides evolutionary developmental biologists with a powerful tool to decipher how changes in regulatory architecture drive phenotypic evolution.
The methodology's ability to integrate single-cell multi-omics data while accounting for cellular context makes it particularly valuable for studying the evolvability of developmental programs. As single-cell technologies continue to advance and atlas-scale projects expand, cRegulon analysis promises to reveal fundamental principles of how cooperative TF interactions constraint and facilitate evolutionary innovation.
Future developments may extend cRegulon to incorporate spatial transcriptomics data, enable cross-species module alignment, and integrate non-TF regulators such as signaling molecules and chromatin modifiers, further enhancing its utility for evolutionary developmental biology research.
The integration of spatial context with multimodal single-cell data is revolutionizing our understanding of Gene Regulatory Networks (GRNs) in evolutionary developmental biology. Frameworks such as PathOmCLIP and Nicheformer represent a paradigm shift in computational biology, enabling researchers to decipher the complex spatiotemporal regulatory logic that governs cell fate decisions, tissue patterning, and evolutionary adaptations. These approaches move beyond traditional single-modality analysis by harmonizing diverse data typesâincluding transcriptomics, epigenomics, proteomics, and high-resolution histologyâwithin their native spatial context. This integration is particularly crucial for GRN inference, as gene regulation is inherently spatial, with regulatory outcomes depending on precise cellular microenvironments and tissue organization.
PathOmCLIP introduces a novel approach to bridging histological imaging with spatial gene expression data through contrastive learning, allowing for the prediction of gene expression patterns directly from tissue morphology. Meanwhile, Nicheformer functions as a transformer-based foundation model trained on an unprecedented scale of spatially resolved cells, enabling the prediction of spatial context for dissociated cells and revealing spatial determinants of gene regulation. Together, these frameworks provide powerful new avenues for constructing more accurate, context-aware GRNs that reflect the true complexity of developmental processes and evolutionary trajectories across species.
Table 1: Core Framework Overview
| Framework | Primary Architecture | Key Innovation | Primary Data Modalities | Scale of Pretraining |
|---|---|---|---|---|
| PathOmCLIP | Contrastive learning model | Aligns histology images with spatial gene expression | Histology, spatial transcriptomics | Validation across 5 tumor types [11] |
| Nicheformer | Transformer-based foundation model | Predicts spatial context for dissociated cells | scRNA-seq, spatial transcriptomics | 57M dissociated + 53M spatially resolved cells [11] [48] |
PathOmCLIP (Pathology Omics Contrastive Language-Image Pretraining) employs a sophisticated multimodal learning approach to align histopathology images with spatial transcriptomic profiles. The methodology can be broken down into several key stages:
Data Acquisition and Preprocessing: Collect paired whole-slide histology images (typically H&E stained) and spatial transcriptomics data from the same tissue section. The spatial transcriptomics data provides gene expression measurements for specific locations (spots) on the tissue, each associated with spatial coordinates.
Image Patch Processing: Extract image patches centered on the spatial transcriptomics spots using the registered coordinates. Process these patches using a convolutional neural network (CNN) encoder to generate compact image feature embeddings.
Gene Expression Processing: The spatial gene expression vector for each spot is processed through a dedicated encoder (e.g., a multilayer perceptron or transformer) to generate gene expression embeddings.
Contrastive Learning Alignment: The core innovation of PathOmCLIP lies in its contrastive learning objective, which brings the image and gene expression embeddings of corresponding spots closer together in a shared latent space while pushing apart non-corresponding pairs.
Model Validation and Application: Validate the model by testing its ability to predict gene expression from histology images alone on held-out datasets. PathOmCLIP has been validated across five different tumor types, demonstrating robust cross-tissue performance [11].
PathOmCLIP Workflow: Illustrates the multimodal alignment of histology images and spatial gene expression.
PathOmCLIP enables several advanced applications critical for GRN inference in developmental biology:
Nicheformer is a transformer-based foundation model specifically designed for single-cell and spatial omics integration. Its protocol involves large-scale pretraining followed by task-specific fine-tuning:
Data Curation and Integration: Assemble a massive, diverse collection of single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics datasets. Nicheformer was pretrained on the largest collection to dateâover 57 million dissociated cells and 53 million spatially resolved cells across 73 human and mouse tissues [11] [48].
Foundation Model Pretraining: The model is pretrained on this colossal corpus using self-supervised learning objectives. This step allows the model to learn universal representations of cellular states and their potential spatial contexts.
Spatial Task Fine-Tuning: The pretrained model is subsequently fine-tuned on specific spatial tasks, such as spatial density prediction or niche and region label prediction. This adapts the general cellular representation to spatially explicit applications.
Spatial Context Prediction: A key application involves predicting the spatial context of dissociated cells. By inputting scRNA-seq data from dissociated cells, Nicheformer can infer their original spatial microenvironment, effectively transferring rich spatial information to vast scRNA-seq datasets.
Downstream Analysis: The model supports various downstream tasks, including spatial clustering, identification of spatially variable genes, and inference of cell-cell communication patterns within niches.
Nicheformer Workflow: Shows the process from large-scale pretraining to spatial context prediction.
Nicheformer provides several unique capabilities for enhancing GRN inference:
When evaluated on their respective tasks, both PathOmCLIP and Nicheformer demonstrate significant advancements over previous methods. PathOmCLIP shows high accuracy in connecting tumor histology with spatial gene expression across multiple cancer types [11]. Nicheformer consistently achieves top performance on novel spatial prediction problems, demonstrating the advantage of its transformer-based architecture and massive training scale [48].
Large-scale benchmarking efforts like HESCAPE (H&E + Spatial ContrAstive PrEtraining) are crucial for evaluating multimodal learning methods that leverage both histology images and gene expression data. Such benchmarks have revealed that effective integration requires carefully addressing technical challenges like batch effects, and that gene expression encoders pretrained on spatial transcriptomics data outperform those trained without spatial context [49].
Table 2: Functional Comparison and Applications
| Feature | PathOmCLIP | Nicheformer | Significance for GRN Inference |
|---|---|---|---|
| Primary Strength | Histology-gene expression alignment | Spatial context prediction for single cells | Links morphology to regulation; provides spatial priors |
| Key Innovation | Cross-modal contrastive learning | Massive-scale pretraining + fine-tuning | Enables novel data integration paradigms |
| Spatial Resolution | Spot-level (spatial transcriptomics) | Single-cell & spot-level | Enables fine-grained spatial regulatory analysis |
| Validation Scope | 5 tumor types [11] | 73 tissues across human & mouse [11] [48] | Demonstrates broad applicability across biological contexts |
| GRN Relevance | Identifies morphology-associated regulators | Reveals spatial determinants of regulation | Connects tissue patterning with transcriptional programs |
Successful implementation of spatial and multimodal integration methods requires specific computational tools and data resources. The following table outlines key components of the research toolkit for working with frameworks like PathOmCLIP and Nicheformer.
Table 3: Research Reagent Solutions for Spatial Multimodal Analysis
| Resource Category | Specific Examples | Function and Application |
|---|---|---|
| Spatial Transcriptomics Platforms | 10x Genomics Visium/Xenium, MERFISH, ISS | Generate paired histology and spatial gene expression data [50] [49] |
| Foundation Models | scGPT, scPlantFormer, Geneformer | Provide pretrained gene expression encoders for transfer learning [11] [49] |
| Histology Encoders | CTransPath, UNI, Phikon | Extract features from histology images for multimodal alignment [49] |
| Benchmarking Frameworks | HESCAPE, BioLLM | Standardized evaluation of multimodal integration methods [11] [49] |
| Data Repositories | DISCO, CZ CELLxGENE Discover | Provide access to millions of single-cell and spatial datasets for pretraining and analysis [11] |
| Multimodal Integration Tools | StabMap, TMO-Net, GIST | Enable integration of diverse omics modalities beyond transcriptomics [11] |
This protocol combines elements from PathOmCLIP and Nicheformer approaches to create a comprehensive workflow for spatial GRN inference in evolutionary developmental biology research.
Tissue Collection and Preservation: Collect developmental tissue samples at specific stages of interest. Immediately snap-freeze in liquid nitrogen or use appropriate fixation methods that preserve RNA quality while maintaining tissue architecture.
Spatial Multimodal Data Generation: Process tissue sections using compatible spatial transcriptomics platforms (e.g., 10x Genomics Xenium or Visium). For technologies requiring separate processing:
Single-Cell RNA Sequencing: For complementary dissociated single-cell analysis, prepare single-cell suspensions from adjacent tissue regions and perform scRNA-seq using standard protocols (10x Chromium, SMART-seq, etc.).
Initial Data Processing:
Multimodal Alignment with PathOmCLIP Framework:
Spatial Context Transfer with Nicheformer:
Spatially-Resolved GRN Construction:
Experimental Validation:
Downstream Analysis and Interpretation:
The integration of single-cell multi-omics data has revolutionized evolutionary developmental biology (Evo-Devo), enabling researchers to decipher the gene regulatory networks (GRNs) that underlie the emergence of novel traits and morphological diversity. Evo-Devo investigates how changes in embryonic development during single generations relate to evolutionary changes that occur between generations [52]. The field has uncovered that distantly related organisms share conserved genetic toolkits, such as Hox genes, which pattern the anterior-posterior axis in both fruit flies and mice, despite their divergent morphologies [53]. The completion of this workflow enables the identification of cell type-specific regulatory programs and the molecular mechanisms driving evolutionary innovations, such as the loss of eyes in cavefish or the diversification of cichlid jaw structures [54] [52].
This application note provides a detailed protocol for constructing GRNs from single-cell multiome data (paired gene expression and chromatin accessibility), framing the process within the context of Evo-Devo research. The workflow progresses from raw data preprocessing and dimensionality reduction to the final inference of regulatory networks, empowering researchers to explore the evolutionary forces that shape developmental programs.
The following workflow diagram outlines the end-to-end process for inferring gene regulatory networks from single-cell multi-omics data, highlighting the key computational stages.
Table 1: Essential reagents and computational tools for single-cell multi-omics GRN inference.
| Item Name | Function/Application | Specifications |
|---|---|---|
| 10x Genomics Multiome Kit | Simultaneous profiling of gene expression and chromatin accessibility in single cells | Enables paired scRNA-seq and scATAC-seq from the same cell |
| SHARE-Seq Protocol | Alternative method for co-profiling transcriptome and epigenome | Captures histone modifications or open chromatin with RNA |
| ENCODE Consortium Data | Atlas-scale external bulk data for regulatory element annotation | Provides diverse cellular contexts for prior knowledge integration |
| TF Motif Databases | Prior knowledge of transcription factor binding specificities | Enables linking accessible regions to potential regulators (e.g., JASPAR, CIS-BP) |
| ChIP-seq Validation Data | Ground truth data for benchmarking GRN predictions | Curated TF-target interactions for specific cell types |
| eQTL Datasets (GTEx/eQTLGen) | Independent validation of cis-regulatory relationships | Links genetic variants to gene expression changes |
Objective: To process raw sequencing data into high-quality count matrices for downstream analysis.
Protocol:
Objective: To project high-dimensional data into a low-dimensional space, enabling the identification of cell populations and states.
Protocol:
Objective: To infer directional regulatory interactions between transcription factors (TFs), regulatory elements (REs), and target genes (TGs).
Protocol:
Table 2: Benchmarking of GRN Inference Methods for Single-Cell Multi-omics Data. AUC = Area Under the Receiver Operating Characteristic Curve; AUPR = Area Under the Precision-Recall Curve.
| Method | Underlying Approach | Key Features | Reported Performance |
|---|---|---|---|
| LINGER [3] | Lifelong Neural Network | Incorporates external bulk data via lifelong learning; uses motif matching as manifold regularization | 4-7x relative increase in AUC vs. baselines; superior cis-regulatory AUC/AUPR across distance groups |
| PRISM-GRN [4] | Bayesian Model | Integrates prior known GRNs with multi-omics data in a probabilistic, biologically interpretable framework | Higher precision in sparse, imbalanced true networks; captures causality well |
| SCENIC+ [3] | Machine Learning | Infers cis-regulatory interactions and TF activations from multiome data | Baseline method for comparison |
| PCC [58] | Correlation (Pearson) | Simple, linear co-expression measure | Lower performance compared to nonlinear/multi-omic methods |
The following diagram illustrates the logical flow for diagnosing and resolving common issues in GRN inference.
The integration of GRN inference with Evo-Devo questions allows for a mechanistic understanding of evolutionary processes. The workflow below shows how to apply GRN inference to a classic Evo-Devo model.
Case Study: Unveiling the Regulatory Logic of Evolutionary Novelties
In single-cell multi-omics research, particularly in the context of Gene Regulatory Network (GRN) inference for evolutionary developmental biology (EvoDevo), two fundamental computational barriers persistently challenge researchers: data sparsity and the curse of dimensionality (COD). Data sparsity in single-cell RNA sequencing (scRNA-seq) arises from technical limitations where only a fraction (â¼1â60%, averaging <10%) of the transcriptome is detected in individual cells, creating numerous "dropout" events where transcripts are missed entirely [59]. Simultaneously, the curse of dimensionality manifests when analyzing high-dimensional data (typically >10,000 genes across thousands of cells), where accumulating technical noise causes detrimental effects including impaired distance metrics, inconsistent statistical results, and broken principal component structures [59] [60]. These challenges collectively obscure true biological signals, complicate the identification of cell types and states, and fundamentally limit our ability to reconstruct accurate GRNsâthe very blueprints that explain how inherited developmental programs generate phenotypic diversity across species [43].
For evolutionary developmental biologists, these technical challenges directly impede investigations into how developmental programs evolve through changes in gene expression and regulatory interactions [43]. Overcoming data sparsity and COD is therefore not merely a computational exercise but a prerequisite for understanding the molecular basis of phenotypic diversity.
The following table summarizes key computational methods designed to address data sparsity and the curse of dimensionality in single-cell multi-omics data, with particular relevance to GRN inference in evolutionary developmental biology.
Table 1: Computational Methods for Addressing Data Sparsity and Dimensionality Challenges
| Method | Primary Function | Key Innovation | Performance Advantages | Applicability to GRN Inference |
|---|---|---|---|---|
| RECODE [59] [60] | Noise reduction for high-dimensional data | Resolves COD without dimension reduction; parameter-free | Recovers expression of lowly expressed genes; enables precise cell fate transition delineation | Improves input data quality for any downstream GRN method |
| LINGER [3] | GRN inference from single-cell multiome data | Lifelong learning incorporating atlas-scale external bulk data | 4-7x relative increase in accuracy over existing methods; reveals complex GWAS regulatory landscapes | Directly infers trans-regulation (TF-TG), cis-regulation (RE-TG), and TF-binding (TF-RE) |
| GLUE [29] | Multi-omics data integration | Graph-linked unified embedding with explicit regulatory interactions | Superior performance in heterogeneous data; enables triple-omics integration; corrects previous atlas annotations | Provides integrated data for GRN inference; directly infers regulatory interactions |
| PRISM-GRN [4] | GRN recovery from multi-omics data | Bayesian model incorporating prior knowledge with biologically interpretable architecture | Higher precision in sparse regulatory interactions; captures causality; works with unpaired omics data | Specifically designed for cell type-specific GRN reconstruction |
| VIA [61] | Trajectory inference in single-cell omics | Lazy-teleporting random walks with MCMC refinement | Captures complex trajectories (cyclic, disconnected); scalable to >1 million cells; preserves global connectivity | Reveals developmental trajectories informing GRN dynamics |
Principle: RECODE (Resolution of the Curse of Dimensionality) employs high-dimensional statistical theory to separate technical noise from biological signal without dimension reduction, specifically tailored for scRNA-seq data with Unique Molecular Identifiers (UMIs) [59] [60].
Step-by-Step Procedure:
Validation Metrics:
Applications in EvoDevo: RECODE enables precise delineation of closely related cell types in developing tissues, identification of rare transitional cell states during organogenesis, and improved detection of subtle expression differences driving evolutionary innovations [59].
Principle: LINGER (Lifelong Neural Network for Gene Regulation) uses lifelong machine learning to incorporate atlas-scale external bulk data and TF motif knowledge as manifold regularization when inferring GRNs from single-cell multiome data [3].
Step-by-Step Procedure:
Model Architecture:
Training Protocol:
Regulatory Inference:
Validation:
Applications in EvoDevo: LINGER reveals evolutionary changes in GRN architecture underlying phenotypic differences between species, identifies key regulatory drivers in developmental pathways, and elucidates how noncoding variants associated with evolutionary innovations alter gene regulation [3].
Table 2: Key Research Reagents and Computational Resources for Single-Cell Multi-omics GRN Inference
| Resource Type | Specific Examples | Function in Research | Application in EvoDevo |
|---|---|---|---|
| Single-cell Multiome Kits | 10X Genomics Multiome ATAC + Gene Expression | Simultaneous measurement of chromatin accessibility and gene expression in same cell | Captures coordinated regulatory changes during evolution of developmental programs |
| Reference Datasets | ENCODE project bulk data [3]; GTEx eQTL data [3] | Provides external context for regulatory inference; validation of cis-regulatory predictions | Enables comparative analysis of regulatory elements across species |
| Prior Knowledge Bases | TF motif databases; curated ChIP-seq datasets [3] | Informs manifold regularization in GRN inference; provides ground truth for validation | Identifies conserved and diverged regulatory interactions in evolution |
| Benchmarking Resources | Gold-standard simultaneous omics datasets (SNARE-seq, SHARE-seq) [29] | Enables rigorous method validation and performance assessment | Establishes confidence in evolutionary inferences from computational predictions |
| Software Tools | LINGER, GLUE, RECODE, PRISM-GRN, VIA [3] [61] [59] | Provides specialized algorithms for each step in analytical pipeline | Enables comprehensive reconstruction of evolutionary changes in developmental GRNs |
Principle: Establish functional validation pipelines to test computational predictions of GRN changes underlying evolutionary innovations in developmental programs [43].
Step-by-Step Procedure:
Functional Testing:
Conservation Analysis:
Validation Metrics:
The integration of advanced computational methods like LINGER, RECODE, and GLUE represents a transformative approach to overcoming the fundamental challenges of data sparsity and dimensionality in single-cell multi-omics. By leveraging lifelong learning to incorporate external data, resolving COD without discarding biological information, and explicitly modeling regulatory interactions across omics layers, these methods enable unprecedented reconstruction of accurate, cell type-specific GRNs. For evolutionary developmental biology, these capabilities open new avenues for investigating how changes in developmental programs produce phenotypic diversity across speciesâaddressing questions that were previously intractable with conventional approaches. As single-cell technologies continue to advance, generating ever-larger and more complex datasets, the computational frameworks described here will play an increasingly central role in unlocking the regulatory logic of evolutionary innovation.
In the field of evolutionary developmental biology, the inference of Gene Regulatory Networks (GRNs) from single-cell multi-omics data represents a pivotal challenge for understanding the molecular mechanisms driving cellular differentiation and lineage specification. Single-cell sequencing technologies enable genome- and epigenome-wide profiling of thousands of individual cells, offering unprecedented biological insights into cellular heterogeneity [62] [31]. However, technical noise and batch effects obscure high-resolution structures, hindering rare-cell-type detection and cross-dataset comparisons essential for robust GRN inference [62]. These artifacts mask subtle biological signals, impair reproducibility, and limit the scope of downstream analyses, particularly when integrating data across multiple experiments or sequencing runs [63]. As the number of single-cell RNA sequencing (scRNA-seq) experiments grows, researchers increasingly combine results across experiments or process cells from the same experiment assayed in separate sequencing runs, gaining cellular throughput at the cost of introducing batch effects [63]. This application note provides a comprehensive framework for mitigating these technical challenges, enabling more accurate GRN inference in single-cell multi-omics studies of evolutionary developmental processes.
Technical noise in single-cell multi-omics data arises from multiple sources, including amplification bias, molecular dropout events, library preparation artifacts, and sequencing depth variations. Batch effects represent systematic technical variations between different experimental batches, sequencing runs, or platforms that are unrelated to biological signals. In the context of GRN inference, these artifacts profoundly impact the accurate reconstruction of regulatory relationships between transcription factors (TFs), cis-regulatory elements (CREs), and their target genes.
Single-cell data are particularly susceptible to these issues due to the inherent sparsity of measurements and the complex nature of multi-omics integration [64]. Technical noise can obscure the covariation patterns between TFs and their target genes that methods like correlation analysis, regression models, and deep learning approaches rely upon for GRN inference [31]. For instance, dropout events in scRNA-seq data can create false negatives in TF-target gene relationships, while batch effects can introduce spurious correlations that lead to incorrect regulatory edge predictions.
Recent benchmarking studies demonstrate that the process of batch correction itself can create measurable artifacts in the data if poorly calibrated [63]. Many published methods alter the data considerably during correction, potentially distorting legitimate biological signals along with technical noise. This highlights the critical need for carefully calibrated noise reduction and batch integration strategies that preserve biologically relevant information while removing technical artifacts.
A comprehensive evaluation of eight widely used batch correction methods for scRNA-seq data revealed significant variations in their performance and tendency to introduce artifacts [63]. The study employed a novel approach to measure the degree to which methods alter data during batch correction, examining both fine-scale distances between cells and cluster-level effects.
Table 1: Performance Comparison of scRNA-seq Batch Correction Methods
| Method | Performance Rating | Artifact Introduction | Recommended for GRN Studies |
|---|---|---|---|
| Harmony | Consistently performs well | Minimal artifacts | Yes |
| Combat | Performs poorly | Detectable artifacts | No |
| ComBat-seq | Performs poorly | Detectable artifacts | No |
| BBKNN | Performs poorly | Detectable artifacts | No |
| Seurat | Performs poorly | Detectable artifacts | No |
| MNN | Performs poorly | Considerable alteration | No |
| SCVI | Performs poorly | Considerable alteration | No |
| LIGER | Performs poorly | Considerable alteration | No |
Among the evaluated methods, Harmony was the only approach that consistently performed well across all testing methodologies without introducing detectable artifacts [63]. Methods including MNN, SCVI, and LIGER performed poorly, often altering the data considerably during the correction process. Combat, ComBat-seq, BBKNN, and Seurat introduced artifacts that could be detected in the testing setup, making them suboptimal choices for preparing data for GRN inference [63].
The RECODE platform represents a significant advancement in noise reduction for single-cell data, simultaneously addressing technical noise and batch effects while preserving full-dimensional data [62]. Unlike methods that rely on dimensionality reduction, RECODE maintains the complete feature space, enabling more accurate downstream analyses including GRN inference.
RECODE employs high-dimensional statistics and noise variance stabilizing normalization to comprehensively address technical noise across diverse single-cell modalities [62]. The platform's recent upgrade includes iRECODE, which specifically addresses batch effects while retaining the method's effectiveness for technical noise reduction. This dual capability makes it particularly valuable for cross-study integration in evolutionary developmental biology research, where datasets often originate from multiple laboratories and experimental conditions.
The algorithm underlying RECODE has undergone recent improvements that substantially enhance both accuracy and computational efficiency [62]. The platform has demonstrated effectiveness across multiple single-cell modalities, including single-cell high-throughput chromosome conformation capture (scHi-C) and spatial transcriptomics, making it suitable for comprehensive multi-omics GRN inference studies.
Principle: Harmony performs batch correction by iteratively clustering cells and correcting dataset-specific effects within each cluster, effectively integrating datasets while preserving biological heterogeneity.
Materials:
Procedure:
Harmony Integration:
Downstream Analysis:
Validation Metrics:
Principle: RECODE employs high-dimensional statistics to simultaneously reduce technical and batch noise while preserving full-dimensional data structure, enabling more accurate GRN inference from single-cell multi-omics data.
Materials:
Procedure:
RECODE Application:
Output Processing:
Integration with GRN Inference:
Principle: LINGER (Lifelong neural network for gene regulation) incorporates atlas-scale external bulk data across diverse cellular contexts and prior knowledge of transcription factor motifs as manifold regularization, significantly improving GRN inference accuracy from single-cell multiome data.
Materials:
Procedure:
Model Pre-training:
Model Refinement:
GRN Inference:
Validation:
Table 2: Key Research Reagent Solutions for Noise-Robust GRN Inference
| Item | Function/Application | Implementation Notes |
|---|---|---|
| Harmony Algorithm | Batch effect correction for scRNA-seq data | Only method consistently performing without artifacts in benchmarks [63] |
| RECODE/iRECODE Platform | Simultaneous technical and batch noise reduction | Preserves full-dimensional data; applicable to diverse single-cell modalities [62] |
| LINGER Framework | GRN inference with external data integration | Achieves 4-7x relative accuracy increase over existing methods [3] |
| ENCODE Bulk Data Resources | External reference for lifelong learning | Provides diverse cellular contexts for regularization [3] |
| TF Motif Databases | Prior knowledge for manifold regularization | Enriches TF-RE connections in regulatory modules [3] |
| ChIP-seq Validation Sets | Ground truth for trans-regulatory assessment | Essential for benchmarking GRN inference accuracy [3] |
| eQTL Resources (GTEx, eQTLGen) | Cis-regulatory validation | Enables assessment of RE-TG linkage accuracy [3] |
| CPI-637 | CPI-637, MF:C22H22N6O, MW:386.4 g/mol | Chemical Reagent |
| CPUY201112 | CPUY201112: Potent HSP90 Inhibitor for Research |
Effective mitigation of technical noise and batch effects is paramount for accurate GRN inference from single-cell multi-omics data in evolutionary developmental biology research. The integration of robust computational approachesâspecifically Harmony for batch correction, RECODE for comprehensive noise reduction, and LINGER for leveraging external data resourcesâenables researchers to overcome the significant challenges posed by technical variability. The protocols outlined in this application note provide a structured framework for implementing these advanced methods, facilitating more reliable identification of regulatory mechanisms driving developmental processes across diverse cellular contexts and experimental conditions. As single-cell technologies continue to evolve, these noise mitigation strategies will become increasingly essential for unlocking the full potential of multi-omics data in deciphering the complex regulatory networks that underlie evolutionary developmental processes.
Gene regulatory networks (GRNs) represent the complex web of interactions between transcription factors, cis-regulatory elements, and their target genes that control developmental processes and evolutionary changes [43] [65]. The emergence of single-cell multi-omics technologies has revolutionized evolutionary developmental biology (EvoDevo) by enabling simultaneous profiling of chromatin accessibility, gene expression, and epigenetic modifications in individual cells [66] [58]. However, the computational frameworks developed to reconstruct GRNs from these dataâparticularly deep learning modelsâincreasingly resemble "black boxes" that offer limited biological insight into their decision-making processes [67] [68] [69].
This model interpretability gap presents a significant challenge for researchers seeking to understand the molecular mechanisms underlying phenotypic evolution. As GRN inference methods grow in complexity, including transformer-based architectures and foundation models trained on millions of single-cell datasets [68], the need for Explainable AI (XAI) techniques becomes increasingly critical. This Application Note provides detailed protocols for applying SHAP and other XAI methods to enhance the biological interpretability of GRN models in evolutionary developmental studies, enabling researchers to bridge the gap between predictive accuracy and mechanistic understanding.
Gene regulatory networks represent the molecular infrastructure that transforms genotypic variations into phenotypic diversity through development. The GRN concept posits that developmental programs exhibit a fundamentally network-like structure, with genetically encoded components linked by recursive regulatory interactions [43]. From an evolutionary perspective, changes to GRN architectureâincluding gene duplications, cis-regulatory modifications, and network co-optionâdrive the emergence of novel traits across evolutionary timescales [44] [70].
In insects, for example, GRN evolution underpins diverse morphological adaptations. Studies in Drosophila species have demonstrated that changes in cis-regulatory regions can lead to new trait acquisition by altering network connectivity, while butterfly wing patterns emerge through co-option processes that reshape color pattern GRNs [70]. The Nodal signaling pathway governing body axis formation in deuterostomes represents another well-characterized example, where GRN rewiring through enhancer hijacking events has altered developmental programs in cephalochordate amphioxus while maintaining functional output [44].
Modern GRN inference leverages diverse mathematical frameworks to reconstruct regulatory relationships from single-cell multi-omic data [58]. The table below summarizes the primary computational approaches:
Table 1: Methodological Foundations for GRN Inference
| Approach | Underlying Principle | Strengths | Limitations |
|---|---|---|---|
| Correlation-based | Guilt-by-association through co-expression | Simple implementation; captures linear and non-linear associations | Cannot distinguish direct vs. indirect regulation; directionality challenges |
| Regression models | Predicts gene expression from TF expression/accessibility | Interpretable coefficients; handles multiple predictors | Overfitting with many predictors; correlated predictors cause instability |
| Probabilistic models | Graphical models capturing dependence between variables | Uncertainty quantification; handles noise effectively | Distributional assumptions may not hold for all genes |
| Dynamical systems | Models system behavior evolving over time | Captures temporal dynamics; interpretable parameters | Computationally intensive; requires time-series data |
| Deep learning | Artificial neural networks learning complex patterns | Captures non-linear interactions; high predictive power | "Black box" nature; extensive data requirements |
Single-cell multi-omics technologies have been particularly transformative for GRN inference, with methods like SHARE-seq and 10x Multiome enabling simultaneous profiling of chromatin accessibility and gene expression in individual cells [58]. These advances have spurred development of sophisticated GRN inference tools including SCENIC+ [65], HALO [66], and Pando [65] that integrate multiple data modalities to reconstruct enhancer-driven regulatory networks.
SHAP values originate from cooperative game theory and provide a unified approach to feature importance attribution by measuring the marginal contribution of each feature to the model's prediction across all possible feature combinations.
Protocol 3.1.1: SHAP Analysis for Transcription Factor Importance
Different GRN inference architectures require specialized XAI approaches:
Protocol 3.2.1: Attention Mechanism Analysis in Transformer Models
Protocol 3.2.2: Integrated Gradients for Deep Neural Networks
Evaluating XAI method effectiveness requires biological ground truth data:
Table 2: XAI Validation Framework for GRN Inference
| Validation Method | Application | Interpretation |
|---|---|---|
| Recovery of known regulatory interactions | Compare top explanations against literature-curated gold standards (e.g., TRRUST, RegNetwork) | Higher precision indicates better biological relevance |
| Enrichment of ChIP-seq supported interactions | Test if high-attribution TF-gene pairs show experimental binding support | Validates mechanistic plausibility of explanations |
| Stability across cell types | Assess consistency of explanations across related cell states | Robust explanations should show gradual changes along differentiation trajectories |
| Conservation across species | Compare explanatory features in orthologous GRNs | Evolutionarily conserved explanations suggest functional importance |
The following diagram illustrates the comprehensive workflow for applying XAI techniques to GRN inference in evolutionary developmental biology:
Diagram 1: Integrated XAI-GRN Workflow for EvoDevo Research
Table 3: Essential Research Reagents and Computational Tools
| Category | Specific Tools/Databases | Application in XAI-GRN Studies |
|---|---|---|
| GRN Inference Methods | SCENIC+ [65], HALO [66], Pando [65], CellOracle [65] | Base models for regulatory network reconstruction from single-cell multi-omics data |
| XAI Libraries | SHAP, Captum, LRP, Transformers Interpret | Model interpretation and feature importance attribution |
| Motif & Annotation Databases | CIS-BP, JASPAR, UniBind [65], pycisTarget [65] | TF-binding site information for validating regulatory predictions |
| Single-cell Multi-omics Platforms | 10x Genomics Multiome, SHARE-seq [58] | Experimental data generation for model training and validation |
| Validation Datasets | ENCODE ChIP-seq, SCREEN enhancer annotations [65] | Ground truth data for benchmarking XAI explanations |
| Specialized Architectures | MIDAA [71], Pathway-Guided Interpretable DL [69] | Knowledge-guided models with inherent interpretability features |
The Nodal signaling pathway governing body axis formation exhibits substantial GRN rewiring in cephalochordate amphioxus compared to other deuterostomes [44]. While the core function in dorsal-ventral and left-right patterning is conserved, the network architecture has undergone significant modification, including:
Protocol 6.2.1: Comparative GRN Analysis with SHAP
The following diagram illustrates the specific GRN rewiring in amphioxus and how XAI techniques pinpoint key evolutionary changes:
Diagram 2: XAI-Powered Analysis of GRN Rewiring in Amphioxus
The application of XAI techniques to single-cell multi-omics data presents significant computational challenges. For large-scale atlas studies encompassing millions of cells, we recommend:
Interpretable AI outputs must undergo rigorous biological validation:
Protocol 7.2.1: Functional Validation of XAI-Generated Hypotheses
The integration of Explainable AI techniques with single-cell multi-omic GRN inference represents a powerful approach for advancing evolutionary developmental biology. By applying the protocols and frameworks outlined in this Application Note, researchers can transform black-box predictions into testable biological hypotheses about the regulatory mechanisms underlying phenotypic diversity. As single-cell technologies continue to evolve and foundation models grow in sophistication [68], the principles of interpretability and biological validation will remain essential for extracting meaningful insights from complex computational models.
In evolutionary developmental biology (EvoDevo), the core objective is to unravel the molecular basis of phenotypic diversity by understanding the gene regulatory networks (GRNs) that control embryonic development [43]. A GRN is a network-like structure where genes (nodes) interact through their expressed products (edges) to control developmental programs [43]. The evolution of developmental programs occurs largely through changes in the connectivity and composition of these GRNs [43].
The advent of single-cell multi-omics technologies presents both an unprecedented opportunity and a significant challenge for GRN inference. Researchers can now simultaneously measure multiple molecular modalitiesâsuch as gene expression (scRNA-seq), chromatin accessibility (scATAC-seq), and DNA methylationâfrom individual cells [29]. This multi-dimensional view is essential because regulatory processes are too complex to reliably model with transcriptomic data alone [17]. However, this richness introduces the critical challenge of feature selection: identifying which molecular features among thousands are most biologically relevant for reconstructing accurate GRNs, particularly when these features interact through complex, non-linear relationships.
Effective feature selection is not merely a technical preprocessing step; it is foundational to generating biologically meaningful GRN models that can reveal how developmental programs evolve. This Application Note provides structured protocols and best practices to navigate this complex analytical landscape.
Feature selection techniques are broadly categorized into three groups, each with distinct advantages, limitations, and suitability for single-cell multi-omics data [72].
Table 1: Categories of Feature Selection Techniques
| Method Category | Core Principle | Key Advantages | Common Techniques | Suitability for Multi-omics GRN Inference |
|---|---|---|---|---|
| Filter Methods | Selects features based on statistical measures (e.g., correlation) independent of the model [72]. | ⢠Computationally fast and efficient⢠Model-agnostic⢠Easy to implement [72] | Correlation criteria, Chi-square tests [72] | Pre-filtering: Useful for initial, rapid reduction of feature space dimensionality before more sophisticated analysis. |
| Wrapper Methods | Uses a predictive model to evaluate different feature subsets based on their performance [72]. | ⢠Model-specific optimization⢠Can capture feature interactions [72] | Recursive Feature Elimination (RFE), Forward/Backward Selection [72] | Limited use: Often computationally prohibitive for the vast number of features in single-cell data. |
| Embedded Methods | Performs feature selection as an integral part of the model training process [72]. | ⢠Balances efficiency and effectiveness⢠Model-specific learning [72] | LASSO (â1-norm), Elastic Net, tree-based importance [72] [73] |
High: Efficiently handles high-dimensionality and can be tailored to specific GRN inference algorithms. |
For single-cell multi-omics, embedded methods are often most practical due to their ability to handle high dimensionality without the computational burden of wrapper methods. Furthermore, specialized non-linear feature selection networks have been developed that use â2,p-norm regularization within neural networks to select discriminative feature subsets while capturing non-linear structure, which is crucial for complex biological data [73].
The following protocols outline a streamlined workflow for feature selection and GRN inference in a single-cell multi-omics context.
This protocol uses SCENIC++, a widely adopted tool for inferring GRNs from single-cell transcriptomics data [17].
1. Input Data Preparation:
loom file object for use with SCENIC.2. Co-expression Module Inference:
3. Regulon Construction & Scoring:
4. Output & Analysis:
Diagram 1: SCENIC workflow for GRN inference from scRNA-seq.
For a more robust GRN inference that incorporates multiple data modalities, the GLUE (Graph-Linked Unified Embedding) framework is recommended [29].
1. Data & Guidance Graph Preparation:
2. Model Initialization & Training:
3. Iterative Optimization & Inference:
4. Batch Effect Correction (Optional):
Diagram 2: Multi-omics GRN inference with GLUE framework.
Table 2: Key Research Reagent Solutions for Single-Cell Multi-Omics
| Item / Tool Name | Function / Application | Relevant Assay |
|---|---|---|
| 10X Multiome Kit | Enables simultaneous measurement of gene expression and chromatin accessibility from the same single cell. | scRNA-seq + scATAC-seq [29] |
| SHARE-Seq | A high-throughput method for jointly profiling gene expression and chromatin accessibility. | scRNA-seq + scATAC-seq [29] |
| snmC-seq | Measures DNA methylation at single-cell resolution. | scMethyl-seq [29] |
| CisTarget Databases | Databases of evolutionarily conserved transcription factor binding motifs used for regulon refinement. | SCENIC/SCENIC+ [17] |
| GLUE Framework | A modular computational framework for integrating unpaired multi-omics data and inferring regulatory interactions. | Multi-omics Integration & GRN Inference [29] |
| Crenigacestat | Crenigacestat, CAS:1421438-81-4, MF:C22H23F3N4O4, MW:464.4 g/mol | Chemical Reagent |
Table 3: Computational Tools for GRN Inference and Feature Selection
| Tool Name | Primary Input Data | Feature Selection & Modelling Approach | Key Feature |
|---|---|---|---|
| SCENIC+ | scRNA-seq (and integrated scATAC-seq) | Linear modeling; signed, weighted interactions [17] | Infers enhancer-driven gene regulatory networks. |
| GLUE | Unpaired multi-omics (e.g., RNA, ATAC) | Non-linear; weighted interactions; uses a guidance graph [29] [17] | Explicitly models interactions across omics layers. |
| CellOracle | scRNA-seq (and unpaired scATAC-seq) | Linear; signed, weighted interactions [17] | Models the effect of in-silico perturbations on cell identity. |
| Pando | Paired or integrated multi-omics | Linear or non-linear; signed, weighted interactions [17] | Utilizes a flexible generalized linear model framework. |
| NFSN | General feature selection | Non-linear neural network with â2,p-norm regularization [73] |
Jointly performs non-linear feature selection and regression. |
In the context of EvoDevo, where the goal is to trace the evolutionary history and rewiring of GRNsâas exemplified by the shift in the Nodal signaling network in amphioxus [44]ârigorous feature selection is not optional. It is the critical step that determines whether inferred networks reflect biological reality or technical artifact.
The protocols and tools outlined here provide a pathway to robust GRN inference. Starting with a well-defined biological question, researchers should:
Inference of Gene Regulatory Networks (GRNs) from single-cell multi-omics data represents a cornerstone of modern evolutionary developmental biology (evo-devo) research, enabling the dissection of molecular circuits that orchestrate cell fate decisions, differentiation trajectories, and evolutionary adaptations. However, this field grapples with significant methodological limitations that compromise biological validity. High rates of false positives and negatives obscure true regulatory interactions, while scalability issues hinder application to the massive datasets now generated. Furthermore, a critical gap exists in dynamic network inference across pseudo-temporal trajectories, which is essential for understanding developmental processes. This Application Note synthesizes recent computational advances that directly address these bottlenecks, providing validated protocols and reagent solutions to empower researchers in constructing more accurate, scalable, and dynamic GRN models from complex single-cell data.
The performance of GRN inference methods is typically evaluated using Area Under the Receiver Operating Characteristic Curve (AUROC) and Area Under the Precision-Recall Curve (AUPRC). The following table summarizes the reported performance of several recently developed methods on benchmark datasets.
Table 1: Performance Metrics of Recent GRN Inference Methods
| Method | Core Innovation | Reported Performance (AUROC/AUPRC) | Key Application Context |
|---|---|---|---|
| inferCSN [74] | Cell state-specific networks via pseudo-temporal windowing | Outperformed GENIE3, SINCERITIES, PPCOR, and LEAP on simulated datasets (BF, CY, LI, LL) | Robust to dataset type and scale; ideal for analyzing T-cell states and tumor subclones |
| DAZZLE [75] | Dropout Augmentation for model regularization | Shows improved performance and increased stability over baseline models (e.g., DeepSEM) | Handles zero-inflation in large datasets (>15,000 genes) with minimal gene filtration |
| MultiGNN [76] | Supervised GNN using multi-omics features | Consistently outperformed other methods across seven different datasets | Effectively integrates scRNA-seq and scATAC-seq to reduce false positives |
| scMKL [21] | Multiple Kernel Learning for interpretable integration | Statistically significant (p<0.001) superior AUROC vs. MLP, XGBoost, SVM on multiple cancer datasets | Identifies key pathways and TF binding sites in breast, prostate, and lung cancer |
This protocol details the procedure for inferring gene regulatory networks that are specific to cell type and state using single-cell RNA-seq data, based on the inferCSN method [74].
Primary Application in Evo-Devo: Uncovering dynamic rewiring of regulatory networks across developmental pseudo-time.
Input Requirements:
Procedure:
Troubleshooting Tip: If the pseudo-time trajectory is inaccurate, the resulting state-specific networks will be confounded. Validate the trajectory using known marker genes.
This protocol describes a supervised learning framework, MultiGNN, that integrates scRNA-seq and scATAC-seq data to infer GRNs with improved distinction between direct and indirect regulations [76].
Primary Application in Evo-Devo: Identifying conserved, direct transcriptional regulation across species by integrating chromatin accessibility.
Input Requirements:
Procedure:
Troubleshooting Tip: The performance of MultiGNN is dependent on the quality of the prior network. Use the most context-specific prior available for your biological system.
This protocol utilizes DAZZLE, a method that enhances GRN inference robustness against the high dropout (zero-inflation) noise characteristic of scRNA-seq data [75].
Primary Application in Evo-Devo: Achieving reliable network inference from datasets with challenging signal-to-noise ratios, such as those from early embryonic stages or rare cell types.
Input Requirements:
Procedure:
Troubleshooting Tip: The proportion of values to augment with zeros is a key parameter. Start with a low value (e.g., 1-5%) and adjust based on the observed dropout rate in your data.
Table 2: Essential Computational Tools and Resources for Single-Cell Multi-omics GRN Inference
| Resource Name | Type | Primary Function in GRN Inference | Relevant Citation |
|---|---|---|---|
| inferCSN | Software Package | Infers cell type and state-specific GRNs from scRNA-seq using pseudo-temporal windowing. | [74] |
| DAZZLE | Software Package | Infers GRNs with improved robustness to scRNA-seq dropout noise via Dropout Augmentation. | [75] |
| MultiGNN | Software Framework | A supervised Graph Neural Network that integrates scRNA-seq and scATAC-seq for GRN inference. | [76] |
| scMKL | Software Package | Provides interpretable integration of multi-omics data for classification and feature analysis. | [21] |
| CZ CELLxGENE / DISCO | Data Repository | Provides curated, massive-scale single-cell datasets for model training and validation. | [11] |
| BEELINE | Benchmarking Framework | Provides standardized datasets and evaluation protocols for benchmarking GRN inference algorithms. | [75] [76] |
| JASPAR / Cistrome | Prior Knowledge Database | Curated transcription factor binding motifs and chromatin profiles for network calibration. | [21] |
A critical application of state-specific GRNs is the identification of active signaling pathways driving developmental transitions. The diagram below illustrates a generalized workflow for processing a multi-omic dataset to reveal a key pathway, such as Wnt signaling, active in a specific cell state.
Workflow Description: Starting with multi-omics data (scRNA-seq and scATAC-seq), a state-specific GRN is inferred for a window of cells along a pseudo-time trajectory (e.g., a progenitor state). Comparative analysis of GRNs across different states (e.g., progenitor vs. differentiated) reveals a sub-network of TFs and target genes that are dynamically rewired. This sub-network, when mapped to prior knowledge (e.g., KEGG, WikiPathways), can be annotated as a specific signaling pathway, such as Wnt. The activity of this pathway, supported by both expression and chromatin accessibility of key genes, provides a testable mechanistic hypothesis for its role in driving the state transition, which can be validated experimentally or through in silico perturbation modeling [11].
Gene Regulatory Network (GRN) inference is a central problem in evolutionary developmental biology and single-cell multi-omics research. The accuracy of inferred regulatory interactions depends critically on robust validation datasets that provide experimental evidence of true molecular relationships. Gold-standard datasets derived from Chromatin Immunoprecipitation followed by sequencing (ChIP-seq), expression Quantitative Trait Loci (eQTL) studies, and genetic perturbation experiments provide essential ground truth for benchmarking computational methods. These resources enable researchers to validate predicted transcription factor (TF)-target gene relationships, enhancer-promoter connections, and causal regulatory interactions, thereby ensuring biological relevance in network models. The integration of these validation frameworks is particularly crucial for studying the evolution of developmental programs, where rewiring of GRNs underlies phenotypic diversity.
The table below summarizes the primary categories of gold-standard datasets used for validating GRN inferences, their specific applications, and key benchmarks for performance evaluation.
Table 1: Gold-Standard Dataset Categories for GRN Validation
| Dataset Type | Biological Question Addressed | Validation Metrics | Reported Performance Benchmarks |
|---|---|---|---|
| ChIP-seq TF Binding | Direct physical TF-DNA interaction | Precision, Recall, F1 score, Area Under ROC Curve (AUC) | Significantly enriched for binding sites (p < 0.05) [77] |
| eQTL Mapping | Genotype-driven expression variation | Area Under Precision-Recall Curve (AUPR) ratio, AUC | Higher AUC/AUPR across different RE-TG distance groups [3] |
| Perturbation Data | Causal regulatory relationships | Concordance with known biological processes | Fourfold to sevenfold relative increase in inference accuracy [3] |
| Chromatin Interaction | Enhancer-target gene assignments | Gene Set Enrichment (GSE) testing concordance | Top EnTDefs significantly outperform nearest-gene approach [78] |
Large-scale ChIP-seq provides the most direct evidence of transcription factor binding and serves as a critical benchmark for evaluating predicted cis-regulatory elements. The following protocol outlines the methodology used to generate a comprehensive ChIP-seq resource in maize:
This protocol generated 2,147,366 reproducible TF-binding peaks, with a median of 16,000 binding sites per TF, which were used to validate the completeness and precision of integrated cis-regulatory elements (iCREs) [77]. Performance benchmarking demonstrated that predictions from all tested CRE identification methods were significantly enriched (p < 0.05) for the binding sites in the ChIP-seq dataset, whereas repeats and transposable elements were depleted [77].
Figure 1: ChIP-seq Experimental Workflow for GRN Validation
Table 2: Essential Research Reagents for ChIP-seq Experiments
| Reagent/Resource | Function | Specific Example |
|---|---|---|
| Avi Epitope Tag | Enables biotinylation for highly specific pull-down | Maize TFs fused to Avi tag [79] |
| Biotin Ligase | Enzymatically biotinylates Avi-tagged TFs | Co-expressed with TF-Avi fusion [79] |
| Streptavidin Magnetic Beads | Capture biotinylated TF-DNA complexes | High signal-to-noise ratio ChIP [79] |
| Hyper-stable Tn5 Transposase | Efficient tagmentation for library prep | Fused to E. coli elongation factor Ts [79] |
| ChIP-seq Gold Standard | Benchmarking CRE predictions | 104 TF ChIP-seq dataset from maize leaf [77] [79] |
Expression Quantitative Trait Loci (eQTL) mapping provides evidence for genetic variants that influence gene expression, serving as a valuable validation resource for predicted enhancer-target gene relationships. The following methodology outlines the application of eQTL data for benchmarking GRN inferences:
This validation approach is particularly valuable for assessing cis-regulatory predictions in single-cell multi-omics GRN inference methods, where linking distal regulatory elements to their target genes remains challenging. The method provides evidence that regulatory predictions align with natural genetic variation that affects gene expression, strengthening their biological relevance.
Genetic perturbation experiments provide causal evidence for regulatory relationships, serving as the highest standard for validating predicted GRN interactions. The following approaches are used to generate and utilize perturbation data:
Perturbation data is especially valuable in evolutionary developmental biology studies, where it can reveal how GRNs have been rewired during evolution. For example, functional genetic evidence in amphioxus demonstrated that a pivotal enhancer hijacking event allowed the emergence of a new GRN for body axis formation, highlighting the mechanistic basis of developmental system drift [44].
Figure 2: Perturbation Data for Causal GRN Validation
Comprehensive validation of GRN inferences requires integrated frameworks that combine multiple gold-standard datasets. The following protocol outlines a systematic approach for benchmarking method performance:
This integrated approach enables rigorous evaluation of GRN inference methods. For example, the LINGER method demonstrated a fourfold to sevenfold relative increase in accuracy over existing approaches when validated against such comprehensive benchmarks [3].
Table 3: Essential Resources for Multi-omics GRN Validation
| Resource Category | Specific Examples | Application in Validation |
|---|---|---|
| Reference Datasets | ENCODE ChIP-seq, GTEx eQTL, FANTOM5 | Benchmarking enhancer-target predictions [3] [78] |
| Software Tools | Seurat, MOFA+, Harmony | Data integration and batch correction [81] [80] |
| GRN Inference Methods | LINGER, PRISM-GRN, scSAGRN | Comparative performance benchmarking [3] [4] [37] |
| Experimental Validation Platforms | Perturb-seq, CROP-seq | Functional confirmation of predictions [80] |
Gold-standard validation datasets from ChIP-seq, eQTL studies, and perturbation experiments provide essential foundations for benchmarking GRN inferences in evolutionary developmental biology research. The integration of these complementary data types enables comprehensive evaluation of both physical binding associations and functional regulatory relationships. As single-cell multi-omics technologies continue to advance, these validation frameworks will become increasingly important for distinguishing true biological signals from computational artifacts, ultimately leading to more accurate models of gene regulatory network evolution and function. Researchers should prioritize methods that demonstrate robust performance across multiple validation datasets rather than optimizing for any single metric, thereby ensuring biological relevance and translational potential in both basic research and drug development applications.
In the field of single-cell multi-omics research, accurately inferring gene regulatory networks (GRNs) is fundamental to understanding the molecular mechanisms governing evolutionary developmental biology. GRNs are collections of molecular regulators, particularly transcription factors (TFs), regulatory elements (REs), and target genes (TGs), that interact to determine gene activation and silencing in specific cellular contexts [3] [31]. The evaluation of computational methods that reconstruct these networks presents significant challenges due to the complexity of biological systems and limitations in establishing complete ground truth data. Performance metrics such as the Area Under the Receiver Operating Characteristic Curve (AUC) and the Area Under the Precision-Recall Curve (AUPR) have emerged as standard quantitative measures for benchmarking GRN inference accuracy against experimentally validated regulatory interactions [3] [82]. These metrics provide researchers, scientists, and drug development professionals with crucial tools for assessing method reliability before applying them to investigate developmental processes or identify therapeutic targets.
The Area Under the Receiver Operating Characteristic Curve (AUC) and Area Under the Precision-Recall Curve (AUPR) are ranking-based metrics that evaluate the ability of GRN inference methods to distinguish true regulatory interactions from non-interactions. AUC measures the overall performance across all possible classification thresholds, plotting the True Positive Rate (sensitivity) against the False Positive Rate (1-specificity). A perfect classifier achieves an AUC of 1, while random guessing yields 0.5 [3] [82]. AUPR focuses specifically on the precision-recall relationship, making it particularly valuable for evaluating performance on imbalanced datasets where true positives are rare compared to the vast number of potential false positives in GRN inference [82]. The AUPR ratio, which compares the AUPR of a method to that of a random predictor, provides a standardized measure of improvement over chance [3].
In practical applications, these metrics are calculated by comparing inferred regulatory strengths against known ground truth networks. For example, LINGER demonstrated a fourfold to sevenfold relative increase in accuracy over existing methods when evaluated using these metrics [3]. The regulatory strength of transcription factor-target gene (TF-TG) pairs is typically ranked, and the resulting ordered list is assessed against confirmed interactions. AUC values quantify how well the method separates true regulators from non-regulators across all thresholds, while AUPR emphasizes the method's performance in identifying true positives despite the overwhelming number of potential false positives inherent in GRN inference tasks [3] [82].
Table 1: Key Characteristics of AUC and AUPR Metrics
| Metric | Interpretation | Strength | Limitation | Ideal Use Case |
|---|---|---|---|---|
| AUC | Probability that a random true positive is ranked higher than a random true negative | Robust summary of overall performance across thresholds | Less informative for highly imbalanced data | Initial method comparison when false positive rate matters |
| AUPR | Relationship between precision and recall at different thresholds | More informative for imbalanced datasets where positives are rare | More difficult to interpret in isolation | Focused evaluation of positive prediction accuracy |
| AUPR Ratio | Ratio of method AUPR to random predictor AUPR | Standardized measure of improvement over chance | Depends on the specific dataset composition | Normalized comparison across different datasets |
Establishing reliable ground truth data represents a fundamental challenge in GRN inference benchmarking. A comprehensive evaluation requires multiple forms of experimental evidence, each with distinct strengths and limitations for validation purposes [82].
Table 2: Categories of Ground Truth Data for GRN Validation
| Ground Truth Category | Description | Examples | Strengths | Limitations |
|---|---|---|---|---|
| Direct TF-TG Binding | Evidence of physical binding between TF and DNA | ChIP-seq, ChIP-ATLAS [3] [83] | Direct evidence of regulatory potential | Does not confirm functional regulation; cell-type specificity challenges |
| Cis-regulatory Validation | Links between regulatory elements and target genes | eQTL studies (GTEx, eQTLGen) [3] | Provides functional evidence of regulation | Limited by statistical power and tissue/cell type availability |
| Model Organism Databases | Curated regulatory interactions from well-studied organisms | RegulonDB [82] | High-quality, curated data | Limited transferability to human and other complex eukaryotes |
| Genetic Perturbation | Evidence from targeted genetic manipulations | Knockout, knockdown, overexpression [82] | Establishes causal relationships | Technically challenging and low-throughput |
In practice, benchmarking studies typically collect multiple ground truth datasets relevant to the biological context. For example, a study of blood cells might collect 20 ChIP-seq datasets from relevant cell types as ground truth for trans-regulatory interactions [3]. Similarly, eQTL data from GTEx and eQTLGen can validate cis-regulatory predictions, with performance assessed across different genomic distance groups between REs and TGs [3]. The selection of appropriate ground truth data requires careful consideration of cellular context, technical quality, and relevance to the biological questions being investigated.
This protocol details the evaluation of trans-regulatory predictions (TF-TG interactions) against transcription factor binding data from ChIP-seq experiments.
Materials and Reagents:
Procedure:
Metric Calculation:
Statistical Validation:
Troubleshooting Tip: Ensure consistent gene annotation between predicted networks and ground truth data, as identifier mismatches can artificially deflate performance metrics.
This protocol describes the evaluation of cis-regulatory interactions (RE-TG) using expression quantitative trait loci (eQTL) data as ground truth.
Materials and Reagents:
Procedure:
Performance Assessment:
Result Interpretation:
Figure 1: Workflow for comprehensive benchmarking of GRN inference methods, highlighting the sequential process from data selection to final conclusion.
The LINGER (Lifelong neural network for gene regulation) framework provides an illustrative case study for comprehensive GRN evaluation. When benchmarked on peripheral blood mononuclear cell (PBMC) data, LINGER demonstrated a fourfold to sevenfold relative increase in accuracy compared to existing methods including standard neural networks (scNN), bulk data-trained models (BulkNN), and correlation-based approaches (PCC) [3].
For trans-regulatory validation, researchers collected 20 ChIP-seq datasets from blood cells as ground truth. LINGER achieved significantly higher AUC and AUPR ratio values compared to other methods, indicating superior performance in identifying true TF-target relationships [3]. For cis-regulatory validation, the method was evaluated using eQTL data from GTEx and eQTLGen, with performance assessed across different genomic distance groups. LINGER maintained higher AUC and AUPR ratios than scNN across all distance categories, demonstrating robust identification of functional RE-TG connections [3].
This benchmarking approach exemplifies rigorous methodology: multiple ground truth sources, comparison against relevant baseline methods, and stratified analysis to understand performance characteristics across different regulatory contexts.
Table 3: Key Research Reagent Solutions for GRN Benchmarking Studies
| Reagent/Resource | Function in GRN Evaluation | Example Sources | Application Notes |
|---|---|---|---|
| ChIP-seq Datasets | Ground truth for direct TF-DNA binding | ChIP-Atlas, ENCODE [3] [83] | Select cell-type relevant datasets; requires careful processing |
| eQTL Resources | Validation of cis-regulatory relationships | GTEx, eQTLGen [3] | Use significant variant-gene pairs at appropriate FDR thresholds |
| Reference Networks | Curated regulatory interactions from model organisms | RegulonDB, DREAM challenges [82] | Useful for initial method development but limited transferability |
| Single-cell Multiome Data | Paired scRNA-seq + scATAC-seq for inference | 10x Multiome, SHARE-seq [3] [84] | Enables simultaneous profiling of gene expression and chromatin accessibility |
| Benchmarking Platforms | Standardized evaluation frameworks | BEELINE [83] | Provides synthetic and real networks for controlled comparisons |
Single-cell sequencing data presents unique challenges for GRN inference evaluation, including technical noise, sparsity (dropouts), and limited independent data points despite large cell numbers [3] [82]. These characteristics can significantly impact benchmarking results by introducing false negatives and reducing statistical power. Evaluation frameworks must account for these data-specific properties when interpreting performance metrics [82]. Methods that incorporate external data sources, such as LINGER's use of atlas-scale bulk data, can help mitigate these limitations [3].
Recent advances in GRN inference include deep learning approaches that leverage graph-based structures to capture global regulatory information [83], disentangled generative models that separate shared and private features across omics layers [84], and methods that explicitly model combinatorial regulation through cRegulons [12]. As these sophisticated approaches emerge, evaluation frameworks must similarly evolve to capture their specific capabilities, such as identifying cooperative TF interactions or predicting context-specific regulatory activities. Future benchmarking efforts may need to incorporate additional metrics beyond AUC and AUPR to fully characterize method performance on these advanced tasks.
Figure 2: Hierarchy of evaluation framework components showing the relationship between ground truth data types and performance metrics in GRN inference benchmarking.
Rigorous evaluation of GRN inference methods using AUC, AUPR, and comprehensive ground truth comparisons is essential for advancing single-cell multi-omics research in evolutionary developmental biology. The protocols and frameworks presented here provide researchers with standardized approaches for assessing method performance, enabling informed selection of appropriate computational tools for investigating gene regulatory mechanisms in development and disease. As the field continues to evolve with emerging technologies and more sophisticated algorithms, these evaluation principles will remain fundamental for validating biological discoveries and ensuring computational reproducibility.
In the field of evolutionary developmental biology, understanding the gene regulatory networks (GRNs) that govern cell identity and fate is paramount. Single-cell multi-omics technologies now enable the joint profiling of chromatin accessibility and gene expression in individual cells, providing an unprecedented opportunity to decipher enhancer-driven GRNs that underlie developmental processes [65]. The computational inference of these networks is a central problem in biology, with implications for understanding evolution, development, and disease mechanisms [3].
Several advanced computational methods have emerged to leverage single-cell multi-omics data for GRN inference. This application note provides a comparative analysis of three leading toolsâLINGER, SCENIC+, and cRegulonâframed within the context of single-cell multi-omics GRN inference for evolutionary developmental biology research. We evaluate their methodologies, performance, and practical applications to guide researchers and drug development professionals in selecting appropriate tools for their specific research needs.
SCENIC+ is a comprehensive computational framework designed specifically for inferring enhancer-driven GRNs from single-cell multi-omics data. The method predicts genomic enhancers alongside candidate upstream transcription factors (TFs) and links these enhancers to candidate target genes [65].
Workflow: SCENIC+ implements a three-step workflow:
Key Output: The output is a set of enhancer-driven regulons (eRegulons), each comprising a TF with its set of target regions and genes, which together form the complete eGRN [65].
LINGER (Lifelong neural network for gene regulation) represents a paradigm shift in GRN inference by incorporating atlas-scale external bulk data across diverse cellular contexts as a manifold regularization. This approach addresses the challenge of learning complex regulatory mechanisms from limited independent data points in single-cell experiments [3].
Workflow: LINGER employs a three-step process centered on lifelong machine learning:
Key Advantage: This unique approach allows LINGER to achieve a fourfold to sevenfold relative increase in accuracy over existing methods, as validated by ChIP-seq data and expression quantitative trait loci (eQTL) studies [3].
cRegulon addresses a critical limitation in existing GRN inference methods by modeling combinatorial regulation of transcription factors. While methods like SCENIC+ infer regulons for single TFs, cRegulon identifies regulatory modules where multiple TFs work together to regulate common target genes, which is fundamental for understanding cell fate decisions [12].
The following diagram illustrates the core methodological differences and shared workflow stages among SCENIC+, LINGER, and cRegulon:
Independent benchmarking studies provide critical insights into the performance of GRN inference methods. The table below summarizes key performance characteristics of SCENIC+, LINGER, and cRegulon based on published evaluations.
Table 1: Performance Comparison of GRN Inference Tools
| Metric | SCENIC+ | LINGER | cRegulon |
|---|---|---|---|
| Accuracy Improvement | Outperforms CellOracle, Pando, FigR, GRaNIE, and SCENIC in TF and target region prediction [65] | 4-7x relative increase in accuracy over existing methods [3] | Outperforms existing approaches in identifying TF combinatorial modules [12] |
| TF Recovery | Best recovery of differentially expressed TFs and TFs with direct ChIP-seq peaks [65] | High recovery of biologically relevant TFs validated by ChIP-seq [3] | Specialized in identifying cooperative TF pairs with validated biological relevance [12] |
| Precision/Recall | High precision and recall for predicted target regions based on ChIP-seq validation [65] | Significantly higher AUC and AUPR ratio across validation datasets [3] | Precise capture of known cooperative TF interactions [12] |
| Experimental Validation | Strong overlap with EBF1, PAX5, and POU2F2 ChIP-seq data in PBMCs; high enhancer activity by STARR-seq [65] | Consistency with eQTL data from GTEx and eQTLGen; validation against ChIP-seq ground truth [3] | Validation on in-silico data and real cell line mixtures confirming biological properties [12] |
| Scalability | 1h-44h running time and 21GB-461GB memory for tested datasets [65] | Efficient utilization of large-scale external data improves learning from limited single-cell data [3] | Designed to reduce GRN complexity through modularity, enabling genome-scale analysis [12] |
Table 2: Methodological Approaches of GRN Inference Tools
| Feature | SCENIC+ | LINGER | cRegulon |
|---|---|---|---|
| Primary Innovation | Large motif collection (32,765 motifs) and enhancer-focused GRNs [65] | Lifelong learning incorporating external bulk data [3] | Combinatorial TF module identification [12] |
| Data Requirements | Paired scRNA-seq + scATAC-seq (can work with integrated data) [17] | Paired scRNA-seq + scATAC-seq with cell type annotations [3] | Paired or unpaired scRNA-seq + scATAC-seq [12] |
| Regulatory Unit | eRegulon (single TF + enhancers + target genes) [65] | Triple interactions (TF-TG, RE-TG, TF-RE) [3] | cRegulon (TF modules + REs + target genes) [12] |
| Modeling Approach | Linear modeling with frequentist framework [17] | Neural network with Bayesian-inspired refinement [3] | Matrix optimization and modularity analysis [12] |
| Key Applications | Master regulator identification; conserved regulation across species; differentiation trajectories [65] | Disease-relevant cell type and driver regulator identification; GWAS interpretation [3] | Cell type annotation; developmental process analysis; cooperative TF identification [12] |
Purpose: To identify enhancer-driven gene regulatory networks governing cell fate decisions in developing tissues.
Materials:
Procedure:
Motif Enrichment Analysis:
Region-to-Gene Linking:
Downstream Analysis:
Validation: Compare predicted target regions with available ChIP-seq data and assess enhancer activity using STARR-seq data [65].
Purpose: To infer GRNs from single-cell multiome data using atlas-scale external data for enhanced accuracy, particularly for disease applications.
Materials:
Procedure:
Model Pre-training:
Lifelong Learning Refinement:
GRN Extraction:
Disease Application:
Validation: Validate trans-regulatory predictions against ChIP-seq data and cis-regulatory predictions against eQTL data from GTEx and eQTLGen [3].
Purpose: To identify combinatorial TF modules that co-regulate target genes and define cell identities.
Materials:
Procedure:
Combinatorial Effect Calculation:
TF Module Identification:
cRegulon Construction:
Biological Interpretation:
Validation: Apply to in-silico simulated data and real cell line mixtures to validate the biological properties of identified regulatory units [12].
Table 3: Key Research Reagents for Single-Cell Multi-omics GRN Inference
| Reagent/Resource | Function | Example Applications |
|---|---|---|
| 10x Genomics Single-Cell Multiome Kit | Simultaneous measurement of gene expression and chromatin accessibility in the same single cell | Generating input data for all three tools; PBMC analysis for benchmarking [3] |
| ENCODE Project Data | Comprehensive collection of functional genomic datasets across diverse cell types | External bulk data for pre-training LINGER; validation of predictions [3] |
| CisTarget Databases | Curated collections of transcription factor binding motifs and regulatory interactions | Motif enrichment analysis in SCENIC+; improved TF identification [65] |
| ChIP-seq Reference Data | Genome-wide mapping of transcription factor binding sites | Validation of predicted TF-target region relationships [65] [3] |
| eQTL Data (GTEx/eQTLGen) | Catalog of genetic variants associated with gene expression levels | Validation of predicted region-to-gene links [3] |
| STARR-seq Data | High-throughput assessment of enhancer activity | Validation of predicted enhancer regions [65] |
The following diagram illustrates how these tools can be integrated to unravel gene regulatory networks in evolutionary developmental biology, from data acquisition to biological insight:
SCENIC+, LINGER, and cRegulon represent distinct but complementary approaches to GRN inference from single-cell multi-omics data. SCENIC+ excels in enhancer-centric network inference with its extensive motif collection and established workflow. LINGER demonstrates groundbreaking accuracy through its lifelong learning approach that leverages external data. cRegulon addresses the critical dimension of combinatorial regulation that is largely overlooked by other methods.
For evolutionary developmental biology research, the choice of tool depends on the specific biological question:
As the field advances, integration of these complementary approaches may provide the most comprehensive understanding of the gene regulatory networks that underlie developmental processes and their evolution.
In evolutionary developmental biology, the inference of Gene Regulatory Networks (GRNs) from single-cell multi-omics data has become a powerful approach for understanding the molecular basis of phenotypic divergence. However, a significant challenge persists: the computational predictions of regulatory interactions, especially those identified through cross-species comparisons, require robust experimental validation to confirm their biological relevance. Cross-species validation ensures that identified regulatory elements and network architectures are genuine drivers of evolutionary change rather than technical artifacts or species-specific peculiarities.
The integration of single-cell technologies has revolutionized GRN inference by enabling the profiling of chromatin accessibility, gene expression, and epigenetic states at unprecedented resolution [85] [86]. These advances allow researchers to move beyond static network models toward dynamic representations of regulatory changes across evolutionary timescales. Nevertheless, the accuracy of inferred networks depends heavily on the validation strategies employed, particularly when comparing distantly related species with distinct genomic architectures.
Recent work on the Tasmanian snow skink (Carinascincus ocellatus) demonstrates an innovative approach to chromosome-level genome validation using Oligo-FISH (Fluorescence In Situ Hybridization) [87]. This methodology provides direct cytogenetic confirmation of computational genome assemblies, which is particularly crucial for accurately resolving microchromosomes that are prevalent in reptilian genomes and notoriously difficult to assemble.
Table 1: Key Components of the Oligo-FISH Validation Protocol
| Component | Specification | Function in Validation |
|---|---|---|
| Oligonucleotide Probes | Thousands of short probes designed from genome assembly | Provide specific binding sites for chromosomal visualization |
| Metaphase Spread Preparation | Single C. ocellatus metaphase slide | Enables chromosomal visualization at condensed state |
| Hybridization Conditions | Standardized buffer and temperature conditions | Ensure specific probe binding to chromosomal targets |
| Imaging & Analysis | Fluorescence microscopy with appropriate filters | Visualize and confirm all 15 chromosomes including microchromosomes |
The protocol involves designing thousands of short oligonucleotide probes directly from the genome assembly itself, then hybridizing them to metaphase chromosome spreads. This single-step approach provides independent cytogenetic confirmation of computational assemblies and has demonstrated utility across multiple skink species, highlighting its potential for comparative genomic studies [87]. The method is particularly valuable for resolving evolutionary rearrangements and validating conserved syntenic blocks across species boundaries.
The emergence of single-cell multi-omics technologies has prompted development of novel computational frameworks for GRN inference that leverage both transcriptomic and chromatin accessibility data [85] [86]. These methods overcome limitations of traditional bulk sequencing by capturing cellular heterogeneity while providing insights into regulatory changes that drive evolutionary adaptations.
Table 2: Benchmarking Metrics for GRN Inference Methods in Evolutionary Studies
| Method Category | Key Principles | Strengths | Limitations for Cross-Species Studies |
|---|---|---|---|
| Tree-Based Methods | Inference of regulatory relationships using tree models | Handles non-linear relationships; robust to noise | May require species-specific parameter tuning |
| Correlation-Based Networks | Weighted gene co-expression network analysis (WGCNA) | Identifies co-regulated modules; preserves network topology | Cannot distinguish direct vs. indirect regulation |
| Integration Methods | Combining ChIP-seq/ATAC-seq with transcriptome data | Provides direct evidence of TF binding | Requires cross-species compatible epigenomic data |
| Multimodal Integration | Simultaneous measurement of transcriptome and epigenome in same cell | Reduces spurious correlations from cellular heterogeneity | Technical challenges in cross-species application |
These inference methods can be applied to cross-species comparisons by analyzing homologous cell types or tissues across evolutionary lineages. For example, studies of craniofacial development in marsupials have revealed significant divergence in cis-regulatory elements despite high conservation of protein-coding genes [88]. Such approaches help identify which aspects of GRN architecture are evolutionarily stable versus those that contribute to phenotypic diversification.
This protocol describes the validation of chromosome-scale genome assemblies using oligonucleotide-based FISH, adapted from Saunders et al. [87] for cross-species applications.
Sample Preparation:
Oligonucleotide Probe Design and Labeling:
In Situ Hybridization:
Post-Hybridization Washes and Detection:
Image Analysis and Interpretation:
This protocol outlines the inference of gene regulatory networks from single-cell multi-omics data for cross-species evolutionary comparisons, based on methodologies reviewed by Badia-I-Mompel et al. [85] [86].
Experimental Design:
Computational Analysis of Single-Cell Data:
GRN Inference and Cross-Species Comparison:
Table 3: Research Reagent Solutions for Cross-Species Validation Studies
| Reagent/Material | Function | Example Application | Considerations for Cross-Species Work |
|---|---|---|---|
| Oligo-FISH Probe Sets | Chromosomal visualization and genome validation | Validating chromosome-scale assemblies [87] | Probe design must account for sequence divergence between species |
| Single-Cell Multiome Kits | Simultaneous profiling of gene expression and chromatin accessibility | GRN inference across species [85] [86] | Protocol optimization may be needed for different species |
| Transposition Enzyme (Tn5) | Tagmentation of accessible chromatin regions | ATAC-seq for regulatory element identification | Enzyme efficiency may vary across species; titration may be required |
| Cross-Species Harmony Integration Algorithms | Computational integration of multi-species single-cell data | Aligning homologous cell types across evolutionary lineages | Requires careful parameterization to preserve biological variation |
| CUT&Tag Reagents | Profiling transcription factor binding and histone modifications | Validation of predicted TF-binding sites | Antibody compatibility across species must be verified |
| Epigenome Editing Tools (CRISPR-dCas9) | Functional validation of regulatory elements | Testing enhancer activity across species | Requires species-specific guide RNA design and delivery optimization |
The integration of cross-species validation approaches with single-cell multi-omics has yielded significant insights into evolutionary mechanisms. For example, studies of craniofacial development have demonstrated that while genes involved in this process are highly conserved between marsupials and placental mammals, their cis-regulatory elements show significant divergence that likely reflects species-specific developmental processes [88]. Similarly, research on trained immunity has revealed evolutionarily conserved transcription factor networks that encode innate immune memory in long-lived stem cells across mammalian species [88].
These approaches are particularly powerful when applied to understanding human evolutionary history. Genomic investigations of ancient West Eurasians have identified signatures of positive selection related to adaptation to M. tuberculosis, revealing how autoimmune trade-offs have shaped modern genetic risk profiles for inflammatory bowel disease [89]. Such findings demonstrate the value of combining evolutionary genomics with functional validation to understand the health implications of our evolutionary past.
When implementing these protocols, researchers should consider that different taxonomic groups may present specific challenges. For example, studies of skinks and other reptiles must account for the presence of microchromosomes that require specialized assembly and validation approaches [87]. Similarly, cross-species comparisons of GRNs must distinguish genuine regulatory rewiring from technical artifacts arising from sequence divergence that affects probe hybridization or antibody binding in validation experiments.
Genome-wide association studies (GWAS) have successfully identified thousands of genetic variants associated with complex traits and diseases. However, over 90% of these variants lie in non-coding regions of the genome, obscuring their functional mechanisms and causal genes [90] [91]. This gap impedes the translation of genetic discoveries into biological insights and therapeutic interventions. Functional genomics approaches, particularly those utilizing single-cell multi-omics data, now enable researchers to map these non-coding variants to cell-type-specific regulatory mechanisms and gene networks.
The integration of single-cell RNA sequencing (scRNA-seq) and single-cell ATAC sequencing (scATAC-seq) provides unprecedented resolution for mapping enhancer-gene interactions and inferring gene regulatory networks (GRNs) across diverse cell types [90] [3]. These advancements are crucial for evolutionary developmental biology research, as they reveal how genetic variation shapes regulatory programs across cell types and developmental trajectories. This application note details computational protocols and experimental frameworks for linking GWAS variants to regulatory mechanisms through functional enrichment analysis and GRN inference.
Table 1: Computational Methods for Regulatory Mechanism Inference
| Method Name | Primary Function | Key Innovation | Data Input Requirements |
|---|---|---|---|
| scMultiMap [90] | Infers cell-type-specific enhancer-gene pairs from multi-omics data | Joint latent-variable model with analytical p-values; 100x faster than existing methods | Paired scRNA-seq and scATAC-seq from same cells |
| LINGER [3] | Infers gene regulatory networks (GRNs) using lifelong learning | Incorporates atlas-scale external bulk data as manifold regularization | Single-cell multiome data (paired gene expression & chromatin accessibility) |
| cRegulon [12] | Identifies combinatorial TF regulatory modules | Models TF cooperativity beyond single-TF regulons | scRNA-seq and scATAC-seq (paired or unpaired) |
The following diagram illustrates the comprehensive workflow for connecting GWAS variants to regulatory mechanisms and potential therapeutic targets:
Purpose: Determine which cell types show enrichment for GWAS signal and regulatory annotations.
Procedure:
Expected Output: Ranked list of cell types with enrichment p-values and fold-enrichment estimates.
Purpose: Acquire paired gene expression and chromatin accessibility data for prioritized cell types.
Procedure:
Alternative Approach: Utilize public single-cell multi-omics datasets from resources like BRAIN Initiative Cell Census Network, Human Cell Atlas, or CellXGene.
Purpose: Identify statistical associations between regulatory elements and potential target genes.
Procedure:
Validation: Compare associations with orthogonal datasets (Promoter Capture Hi-C, PLAC-seq) where available.
Purpose: Reconstruct comprehensive GRNs including TF-RE and TF-TG interactions.
Procedure:
Alternative Approach: Apply cRegulon for combinatorial TF module identification [12].
Purpose: Connect non-coding GWAS variants to their regulated genes through statistical colocalization.
Procedure:
Interpretation: Prioritize gene-variant pairs with posterior probability of colocalization (PP4) > 0.8 and SMR p-value < 2.5Ã10^-6.
Purpose: Experimentally validate computational predictions and prioritize candidates for therapeutic development.
Procedure:
Table 2: Key Research Reagents and Computational Resources
| Resource Type | Specific Examples | Function/Purpose | Access Information |
|---|---|---|---|
| Reference Datasets | ENCODE Epigenome Roadmap [3], GTEx eQTLs [3], BRAIN Initiative Cell Census | Provide external regulatory annotations and expression quantitative trait loci for prioritization and validation | Publicly available from respective consortia websites |
| Analysis Tools | scMultiMap [90], LINGER [3], cRegulon [12], COLOC, SMR | Perform statistical inference of enhancer-gene links, GRNs, and variant-to-function mapping | GitHub repositories with documentation |
| Experimental Reagents | 10x Genomics Multiome Kit, Luciferase Reporter Vectors, CRISPRa/i Systems | Generate multi-omics data and validate computational predictions experimentally | Commercial vendors (10x Genomics, Addgene, etc.) |
| Cell Type Markers | Microglia (TMEM119, CX3CR1), Astrocytes (GFAP), Neuronal (RBFOX3) | Annotate cell types in single-cell data and interpret cell-type-specific mechanisms | Published cell type atlases and marker databases |
Recent applications in Alzheimer's disease demonstrate the power of this integrated approach. A multi-omics analysis of AD integrated five GWAS datasets with two single-cell eQTL datasets and one bulk eQTL dataset, identifying 28 candidate causal genes for AD [92]. The study revealed that microglia contributed the highest number of candidate genes, followed by excitatory neurons and astrocytes, highlighting the cell-type-specific nature of AD genetic architecture.
The analytical workflow employed SMR and Bayesian colocalization to prioritize genes, followed by enhancer activity analysis using H3K27ac and ATAC-seq data. This led to the discovery that AD-risk variants associated with candidate causal genes are frequently located near or within enhancers specifically active in disease-relevant cell types. For example, the novel candidate gene PABPC1 was found to be associated with an AD-risk variant located within an enhancer active only in astrocytes [92].
The following diagram illustrates key signaling pathways enriched for Alzheimer's disease risk genes identified through functional genomics approaches:
The integration of single-cell multi-omics data with GWAS signals provides a powerful framework for mapping non-coding variants to cell-type-specific regulatory mechanisms. The protocols outlined hereâfrom cell type identification and enhancer mapping to GRN inference and experimental validationâenable researchers to bridge the gap between genetic associations and biological mechanisms. For evolutionary developmental biology research, these approaches offer particular promise for understanding how genetic variation shapes regulatory programs across cell types and developmental trajectories, ultimately accelerating the identification of therapeutic targets for complex diseases.
The integration of single-cell multi-omics data with advanced computational models marks a paradigm shift in GRN inference, moving from correlative maps to predictive, mechanistic models of gene regulation. The key takeaways are the critical importance of leveraging large-scale external data, as demonstrated by LINGER, and the need to model combinatorial TF interactions, as enabled by cRegulon, to truly capture the logic of development and disease. While challenges in data sparsity, model interpretability, and standardization persist, the field is rapidly advancing through foundational models and federated computational ecosystems. Future directions point towards the integration of spatiotemporal data, the application of these networks for in silico drug perturbation screening, and the translation of these insights into clinically actionable strategies for precision medicine, ultimately bridging the gap between cellular omics and therapeutic innovation.