ChatGPT vs Gemini vs Grok: RNA-seq Analysis

Prompt
RNA-seq data analysis - differential expression and pathway enrichment
ChatGPTOpenAI
RNA-seq data analysis - differential expression and pathway enrichment

Certainly! Here's an overview and guide on RNA-seq data analysis specifically focusing on differential expression and pathway enrichment:


RNA-seq Data Analysis: Differential Expression & Pathway Enrichment

1. Overview


  • RNA-seq measures gene expression by sequencing RNA transcripts.

  • Differential expression (DE) analysis identifies genes with significant changes in expression across conditions.

  • Pathway enrichment determines if certain biological pathways or gene sets are overrepresented among DE genes.



2. Workflow for RNA-seq Differential Expression Analysis

Step 1: Quality Control (QC)


  • Tools: FastQC, MultiQC

  • Check read quality, adapter contamination, duplication levels.


Step 2: Read Alignment or Quasi-Mapping


  • Align reads to reference genome or transcriptome.

  • Tools: HISAT2, STAR for alignment; Salmon, Kallisto for alignment-free quantification.


Step 3: Quantification


  • Obtain raw read counts per gene.

  • Tools: featureCounts, HTSeq-count (for aligned reads)

  • Alternatively, obtain transcript counts directly from Salmon/Kallisto.


Step 4: Data Normalization & Filtering


  • Import count data into DE analysis tools.

  • Filter out lowly expressed genes to reduce noise.


Step 5: Differential Expression Analysis


  • Use statistical packages designed for count data:

  • DESeq2 (R)

  • edgeR (R)

  • limma-voom (R)

  • Model expression changes between conditions.

  • Obtain log2 fold changes, p-values, adjusted p-values (FDR).



3. Differential Expression Analysis Example (R with DESeq2)

library(DESeq2)

# Load count matrix and sample metadata
counts <- read.csv("counts.csv", row.names=1)
colData <- read.csv("sample_info.csv", row.names=1)

# Create DESeq2 dataset
dds <- DESeqDataSetFromMatrix(countData = counts,
colData = colData,
design = ~ condition)

# Filter low counts
dds <- dds[rowSums(counts(dds)) > 10, ]

# Run DE analysis
dds <- DESeq(dds)
res <- results(dds)

# Summary and filtering significant genes
summary(res)
sig_genes <- subset(res, padj < 0.05 & abs(log2FoldChange) > 1)

# Save results
write.csv(as.data.frame(sig_genes), "DE_genes.csv")


4. Pathway Enrichment Analysis

After identifying DE genes, test if certain biological pathways/gene sets are enriched.

Common Databases:


  • KEGG

  • Reactome

  • Gene Ontology (GO)

  • MSigDB (Molecular Signatures Database)


Approaches:


  • Over-Representation Analysis (ORA): Test enrichment in DE gene list (usually up/down separately).

  • Gene Set Enrichment Analysis (GSEA): Uses whole ranked gene list (based on log2FC or statistics).



5. Pathway Enrichment Examples in R

Over-Representation Analysis Using clusterProfiler

library(clusterProfiler)
library(org.Hs.eg.db) # for human gene annotation

# Convert gene symbols to Entrez IDs
sig_genes_symbols <- rownames(sig_genes)
entrez_ids <- mapIds(org.Hs.eg.db, keys=sig_genes_symbols, column="ENTREZID", keytype="SYMBOL", multiVals="first")

# Remove NAs
entrez_ids <- na.omit(entrez_ids)

# KEGG enrichment
kegg_enrich <- enrichKEGG(gene = entrez_ids,
organism = 'hsa',
pvalueCutoff = 0.05)

# View results
head(kegg_enrich)

Gene Set Enrichment Analysis (GSEA)

library(clusterProfiler)

# Prepare ranked gene list: named numeric vector log2FC indexed by Entrez IDs
all_genes <- res$log2FoldChange
names(all_genes) <- mapIds(org.Hs.eg.db, keys=rownames(res), column="ENTREZID", keytype="SYMBOL", multiVals="first")
all_genes <- na.omit(all_genes)
all_genes <- sort(all_genes, decreasing=TRUE)

gsea_results <- gseKEGG(geneList = all_genes,
organism = 'hsa',
nPerm = 1000,
minGSSize = 10,
pvalueCutoff = 0.05,
verbose = FALSE)

head(gsea_results)


6. Visualization


  • MA-plot or volcano plot of DE genes.

  • Heatmaps of top DE genes.

  • Dotplots or barplots of enriched pathways.

  • Enrichment plots for GSEA.


Examples:

library(EnhancedVolcano)

EnhancedVolcano(res,
lab = rownames(res),
x = 'log2FoldChange',
y = 'padj',
pCutoff = 0.05,
FCcutoff = 1)

library(pheatmap)
# Heatmap of normalized counts for top DE genes
top_genes <- head(order(res$padj), 50)
norm_counts <- counts(dds, normalized=TRUE)
pheatmap(norm_counts[top_genes, ], cluster_rows=TRUE, cluster_cols=TRUE)


7. Additional Tips


  • Always check data quality and normalization.

  • Use appropriate models accounting for batch effects or covariates.

  • Adjust p-values (FDR) to control false positives.

  • Interpret pathway enrichment in biological context.

  • Consider multiple testing when interpreting results.



If you want, I can help you with specific code or interpreting results based on your dataset. Feel free to share data details!

10:48
GeminiGoogle
RNA-seq data analysis - differential expression and pathway enrichment

Okay, let's break down RNA-seq data analysis focusing on differential expression (DE) and pathway enrichment. This is a common workflow for understanding how gene expression changes in response to different conditions and what biological processes are affected.

1. RNA-seq Data & Objectives

  • RNA-seq Data: You'll start with raw reads from your RNA-seq experiment. These reads represent the RNA transcripts present in your samples. Your samples might be cells treated differently (e.g., drug vs. control), different tissues, different time points, etc.

  • Objectives: The core goals are:

  • Differential Expression (DE): Identify genes that show statistically significant differences in expression levels between the conditions you're comparing.

  • Pathway Enrichment: Determine which biological pathways (e.g., metabolic pathways, signaling pathways, disease-related pathways) are significantly enriched with differentially expressed genes. This helps you understand the biological implications of the expression changes.


2. Workflow Outline (Key Steps)

Here's a general workflow. Specific tools and parameters will depend on your experimental design, data quality, and research question.

  • Experimental Design & Sample Preparation:

  • Define your groups: Clearly define the experimental conditions you want to compare (e.g., treatment vs. control, different cell types, stages of development). Good experimental design is crucial.

  • Biological Replicates: Have enough biological replicates per group. More replicates generally lead to more robust and statistically significant results. A general rule of thumb is at least 3 biological replicates per condition, but power analysis can determine the optimal number.

  • RNA Extraction & Library Preparation: Ensure high-quality RNA extraction and library preparation to minimize biases. Different library prep protocols exist (e.g., polyA selection, rRNA depletion, strand-specific) that can affect the results.

  • Quality Control (QC):

  • FastQC: Use FastQC to assess the quality of your raw reads (per-base sequence quality, adapter contamination, overrepresented sequences, etc.).

  • MultiQC: Summarize QC results from multiple samples and tools.

  • Read Alignment/Mapping:

  • Alignment to a Reference Genome: Align the reads to a reference genome using tools like:

  • STAR: A very popular and fast aligner. Handles spliced reads well.

  • HISAT2: Another fast and accurate spliced aligner, often used as an alternative to STAR.

  • Bowtie2: Generally faster, but may be less accurate for RNA-seq, especially with complex splicing.

  • Genome Indexing: Create an index of the reference genome to speed up the alignment process. You'll need to create this index before running the aligner.

  • Alignment Parameters: Choose appropriate alignment parameters (e.g., gap opening/extension penalties, mismatch limits) based on your data and the aligner's documentation.

  • Handling Paired-End Data: If you have paired-end reads, configure the aligner accordingly.

  • Read Quantification (Gene/Transcript Counting):

  • Generate Count Matrix: After alignment, count the number of reads that map to each gene or transcript. Tools like:

  • featureCounts (from the Subread package): Fast and accurate, commonly used for gene-level quantification.

  • htseq-count (from HTSeq): Another widely used tool for gene-level counting.

  • Salmon/Kallisto (pseudo-alignment): These tools perform pseudo-alignment and directly estimate transcript abundances. They are faster than alignment-based methods and are becoming increasingly popular.

  • Differential Expression Analysis:

  • Normalization: Normalize the read counts to account for differences in library size and RNA composition between samples. Common normalization methods include:

  • DESeq2's normalization: Calculates size factors based on the median of ratios.

  • TMM (Trimmed Mean of M-values, implemented in edgeR): Removes the most extreme values from the data before calculating normalization factors.

  • RPKM/FPKM/TPM: While these were commonly used in the past, they are generally discouraged now. DESeq2's or TMM normalization methods are preferred.

  • Statistical Modeling: Use a statistical model to test for differential expression, accounting for variability between replicates.

  • DESeq2: A popular R package based on the negative binomial distribution. Handles complex experimental designs and batch effects well.

  • edgeR: Another R package, also based on the negative binomial distribution. Offers different normalization and dispersion estimation methods.

  • limma-voom: An R package that uses linear models after applying a variance-stabilizing transformation (voom) to the count data.

  • Multiple Testing Correction: Adjust p-values for multiple testing to control the false discovery rate (FDR). Common methods include:

  • Benjamini-Hochberg (BH) / FDR: Controls the expected proportion of false positives among the rejected hypotheses.

  • Bonferroni: A more conservative method that controls the family-wise error rate (FWER).

  • Results Interpretation: Analyze the results, focusing on genes with statistically significant differential expression (e.g., adjusted p-value < 0.05, absolute log2 fold change > a chosen threshold).

  • Pathway Enrichment Analysis:

  • Gene Set Enrichment Analysis (GSEA): Determines whether a predefined set of genes (e.g., genes in a particular pathway) shows statistically significant, concordant differences between two biological states.

  • GSEA software: The Broad Institute's GSEA software is a widely used tool.

  • fgsea (Fast Gene Set Enrichment Analysis): An R package that provides a fast and flexible implementation of GSEA.

  • Over-Representation Analysis (ORA): Tests whether a set of differentially expressed genes is enriched for genes associated with specific pathways or Gene Ontology (GO) terms.

  • DAVID (Database for Annotation, Visualization and Integrated Discovery): A web-based tool for functional annotation and enrichment analysis.

  • clusterProfiler (R package): A popular R package for GO and KEGG enrichment analysis.

  • Enrichr: Another web-based tool for enrichment analysis, with a large collection of gene sets.

  • Pathway Databases: Choose appropriate pathway databases based on your organism and research question (e.g., KEGG, GO, Reactome, MSigDB).

  • Visualization: Visualize the enriched pathways using bar plots, dot plots, or network graphs.

  • Further Analysis & Interpretation:

  • Gene Ontology (GO) Enrichment: Identify GO terms (biological process, molecular function, cellular component) that are enriched in your DE genes.

  • Network Analysis: Build networks of interacting genes and proteins to understand how the DE genes relate to each other. Tools like Cytoscape can be used for network visualization and analysis.

  • Functional Validation: Validate your findings using independent experimental techniques (e.g., qPCR, Western blotting, functional assays).

  • Integrative Analysis: Combine RNA-seq data with other omics data (e.g., proteomics, metabolomics) to gain a more comprehensive understanding of the biological system.


Detailed Explanation of Key Steps
  • Alignment (Step 3):

  • Splicing: RNA-seq reads often span exon-exon junctions (splice junctions). Aligners like STAR and HISAT2 are designed to handle these spliced reads efficiently.

  • Multiple Mapping: Reads can sometimes map to multiple locations in the genome, especially in regions with repetitive sequences or gene families. Consider how the aligner handles multiple mapping reads. Some aligners randomly assign reads to one of the possible locations, while others report all possible locations. This can impact downstream quantification.

  • Strand Specificity: If your library preparation was strand-specific (i.e., you know the direction of transcription), configure the aligner accordingly. This will improve the accuracy of the quantification.

  • Quantification (Step 4):

  • Gene vs. Transcript Level: You can quantify reads at the gene level (counting reads that map to any exon of a gene) or at the transcript level (counting reads that map to specific isoforms of a gene). Transcript-level analysis is more complex but can reveal isoform-specific regulation.

  • Read Counting Considerations: When using alignment-based methods (featureCounts, htseq-count), consider the overlap resolution mode (how to handle reads that overlap multiple features).

  • Differential Expression (Step 5):

  • Normalization is Critical: Normalization is essential to remove technical biases and ensure that you're comparing expression levels across samples in a fair way. Different normalization methods have different assumptions and may be more appropriate for certain datasets.

  • Model Design: The statistical model you use in DESeq2, edgeR, or limma-voom should reflect your experimental design. Include any relevant covariates (e.g., batch effects, sex, age) in the model.

  • Log2 Fold Change: The log2 fold change represents the difference in expression between two conditions on a logarithmic scale. A log2 fold change of 1 means a 2-fold increase in expression, while a log2 fold change of -1 means a 2-fold decrease in expression.

  • Shrinking Log2 Fold Changes: DESeq2 and other tools offer the option to "shrink" log2 fold changes, especially for genes with low counts or high dispersion. This can improve the accuracy of the estimates.

  • Pathway Enrichment (Step 6):

  • Choose the Right Tool: GSEA is generally preferred over ORA because it considers the expression levels of all genes, not just the DE genes. This can make it more sensitive to subtle changes in pathway activity.

  • Database Selection: The choice of pathway database is important. KEGG is a comprehensive database of metabolic pathways, while GO is a hierarchical classification of gene functions. Reactome is a curated database of biological pathways. MSigDB contains many different gene sets, including pathway databases and curated gene sets.

  • Interpretation: Pathway enrichment results should be interpreted carefully. Just because a pathway is enriched does not necessarily mean that it is the most important pathway. Consider the biological context of your experiment and the specific genes that are driving the enrichment.


Tools and Technologies
  • Programming Languages: R and Python are the most common languages for RNA-seq data analysis. R is particularly strong for statistical analysis and visualization, while Python is versatile for data manipulation and scripting.

  • R Packages: DESeq2, edgeR, limma, clusterProfiler, fgsea, ggplot2, dplyr

  • Python Packages: Biopython, pandas, numpy, scikit-learn

  • Command-Line Tools: FastQC, STAR, HISAT2, Bowtie2, featureCounts, Salmon, Kallisto

  • Cloud Computing: For large datasets, cloud computing platforms like AWS, Google Cloud, or Azure can provide the necessary computational resources.


Example Workflow (Using R and DESeq2)

# Install necessary packages (if not already installed)
if (!requireNamespace("BiocManager", quietly = TRUE))
install.packages("BiocManager")
BiocManager::install(c("DESeq2", "tximport", "readr", "ggplot2"))

# Load libraries
library(DESeq2)
library(tximport)
library(readr)
library(ggplot2)

# 1. Create a sample table (replace with your actual sample information)
samples <- data.frame(
sampleName = c("sample1", "sample2", "sample3", "sample4", "sample5", "sample6"),
condition = factor(c("control", "control", "control", "treatment", "treatment", "treatment"))
)

# 2. Path to your count files (assuming you used featureCounts)
file_paths <- file.path("./counts", paste0(samples$sampleName, ".txt")) #Adapt if needed!

# 3. Create a data frame with sample names and file paths
files <- setNames(file_paths, samples$sampleName)

# 4. Create a DESeqDataSet object
dds <- DESeqDataSetFromHTSeqCount(sampleTable = samples,
directory = "./counts", #directory with count files
design= ~ condition)

# 5. Remove low count genes (optional but recommended)
keep <- rowSums(counts(dds)) >= 10 # at least 10 reads in total across samples
dds <- dds[keep,]

# 6. Run DESeq2
dds <- DESeq(dds)

# 7. Extract results
res <- results(dds)

# 8. Adjust p-values for multiple testing (FDR)
res <- results(dds, alpha = 0.05) #FDR control

# 9. Order by adjusted p-value
resOrdered <- res[order(res$padj),]

# 10. Print results summary
summary(resOrdered)

# 11. Save results to a file
write.csv(as.data.frame(resOrdered), file="deseq2_results.csv")

# 12. Volcano plot (optional)
plot(resOrdered$log2FoldChange, -log10(resOrdered$padj),
xlab="log2 Fold Change", ylab="-log10(FDR)",
main="Volcano Plot", pch=16)
abline(h = -log10(0.05), col = "red") # Add line for FDR threshold

# --- Pathway Enrichment Analysis (using clusterProfiler) ---

# 1. Extract significant genes (adjust pvalue and log2fc thresholds as needed)
sig_genes <- rownames(resOrdered[which(resOrdered$padj < 0.05 & abs(resOrdered$log2FoldChange) > 1),])

# 2. Install and load clusterProfiler (if needed)
if (!requireNamespace("BiocManager", quietly = TRUE))
install.packages("BiocManager")
BiocManager::install("clusterProfiler")
library(clusterProfiler)

# 3. Perform GO enrichment analysis (replace 'org.Hs.eg.db' with your organism's annotation package)
if (!requireNamespace("BiocManager", quietly = TRUE))
install.packages("BiocManager")
BiocManager::install("org.Hs.eg.db") # Human annotation, adapt to your organism (Mm for mouse, etc.)
library(org.Hs.eg.db)

go_enrich <- enrichGO(gene = sig_genes,
OrgDb = org.Hs.eg.db,
ont = "BP", # Biological Process
pAdjustMethod = "BH",
pvalueCutoff = 0.05,
qvalueCutoff = 0.1,
readable = TRUE) # gene ID to gene symbol

# 4. Visualize GO enrichment results
dotplot(go_enrich, showCategory=10)
barplot(go_enrich, showCategory=10)

# 5. Save GO enrichment results
write.csv(as.data.frame(go_enrich), file = "go_enrichment.csv")

Important Considerations

  • Batch Effects: Batch effects are systematic variations that can arise from processing samples at different times, using different reagents, or running them on different sequencing machines. If batch effects are present, it's crucial to account for them in your statistical model. DESeq2 and limma-voom have built-in methods for handling batch effects.

  • Experimental Design: A well-designed experiment is essential for obtaining meaningful results. Consider factors like:

  • Replicates: Adequate biological replicates are crucial for statistical power.

  • Randomization: Randomize the order in which samples are processed to minimize bias.

  • Blocking: Use blocking to control for known sources of variation (e.g., batch effects).

  • Data Visualization: Visualize your data at every step of the analysis to identify potential problems and to gain insights into the results.


This is a comprehensive overview. Remember that RNA-seq analysis can be complex, and it's important to understand the assumptions and limitations of the different tools and methods. Consult the documentation for the specific tools you're using and seek advice from experienced bioinformaticians if needed. Good luck!

10:48
GrokxAI
RNA-seq data analysis - differential expression and pathway enrichment

RNA-seq data analysis for differential expression and pathway enrichment is a common workflow in bioinformatics used to identify genes that are differentially expressed between conditions (e.g., disease vs. control) and to understand the biological pathways or processes they are involved in. Below is a step-by-step guide to performing this analysis. I’ll assume you’re using R for the analysis, as it is one of the most popular tools (with packages like DESeq2 for differential expression and clusterProfiler for pathway enrichment). If you are using Python or command-line tools, let me know, and I can adjust accordingly.


Overview of the Workflow


  • Data Preprocessing: Quality control and alignment of raw RNA-seq reads.

  • Quantification: Generate a count matrix of gene expression.

  • Differential Expression Analysis: Identify differentially expressed genes (DEGs) between conditions.

  • Pathway Enrichment Analysis: Map DEGs to biological pathways or gene sets to interpret their functional significance.

  • Visualization: Create plots to summarize and communicate results.



Step 1: Data Preprocessing and Quantification


Tools Needed:


  • Quality control: FastQC, MultiQC

  • Trimming (optional): Trimmomatic, Cutadapt

  • Alignment: STAR, HISAT2

  • Quantification: featureCounts (from Subread), HTSeq-count, or Salmon/Kallisto (for pseudo-alignment)


Steps:


  • Download raw data: Obtain FASTQ files (RNA-seq reads) from sequencing or public databases (e.g., GEO, SRA).

  • Quality check: Use FastQC to assess the quality of raw reads. Look for issues like adapter contamination or low-quality bases.

  • Trimming (if needed): Use Trimmomatic to remove adapters or low-quality bases.

  • Alignment: Align reads to a reference genome using STAR or HISAT2. This produces BAM files.

  • Example for STAR:

STAR --runThreadN 8 --genomeDir /path/to/genome --readFilesIn sample1_1.fastq sample1_2.fastq --outFileNamePrefix sample1_

  • Quantification: Generate a gene count matrix using featureCounts or Salmon.

  • Example for featureCounts:

featureCounts -a /path/to/annotation.gtf -o counts.txt -T 8 *.bam

Output: A count matrix (rows = genes, columns = samples).


Step 2: Differential Expression Analysis in R


Packages Needed:


  • DESeq2 (for differential expression)

  • tximport (if using Salmon/Kallisto)

  • dplyr, ggplot2 (for data manipulation and visualization)


Steps:


  • Load data into R:

If you used featureCounts, read the count matrix directly. If using Salmon/Kallisto, use tximport to import transcript-level quantifications.

library(DESeq2)
library(tximport) # Optional, if using Salmon/Kallisto

# Read count matrix (from featureCounts)
counts <- read.table("counts.txt", header=TRUE, row.names=1)

# Read sample metadata (e.g., condition: control vs treated)
colData <- data.frame(condition = c("control", "control", "treated", "treated"),
row.names = colnames(counts))


  • Create a DESeqDataSet:

dds <- DESeqDataSetFromMatrix(countData = counts,
colData = colData,
design = ~ condition)

  • Filter low-count genes (optional but recommended):

keep <- rowSums(counts(dds)) >= 10
dds <- dds[keep,]

  • Run DESeq2 analysis:

dds <- DESeq(dds)
res <- results(dds, contrast=c("condition", "treated", "control"))

  • Extract differentially expressed genes (DEGs):

Filter based on adjusted p-value (padj) and log2 fold change (log2FC).
degs <- subset(res, padj < 0.05 & abs(log2FoldChange) > 1)
write.csv(as.data.frame(degs), file="degs.csv")

  • Visualizations:

  • MA plot:

plotMA(res, ylim=c(-2,2))

  • Volcano plot (using EnhancedVolcano or ggplot2):

library(EnhancedVolcano)
EnhancedVolcano(res,
lab = rownames(res),
x = 'log2FoldChange',
y = 'padj')


Step 3: Pathway Enrichment Analysis


Packages Needed:


  • clusterProfiler (for GO and KEGG enrichment)

  • org.Hs.eg.db (for human gene annotation, adjust for other organisms)

  • enrichplot, DOSE (for visualization)


Steps:


  • Prepare gene list:

Extract gene IDs (e.g., Ensembl IDs, Entrez IDs) from DEGs. If needed, convert IDs using packages like biomaRt.
deg_genes <- rownames(degs)

  • Perform GO enrichment (Gene Ontology):

library(clusterProfiler)
library(org.Hs.eg.db)

go_enrich <- enrichGO(gene = deg_genes,
OrgDb = org.Hs.eg.db,
keyType = "ENSEMBL", # Adjust based on your gene ID type
ont = "BP", # Biological Process; can also use "MF" or "CC"
pAdjustMethod = "BH",
pvalueCutoff = 0.05,
qvalueCutoff = 0.2)
write.csv(as.data.frame(go_enrich), file="go_enrichment.csv")


  • Perform KEGG pathway enrichment:

kegg_enrich <- enrichKEGG(gene = deg_genes,
organism = "hsa", # For human; adjust for other species
keyType = "kegg", # Adjust based on ID type
pAdjustMethod = "BH",
pvalueCutoff = 0.05,
qvalueCutoff = 0.2)
write.csv(as.data.frame(kegg_enrich), file="kegg_enrichment.csv")

  • Visualizations:

  • Dot plot:

library(enrichplot)
dotplot(go_enrich, showCategory=10)
dotplot(kegg_enrich, showCategory=10)

  • Bar plot:

barplot(go_enrich, showCategory=10)


Step 4: Interpretation and Reporting


  • Differential Expression Results: Focus on genes with significant padj (< 0.05) and meaningful log2FC (e.g., >1 or <-1). Check known biology or literature for top DEGs.

  • Pathway Enrichment Results: Look at enriched GO terms and KEGG pathways. Terms with low p-values and high gene counts are often biologically relevant.

  • Cross-Validation: Use multiple databases (e.g., GO, KEGG, Reactome) to confirm findings.

  • Visualization: Summarize results in figures (e.g., volcano plots for DEGs, dot plots for enriched pathways) for publications or reports.



Additional Notes


  • Normalization: DESeq2 automatically normalizes counts using the median of ratios method. Ensure your data isn’t pre-normalized if using DESeq2.

  • Batch Effects: If your data has batch effects, use packages like sva or include batch as a covariate in the DESeq2 design.

  • Organism: Adjust gene annotation databases and KEGG organism codes based on your study species (e.g., org.Mm.eg.db for mouse).

  • Alternative Tools:

  • For differential expression: edgeR, limma-voom

  • For pathway analysis: g:Profiler, DAVID (web-based)



Example R Script (Summary)


# Load libraries
library(DESeq2)
library(clusterProfiler)
library(org.Hs.eg.db)
library(enrichplot)

# Step 1: Load and prepare data
counts <- read.table("counts.txt", header=TRUE, row.names=1)
colData <- data.frame(condition = c("control", "control", "treated", "treated"),
row.names = colnames(counts))
dds <- DESeqDataSetFromMatrix(countData = counts, colData = colData, design = ~ condition)
dds <- DESeq(dds)
res <- results(dds, contrast=c("condition", "treated", "control"))

# Step 2: Extract DEGs
degs <- subset(res, padj < 0.05 & abs(log2FoldChange) > 1)
deg_genes <- rownames(degs)

# Step 3: GO and KEGG enrichment
go_enrich <- enrichGO(gene = deg_genes, OrgDb = org.Hs.eg.db, keyType = "ENSEMBL", ont = "BP")
kegg_enrich <- enrichKEGG(gene = deg_genes, organism = "hsa")

# Step 4: Visualize
dotplot(go_enrich, showCategory=10)
dotplot(kegg_enrich, showCategory=10)
write.csv(as.data.frame(degs), "degs.csv")
write.csv(as.data.frame(go_enrich), "go_enrichment.csv")
write.csv(as.data.frame(kegg_enrich), "kegg_enrichment.csv")


Troubleshooting and Common Issues


  • Low number of DEGs: Check if the experimental design has sufficient power (e.g., enough replicates

10:48

Analysis

This comparison demonstrates the different approaches each AI model takes when responding to the same prompt. Here are the key differences observed:

Response Characteristics

ChatGPT: Provides a direct response with 59 sentences.

Gemini: Provides a direct response with 199 sentences.

Grok: Provides a direct response with 136 sentences.

Key Takeaways

  • Each model brings unique strengths to this type of query
  • Response styles vary significantly between models
  • Consider your specific use case when choosing between these models

Try This Comparison Yourself

Want to test these models with your own prompts? Visit SNEOS.com to compare AI responses side-by-side in real-time.


This comparison was generated using the SNEOS AI Comparison ToolPublished: October 01, 2025 | Models: ChatGPT, Gemini, Grok