👻Intro to Computational Biology Unit 7 – Gene Expression Analysis

Gene expression analysis is a crucial field in computational biology, focusing on how genetic information becomes functional molecules in cells. It explores the complex processes of transcription and translation, along with the intricate regulatory mechanisms that control gene activity. This unit covers key concepts, technologies, and computational tools used in gene expression studies. From high-throughput sequencing to statistical analysis methods, it provides a comprehensive overview of how researchers investigate gene activity patterns in various biological contexts and their applications in medicine and biotechnology.

Key Concepts in Gene Expression

  • Gene expression process converts genetic information encoded in DNA into functional gene products (proteins or non-coding RNAs)
  • Involves two main steps: transcription (DNA to RNA) and translation (RNA to protein)
  • Tightly regulated at multiple levels (transcriptional, post-transcriptional, translational, and post-translational)
  • Differential gene expression drives cellular differentiation and specialization
    • Allows cells with identical genomes to have distinct functions (neurons vs. muscle cells)
  • Gene expression patterns change in response to environmental stimuli, developmental stages, and disease states
  • Studying gene expression provides insights into cellular functions, regulatory networks, and disease mechanisms
  • High-throughput technologies (microarrays, RNA-seq) enable genome-wide expression profiling

DNA to RNA: Transcription Basics

  • Transcription initiates gene expression by synthesizing RNA from a DNA template
  • Carried out by RNA polymerase enzymes (RNA polymerase II for most protein-coding genes)
  • Transcription factors bind specific DNA sequences to recruit RNA polymerase and regulate transcription initiation
  • Consists of three main stages: initiation, elongation, and termination
    • Initiation: RNA polymerase binds promoter region and separates DNA strands
    • Elongation: RNA polymerase moves along the template strand, synthesizing complementary RNA
    • Termination: RNA polymerase releases the newly synthesized RNA and dissociates from DNA
  • Eukaryotic transcripts undergo post-transcriptional modifications (5' capping, splicing, 3' polyadenylation)
  • Alternative splicing generates multiple mRNA isoforms from a single gene, increasing proteome diversity

RNA to Protein: Translation Overview

  • Translation converts the genetic information in mRNA into a polypeptide chain
  • Occurs in the cytoplasm on ribosomes, large RNA-protein complexes
  • Genetic code specifies the correspondence between mRNA codons and amino acids
    • Each codon (triplet of nucleotides) encodes a specific amino acid or stop signal
  • tRNAs act as adaptor molecules, carrying amino acids to the ribosome and recognizing codons via anticodon sequences
  • Consists of three main stages: initiation, elongation, and termination
    • Initiation: Ribosomal subunits assemble on the mRNA with the help of initiation factors
    • Elongation: tRNAs deliver amino acids, which are linked together by peptide bonds
    • Termination: Release factors recognize stop codons and trigger polypeptide release
  • Post-translational modifications (folding, cleavage, chemical modifications) generate mature, functional proteins

Gene Regulation Mechanisms

  • Gene regulation controls the timing, location, and level of gene expression
  • Transcriptional regulation involves controlling the rate of transcription initiation
    • Transcription factors bind regulatory DNA sequences (promoters, enhancers, silencers) to activate or repress transcription
    • Chromatin structure and epigenetic modifications (DNA methylation, histone modifications) influence gene accessibility
  • Post-transcriptional regulation targets mRNA stability, localization, and translation efficiency
    • MicroRNAs (miRNAs) and RNA-binding proteins (RBPs) are key regulators at this level
  • Translational regulation controls the rate of protein synthesis from mRNA
    • Includes mechanisms like ribosome recruitment, start codon recognition, and translational repression
  • Post-translational regulation modifies protein activity, stability, and localization
    • Phosphorylation, ubiquitination, and other modifications can alter protein function and half-life
  • Feedback loops and regulatory networks enable precise control and coordination of gene expression

High-Throughput Sequencing Technologies

  • High-throughput sequencing (HTS) technologies enable massive parallel sequencing of DNA or RNA
  • RNA sequencing (RNA-seq) is widely used for transcriptome profiling and gene expression analysis
    • Provides a quantitative measure of transcript abundance across the genome
  • Involves converting RNA to cDNA, fragmentation, adapter ligation, and sequencing
  • Generates millions of short reads that are mapped back to a reference genome or transcriptome
  • Offers several advantages over microarrays: higher sensitivity, dynamic range, and ability to detect novel transcripts
  • Single-cell RNA-seq (scRNA-seq) allows expression profiling at the individual cell level
    • Captures cell-to-cell heterogeneity and identifies rare cell types
  • Other HTS applications: ChIP-seq (protein-DNA interactions), ATAC-seq (chromatin accessibility), ribosome profiling (translation)

Computational Tools for Expression Analysis

  • Quality control: Assessing read quality, trimming adapters, and filtering low-quality reads (FastQC, Trimmomatic)
  • Read alignment: Mapping reads to a reference genome or transcriptome (STAR, HISAT2, Bowtie2)
  • Quantification: Estimating transcript or gene abundance from aligned reads (featureCounts, HTSeq, Kallisto)
  • Normalization: Adjusting for differences in library size and composition (TPM, RPKM, DESeq2, edgeR)
  • Differential expression analysis: Identifying genes with significant expression changes between conditions (DESeq2, edgeR, limma)
  • Clustering: Grouping samples or genes based on expression patterns (hierarchical clustering, k-means, t-SNE)
  • Pathway and gene set enrichment analysis: Identifying overrepresented biological functions or pathways (GSEA, GO enrichment)
  • Data integration: Combining expression data with other omics data types (ChIP-seq, ATAC-seq, proteomics)

Statistical Methods in Gene Expression Studies

  • Normalization methods account for technical biases and enable fair comparisons across samples
    • Common methods: TPM (transcripts per million), RPKM (reads per kilobase per million), DESeq2, edgeR
  • Differential expression analysis identifies genes with significant expression changes between conditions
    • Based on statistical tests (e.g., Wald test, likelihood ratio test) and fold change thresholds
    • Multiple testing correction controls false positives (FDR, Bonferroni)
  • Clustering algorithms group samples or genes based on expression similarity
    • Hierarchical clustering: Builds a tree-like structure based on pairwise distances
    • K-means clustering: Partitions data into a predefined number of clusters
  • Principal component analysis (PCA) reduces data dimensionality and visualizes major sources of variation
  • Gene set enrichment analysis (GSEA) assesses the enrichment of predefined gene sets in ranked gene lists
  • Machine learning methods (e.g., random forests, support vector machines) can predict sample classes or outcomes based on expression signatures

Interpreting and Visualizing Expression Data

  • Heatmaps display expression levels across samples and genes using color gradients
    • Rows (genes) and columns (samples) are often clustered to reveal patterns
  • Volcano plots combine statistical significance (log10(pvalue)-log_{10}(p-value)) and magnitude of change (log2(foldchange)log_2(fold change))
    • Helps identify genes with large and significant expression changes
  • MA plots compare expression levels (log2(meanexpression)log_2(mean expression)) and fold changes (log2(foldchange)log_2(fold change))
    • Useful for assessing differential expression analysis results and identifying outliers
  • Principal component analysis (PCA) plots visualize sample relationships in reduced dimensional space
  • Gene set enrichment plots (e.g., GSEA enrichment plot, GO term bar plots) summarize the enrichment of biological functions or pathways
  • Network diagrams depict gene-gene interactions, co-expression relationships, or regulatory networks
  • Interactive visualization tools (e.g., Shiny apps, Plotly) enable dynamic exploration of expression data

Applications in Research and Medicine

  • Identifying disease biomarkers: Expression signatures associated with disease states can serve as diagnostic or prognostic markers
  • Drug discovery and development: Expression profiling can identify drug targets, assess drug efficacy, and predict side effects
  • Studying cellular differentiation and development: Expression dynamics during cell fate transitions provide insights into developmental processes
  • Characterizing tumor heterogeneity: Single-cell expression profiling reveals subpopulations within tumors with distinct properties
  • Investigating host-pathogen interactions: Expression changes in host cells upon infection shed light on immune responses and pathogenesis
  • Precision medicine: Expression-based patient stratification can guide personalized treatment decisions
  • Functional genomics: Integrating expression data with other omics data types to understand gene functions and regulatory networks
  • Comparative transcriptomics: Comparing expression patterns across species to study evolution and conservation of biological processes


© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.

© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.