Dictyostelium discoideum is a cellular slime mold that is valuable in cell and developmental biology research because of its simple life cycle and ease of use . Recent studies suggest that Dictyostelium may be a source of novel lead compounds for pharmacological and medical research . The Elongator complex, highly conserved among eukaryotes, plays roles in transcription regulation, tRNA modification, and α-tubulin acetylation .
The Elongator complex consists of six subunits (Elp1–Elp6), organized into two sub-complexes: the core sub-complex Elp123 (Elp1–Elp3) and the accessory sub-complex Elp456 (Elp4–Elp6) .
Elp2 is the second largest subunit of the Elongator complex, characterized by two WD40 propeller domains . Along with Elp1, Elp2 contributes to the stability of the Elp123 sub-complex and integrates signals from different factors that regulate Elongator activity .
The Elongator complex regulates neurotransmitter release and synapse formation, as demonstrated in Drosophila neurons . In vitro studies using the HeLa cell line confirmed that Elongator directly interacts with RNAPII and facilitates transcription in a chromatin- and acetyl-CoA-dependent manner . The Elongator complex has also been reported to play distinct nuclear roles, including paternal DNA demethylation in mouse zygotes and involvement in microRNA (miRNA) biogenesis in Arabidopsis .
Dictyostelium discoideum has emerged as a valuable expression system for the production of eukaryotic proteins .
Causative mutations in the ELP2 gene are found in patients with neurodevelopmental disorders (NDDs), including intellectual disability (ID), autism, and epilepsy . A novel missense mutation in ELP1 was identified in siblings with intellectual disability and global developmental delay .
KEGG: ddi:DDB_G0275651
STRING: 44689.DDB0231759
Dictyostelium discoideum offers several distinct advantages for studying the Elongator complex and specifically elp2. As a social amoeba, it has been used for nearly a century as an inexpensive and high-throughput model system for studying fundamental cellular and developmental processes including cell movement, chemotaxis, differentiation, and autophagy . Its unique life cycle comprises both unicellular growth and multicellular developmental phases that occur over just 24 hours, allowing for rapid detection of developmental phenotypes .
The fully sequenced, low-redundancy, haploid genome of Dictyostelium provides a less complex system while still maintaining many genes and signaling pathways found in more complex eukaryotes . This genomic simplicity, combined with the ease of introducing gene disruptions, makes it possible to study elp2 function in a true multicellular context with measurable phenotypic outcomes . Additionally, available insertional mutant libraries facilitate pharmacogenetic screens that have enhanced our understanding of bioactive compounds at the cellular level .
The Elongator complex is a highly conserved multi-subunit complex (Elp1-6) involved in several cellular processes, with its best-characterized function being the modification of transfer RNAs (tRNAs). In Dictyostelium and other organisms, the Elongator complex participates in a three-step modification pathway that introduces chemical modifications at wobble uridines at position 34 in tRNAs, which serve to optimize codon translation rates .
Within this complex, elp2 (Elongator complex protein 2) serves primarily as a structural component that helps maintain the integrity and stability of the entire complex. It typically contains WD40 repeat domains that form a β-propeller structure, providing a platform for protein-protein interactions. While not directly catalytic, elp2 is essential for the proper functioning of the complex, as its absence can disrupt the modification pathway and lead to translation defects, particularly affecting genes with specific codon biases.
In Dictyostelium discoideum, the Elongator-dependent modification pathway introduces chemical modifications at wobble uridines at position 34 in specific tRNAs through a three-step process . This pathway is critical for optimizing codon translation rates and ensuring accurate protein synthesis.
The process begins with the Elongator complex (including elp2) catalyzing the addition of a carboxymethyl (cm) group to the wobble uridine. This modification is followed by additional enzymatic steps that can include thiolation to form 5-carboxymethyl-2-thiouridine (mcm5s2U) depending on the specific tRNA species . These modifications alter the structure and chemical properties of the tRNA, influencing its interaction with mRNA codons during translation.
Disruption of this pathway, such as through mutations in elp2 or other Elongator components, can lead to defective tRNA modification, resulting in translation inefficiencies, particularly for specific codon-biased genes. This can manifest as numerous cellular phenotypes affecting growth, development, and stress responses.
For optimal expression of recombinant elp2 in Dictyostelium discoideum, researchers should consider several key parameters:
Vector Selection and Design:
Extrachromosomal or integrating vectors containing strong, constitutive promoters like actin15 or discoidin promoters typically provide robust expression
For controlled expression, inducible systems such as tetracycline-regulated promoters may be preferable
N-terminal tags generally interfere less with elp2 function compared to C-terminal tags
Include appropriate selection markers (G418, Blasticidin S, or Hygromycin B) for stable transformant selection
Culture and Expression Conditions:
| Parameter | Recommended Condition | Notes |
|---|---|---|
| Temperature | 22°C | Higher temperatures may reduce protein stability |
| Media | HL5 medium with glucose | Supplementation with glucose enhances growth |
| Cell density | 2-5 × 10^6^ cells/ml | Mid-log phase provides optimal protein yields |
| Shaking speed | 150-180 rpm | Proper aeration improves expression |
| Expression time | 24-48 hours post-induction | For inducible systems |
Purification Considerations:
Gentle lysis using non-ionic detergents (0.5% Triton X-100) preserves protein structure
Include protease inhibitors to prevent degradation
Buffer conditions (pH 7.0-7.5) should be optimized to maintain elp2 stability
Consider adding low concentrations of glycerol (5-10%) in storage buffers
For validation of expression, Western blotting using antibodies against the tag or elp2-specific antibodies, coupled with functional assays to confirm activity, is essential before proceeding with further experiments.
Creating reliable elp2 knockout models in Dictyostelium requires careful design and comprehensive validation:
CRISPR-Cas9 Approach:
Design sgRNAs targeting exonic regions of elp2, preferably early exons
Use Dictyostelium-optimized Cas9 expression vectors
Include a drug resistance cassette flanked by homology arms for selection
Screen transformants initially by PCR across the target region
Homologous Recombination Method:
Generate a construct containing a selection marker (e.g., Blasticidin resistance cassette) flanked by 5' and 3' homology arms (500-1000 bp each) from the elp2 gene
Transform Dictyostelium cells by electroporation
Select transformants with appropriate antibiotics
Screen for successful gene disruption
Comprehensive Validation Strategy:
| Validation Method | Purpose | Technical Details |
|---|---|---|
| Genomic PCR | Confirm disruption | Use primers flanking the integration site and within the selection marker |
| Southern blotting | Verify proper integration | Identify changes in restriction fragment patterns |
| RT-PCR & qRT-PCR | Confirm loss of expression | Target multiple regions of the transcript |
| Western blotting | Verify protein absence | Use elp2-specific antibodies or against tagged protein |
| tRNA modification analysis | Confirm functional impact | LC-MS analysis of modified nucleosides |
| Phenotypic assessment | Characterize mutant | Evaluate growth, development, and stress responses |
| Complementation test | Validate specificity | Reintroduce wild-type elp2 and assess rescue |
A properly validated elp2 knockout should demonstrate clear genomic integration of the disruption construct, absence of elp2 mRNA and protein, altered tRNA modification profiles, and distinctive phenotypes that can be rescued by reintroduction of wild-type elp2.
When investigating elp2 function in Dictyostelium, researchers should systematically assess multiple phenotypic parameters:
Growth and Developmental Phenotypes:
Growth rate in axenic culture and on bacterial lawns
Timing of developmental progression (aggregation, mound formation, slug formation, culmination)
Morphology of multicellular structures
Spore viability and germination efficiency
Cellular Processes:
Chemotactic responses to cAMP and folate gradients
Cell motility parameters (speed, directionality, persistence)
Phagocytosis and macropinocytosis efficiency
Cytokinesis and cell division patterns
Molecular Phenotypes:
Stress Responses:
Sensitivity to oxidative stress (H₂O₂, paraquat)
Tolerance to heat shock
Response to nutrient limitation
Resistance to translational inhibitors
These phenotypic analyses should be performed with appropriate controls, including wild-type cells and complemented mutants, to ensure that observed effects are specifically attributable to elp2 disruption.
Mass spectrometry provides powerful tools for characterizing tRNA modifications affected by elp2 function:
Sample Preparation Pipeline:
Extract total RNA from wild-type and elp2 mutant Dictyostelium
Enrich tRNA fraction by size exclusion or specific purification methods
Enzymatically digest tRNAs to nucleosides using nuclease P1 and phosphatase
Separate nucleosides by reversed-phase HPLC
Detect modified nucleosides by mass spectrometry
LC-MS/MS Analysis Strategy:
| Analytical Approach | Application | Technical Parameters |
|---|---|---|
| Targeted MRM/PRM | Quantification of known modifications | Monitoring specific precursor-to-product transitions for mcm⁵s²U, mcm⁵U |
| Untargeted scanning | Discovery of novel modifications | Full scan MS with data-dependent MS/MS |
| Comparative analysis | Differential modification | Label-free quantification with internal standards |
Data Analysis and Interpretation:
Use modification-specific mass transitions and retention times for identification
Calculate modification index (ratio of modified to unmodified nucleosides)
Compare modification levels between wild-type and elp2 mutant samples
Correlate modification changes with phenotypic outcomes
Example Data Representation:
| tRNA Isoacceptor | Modification | Wild-type (%) | elp2 Knockout (%) | Fold Change |
|---|---|---|---|---|
| tRNA^Lys^UUU | mcm⁵s²U | 82.3 ± 3.1 | 7.2 ± 1.8 | -11.4 |
| tRNA^Glu^UUC | mcm⁵s²U | 78.6 ± 2.7 | 6.4 ± 2.2 | -12.3 |
| tRNA^Gln^UUG | mcm⁵s²U | 75.9 ± 3.5 | 9.1 ± 2.6 | -8.3 |
| tRNA^Arg^UCU | mcm⁵U | 67.2 ± 4.0 | 11.8 ± 3.1 | -5.7 |
This comprehensive MS-based approach allows researchers to precisely quantify the impact of elp2 disruption on specific tRNA modifications, providing molecular insights into the mechanisms underlying observed phenotypes.
Investigating elp2 protein interactions requires multiple complementary approaches:
Affinity Purification-Mass Spectrometry (AP-MS):
Express tagged elp2 (GFP, FLAG, or HA) in Dictyostelium
Optimize lysis conditions to preserve native interactions (mild detergents, physiological salt)
Perform affinity purification using tag-specific matrices
Analyze co-precipitated proteins by LC-MS/MS
Filter against control pulldowns to identify specific interactors
Proximity Labeling Methods:
BioID: Fuse elp2 to a biotin ligase (BirA*) to biotinylate proximal proteins
APEX2: Fuse elp2 to an engineered peroxidase for proximity labeling
These methods capture both stable and transient interactions in living cells
Yeast Two-Hybrid Screening:
Use elp2 as bait against a Dictyostelium cDNA library
Validate positive interactions by co-immunoprecipitation
Map interaction domains using truncated constructs
Co-immunoprecipitation and Western Blotting:
Target specific candidate interactors based on predictions or preliminary screens
Use reciprocal co-immunoprecipitations to confirm interactions
Include appropriate controls (tag-only, unrelated proteins)
Fluorescence-Based Interaction Studies:
Förster Resonance Energy Transfer (FRET) between fluorescently tagged proteins
Fluorescence Correlation Spectroscopy (FCS) to detect complex formation
Bimolecular Fluorescence Complementation (BiFC) to visualize interactions in vivo
Data Analysis and Visualization:
| Analysis Approach | Purpose | Implementation |
|---|---|---|
| Interaction network mapping | Visualize connectivity | Cytoscape with GO term enrichment |
| Interaction specificity scoring | Filter nonspecific binders | Compare against CRAPome database |
| Domain mapping | Identify interaction interfaces | Test truncated or mutated constructs |
| Comparative analysis | Evolutionary conservation | Compare with known interactions in other species |
When presenting elp2 interaction data, it's essential to classify interactions based on confidence (direct vs. indirect, strength of evidence) and functional relevance to the Elongator complex activity.
Advanced imaging approaches provide crucial insights into elp2 localization and dynamics:
Sample Preparation Optimization:
Express fluorescently tagged elp2 (GFP, mNeonGreen) at near-endogenous levels
Validate tag functionality by complementation testing
For immunofluorescence, optimize fixation to preserve native structures (4% paraformaldehyde)
Use specific antibodies with validated specificity for immunodetection
High-Resolution Imaging Techniques:
| Technique | Application | Technical Parameters |
|---|---|---|
| Confocal microscopy | Co-localization studies | 63x/1.4 NA objective, Airy disk = 1 |
| STED super-resolution | Sub-diffraction localization | Depletion laser 592 nm, 80-150 nm resolution |
| SIM (Structured Illumination) | Improved resolution | 100-120 nm resolution, 3D reconstruction |
| PALM/STORM | Single-molecule localization | 20-50 nm resolution, photoswitchable fluorophores |
Live-Cell Imaging Approaches:
Spinning disk confocal for rapid acquisition with minimal phototoxicity
Light sheet microscopy for extended time-lapse imaging during development
Single-particle tracking to follow elp2-containing complexes
FRAP (Fluorescence Recovery After Photobleaching) to measure mobility and binding dynamics
Quantitative Analysis Methods:
Co-localization analysis with organelle markers using Pearson's or Mander's coefficients
Intensity distribution analysis across cellular compartments
Tracking of dynamic changes during cell cycle or development
Quantification of nuclear/cytoplasmic distribution ratios
Environmental and Genetic Perturbations:
Localization changes under stress conditions (oxidative stress, nutrient limitation)
Redistribution during developmental transitions
Dependence on other Elongator components (localization in other Elongator mutants)
Effects of inhibitors of specific cellular processes
By combining these advanced imaging approaches with appropriate controls and quantitative analysis, researchers can gain detailed insights into the spatial and temporal dynamics of elp2 function in Dictyostelium.
When confronted with contradictory results in elp2 research, a systematic troubleshooting approach is essential:
Systematic Analysis Framework:
Methodological Differences:
Compare experimental protocols in detail (buffer compositions, incubation times, temperatures)
Assess the sensitivity and specificity of detection methods
Consider temporal factors (acute vs. chronic effects, developmental timing)
Biological Variables:
Strain background differences (axenic vs. non-axenic strains)
Growth conditions and media composition
Cell density and growth phase during experiments
Developmental stage variations
Technical Considerations:
Reagent quality, specificity, and batch effects
Equipment calibration and settings
Data processing approaches and cutoff criteria
Resolution Strategy Table:
| Contradictory Area | Potential Causes | Resolution Approach | Example Case |
|---|---|---|---|
| Localization patterns | Different tags or fixation methods | Test multiple tags and positions | N-terminal tag shows nuclear enrichment while C-terminal shows uniform distribution |
| Interaction partners | Condition-dependent interactions | Systematic variation of lysis conditions | elp2 associates with translation factors only during nutrient stress |
| Phenotypic severity | Genetic background or environmental factors | Create mutations in multiple strains | elp2 disruption causes severe growth defects in AX2 but mild effects in AX3 |
| tRNA modification levels | Technical sensitivity differences | Apply multiple orthogonal methods | LC-MS shows complete loss while northern blot shows partial reduction |
Reconciliation Approaches:
Design definitive experiments that directly address the contradiction
Implement multiple complementary techniques to measure the same parameter
Conduct side-by-side experiments controlling for all variables
Systematically vary conditions to identify parameters that explain divergent results
Consider that contradictory results may reveal condition-specific functions
When reporting apparently contradictory findings, researchers should transparently acknowledge the discrepancies, present all relevant data, discuss possible explanations, and propose experiments that could resolve the remaining questions.
Computational methods provide powerful tools for analyzing elp2 conservation:
Sequence Analysis Pipeline:
Retrieval and Alignment:
Collect elp2 sequences from diverse organisms (unicellular to mammals)
Perform multiple sequence alignment using MUSCLE or Clustal Omega
Refine alignments manually focusing on functional domains
Conservation Analysis:
Calculate per-residue conservation scores
Identify highly conserved motifs and domains
Map conservation onto known or predicted structures
Phylogenetic Analysis:
Construct maximum likelihood trees to infer evolutionary relationships
Analyze patterns of selection (dN/dS ratio)
Identify lineage-specific adaptations
Structural Bioinformatics:
Homology modeling based on solved structures of WD40 proteins
Threading approaches when sequence identity is low
Molecular dynamics simulations to compare dynamic properties
Identification of conserved interaction surfaces
Functional Prediction:
Analysis of co-evolving residues to predict functional dependencies
Prediction of post-translational modification sites
Identification of conformational switches
Domain architecture comparison across species
Visualization and Presentation:
| Analysis Type | Visualization Method | Interpretation Focus |
|---|---|---|
| Sequence conservation | Heat maps with conservation scores | Identify universally conserved residues |
| Structural mapping | 3D models with conservation coloring | Locate functional surfaces and interfaces |
| Phylogenetic relationships | Annotated trees with functional data | Detect correlation between sequence and function |
| Domain architecture | Schematic diagrams across species | Identify lineage-specific features |
Example Conservation Analysis Table:
| Region | Position | Dictyostelium | Yeast | Human | Conservation Score | Predicted Function |
|---|---|---|---|---|---|---|
| WD40-1 | 120-160 | LSGGQRSVRIWDL | LSGGQKTVRIWDL | LSGGQKTVRIWDL | 0.92 | Core structural element |
| WD40-2 | 210-250 | VTASADEMRCIWD | VTGSADKMRCIWD | VSGSADKMRCLWD | 0.87 | Protein interaction surface |
| Linker | 300-320 | PGSQSTLNK | PAGTSTFSK | PSSASTFNK | 0.45 | Flexible connector |
| C-term | 490-510 | DELLSRFK | DELLQRFQ | DELLQRFK | 0.78 | Regulatory domain |
This comprehensive bioinformatic analysis can reveal which aspects of elp2 structure and function are evolutionarily conserved, providing insights into fundamental mechanisms versus species-specific adaptations.
Quantifying the impact of elp2 mutations on translation requires multi-level analysis:
Global Translation Assessment:
Polysome Profiling:
Separate polysomes by sucrose gradient ultracentrifugation
Compare polysome-to-monosome ratios between wild-type and elp2 mutants
Identify mRNAs with altered ribosome occupancy by microarray or RNA-seq
Ribosome Profiling:
Sequence ribosome-protected mRNA fragments
Analyze ribosome density and distribution
Calculate translation efficiency (TE) for each transcript
Metabolic Labeling:
Pulse labeling with radioactive or non-radioactive amino acids
Measure global protein synthesis rates
Analyze synthesis rates of specific proteins by immunoprecipitation
Codon-Specific Translation Analysis:
Reporter Systems:
Luciferase constructs with varying codon usage
GFP reporters with synonymous codon substitutions
Dual reporters for measuring relative translation efficiency
Codon-Specific Translation Efficiency:
Calculate A-site codon occupancy from ribosome profiling data
Measure ribosome dwell times at specific codons
Correlate translation defects with tRNA modification levels
Proteome-Wide Impact Assessment:
Quantitative Proteomics:
SILAC or TMT labeling for precise protein quantification
Label-free quantification to identify differentially expressed proteins
Correlation of protein changes with mRNA levels to identify translation effects
Bioinformatic Analysis:
Codon adaptation index (CAI) analysis of affected genes
GO term enrichment of differentially translated mRNAs
Motif analysis of 5' and 3' UTRs of affected transcripts
Example Quantitative Translation Table:
| Measurement | Method | Wild-type | elp2 Mutant | p-value | Interpretation |
|---|---|---|---|---|---|
| Global protein synthesis | ³⁵S-Met incorporation | 100 ± 7.2% | 68.3 ± 5.4% | <0.001 | Significant global reduction |
| Polysome-to-monosome ratio | Polysome profiling | 2.8 ± 0.3 | 1.5 ± 0.2 | <0.001 | Reduced polysome formation |
| AAA-rich reporter | Luciferase assay | 100 ± 5.1% | 42.7 ± 4.8% | <0.001 | Severe defect in AAA codon translation |
| CAC-rich reporter | Luciferase assay | 100 ± 4.7% | 96.3 ± 5.2% | 0.412 | No significant effect on CAC codons |
This multi-level quantitative approach provides a comprehensive assessment of how elp2 disruption affects translation at both the global and codon-specific levels, connecting molecular defects in tRNA modification to functional outcomes in protein synthesis.
Research on Dictyostelium elp2 provides valuable insights into human disease mechanisms:
Neurological Disorders Connection:
Mutations in human Elongator components are linked to familial dysautonomia, intellectual disability, and amyotrophic lateral sclerosis
Dictyostelium elp2 studies can reveal fundamental mechanisms of neurodegeneration related to translation defects
The social amoeba provides a simplified system for studying conserved pathways affected in complex human diseases
Cancer Biology Applications:
Altered translation control is a hallmark of many cancers
Dictyostelium models can reveal how Elongator dysfunction affects cell migration, a process relevant to metastasis
Screening approaches using Dictyostelium elp2 mutants can identify compounds that modulate tRNA modification pathways
Model System Advantages:
Rapid generation of mutants and observation of phenotypes
Ability to perform high-throughput genetic and chemical screens
Simplified genetic background compared to human cells
Translational Research Strategies:
| Disease Connection | Dictyostelium Approach | Translational Value |
|---|---|---|
| Neurodegeneration | Study proteostasis in elp2 mutants | Identify pathways protecting against proteotoxicity |
| Cancer | Examine migration defects in elp2 mutants | Discover targets for anti-metastatic therapies |
| Metabolic disorders | Analyze metabolic changes in elp2 mutants | Reveal links between translation and metabolism |
| Stress sensitivity | Test various stressors on elp2 mutants | Identify protective pathways that could be therapeutic targets |
By leveraging the experimental advantages of Dictyostelium while focusing on conserved mechanisms, researchers can use elp2 studies to generate hypotheses and potential therapeutic approaches relevant to human disease .
Cutting-edge technologies are poised to transform elp2 research:
Single-Cell Technologies:
Single-cell transcriptomics to reveal cell-to-cell variability in responses to elp2 disruption
Single-cell proteomics to detect translation defects at individual cell level
Spatial transcriptomics to map elp2-dependent effects during multicellular development
Advanced Genetic Engineering:
CRISPR base editing for precise mutation introduction without double-strand breaks
Optogenetic control of elp2 expression or activity
Degron systems for rapid and reversible elp2 depletion
Structural Biology Approaches:
Cryo-electron microscopy of the entire Elongator complex
Hydrogen-deuterium exchange mass spectrometry to map dynamic interactions
Integrative structural biology combining multiple data sources
Systems Biology Integration:
Multi-omics approaches combining transcriptomics, proteomics, and metabolomics
Flux analysis to understand metabolic consequences of elp2 disruption
Mathematical modeling of translation dynamics under elp2 perturbation
Emerging Imaging Technologies:
| Technology | Application to elp2 Research | Advantage Over Current Methods |
|---|---|---|
| Lattice light-sheet microscopy | 4D tracking of elp2 dynamics | Higher spatiotemporal resolution with less phototoxicity |
| Expansion microscopy | Nanoscale localization of elp2 | Achieves super-resolution with standard microscopes |
| Correlative light-electron microscopy | Ultrastructural context of elp2 | Combines molecular specificity with ultrastructural detail |
| Label-free imaging (SRS, THG) | Native elp2 complex visualization | Avoids artifacts from fluorescent tags |
By leveraging these emerging technologies, researchers can address current limitations in studying elp2 function, including challenges in detecting transient interactions, visualizing dynamic processes, and connecting molecular changes to cellular phenotypes.
Investigating elp2's role in stress response requires systematic experimental approaches:
Comprehensive Stress Panel:
Oxidative stress (H₂O₂, paraquat, menadione)
Heat shock (30-37°C)
Osmotic stress (sorbitol, NaCl)
Nutrient limitation (amino acid starvation, glucose restriction)
Translation stress (cycloheximide, puromycin)
DNA damage (UV irradiation, MMS)
Experimental Design Parameters:
| Parameter | Options | Considerations |
|---|---|---|
| Stress intensity | Dose-response curves | Identify both sublethal and lethal thresholds |
| Timing | Acute vs. chronic exposure | Distinguish immediate vs. adaptive responses |
| Cellular state | Growth phase, developmental stage | Identify context-dependent sensitivities |
| Readouts | Survival, growth, gene expression | Capture multiple response dimensions |
| Genetic background | Various Dictyostelium strains | Control for strain-specific effects |
Advanced Analytical Approaches:
Time-resolved analysis:
Temporal profiling of stress responses
Identification of early vs. late response genes
Dynamic changes in proteome and metabolome
Selective rescue experiments:
Expression of specific tRNAs with modified nucleosides
Overexpression of stress response factors
Chemical complementation with translation enhancers
Mechanistic dissection:
Ribosome profiling under stress conditions
Analysis of protein aggregation and misfolding
Measurement of translation fidelity under stress
Integrated Multi-omics Strategy:
Parallel analysis of transcriptome, proteome, and metabolome
Pathway enrichment to identify affected cellular processes
Network analysis to reveal stress response organization
This comprehensive experimental strategy will reveal how elp2 disruption affects stress response pathways, potentially identifying specific stresses where tRNA modification plays a particularly critical role and uncovering mechanisms linking translation control to stress adaptation.