| Feature | Danio rerio TADA3 (tada3l) | Human TADA3 (TADA3) |
|---|---|---|
| Genome Location | OneToOne homolog (GeneCards) | 3p25.3 |
| Amino Acid Identity | ~72.5% (GeneCards) | 455 aa (human) |
| Key Domains | DNA-binding, adaptor modules | PCAF/ATAC interaction sites |
| Isoforms | Not explicitly reported | 4+ splice variants |
Key Insight: Zebrafish tada3l shares conserved domains with human TADA3, including regions critical for interactions with transcription factors and HAT complexes .
Human TADA3 is essential for:
Chromatin Remodeling: Part of the PCAF/ATAC complex, acetylating histones (e.g., H3K9) to enhance transcription .
p53-Mediated Apoptosis: Stabilizes and activates p53, a tumor suppressor critical for DNA damage responses .
Developmental Regulation: Linked to cell cycle progression and differentiation .
Embryogenesis: Likely involved in early development, given conserved HAT complex functions.
Stress Response: Potential role in DNA damage signaling, mirroring human p53 interactions .
While no direct protocols are documented for zebrafish TADA3, analogous methods for Danio rerio proteins (e.g., APR3/Atraid) suggest:
Cloning: Full-length cDNA subcloned into bacterial expression vectors (e.g., pET-21a) .
Expression: E. coli systems (e.g., Rosetta-gami2) with IPTG induction .
Purification: His-tag affinity chromatography, yielding >90% purity .
Example: Recombinant zebrafish APR3 (27–230 aa) is expressed as a His-tagged protein in E. coli, lyophilized, and stored at -20°C .
Note: Polyclonal antibodies (e.g., ABIN2854371) show predicted reactivity with zebrafish TADA3 based on sequence homology .
Cancer Models: Zebrafish TADA3 may regulate oncogenic pathways, given its role in p53 stabilization .
Neurodevelopment: Potential overlap with autism-associated transcriptional regulators (e.g., 5TRa loci) .
Epigenetic Studies: Investigating HAT activity in zebrafish using recombinant TADA3.
Tada3 (Transcriptional adaptor 3) in Danio rerio functions as a component of histone acetyltransferase (HAT) complexes, playing crucial roles in chromatin modulation and transcriptional regulation. Similar to its human ortholog, zebrafish tada3 likely participates in the PCAF and ATAC complexes that have histone acetyltransferase activity on histones H3 and H4 . It serves as a transcriptional activator adaptor that links transcriptional activators bound to specific promoters to histone acetylation and the transcriptional machinery .
To investigate its primary function, researchers should:
Perform comparative sequence analysis between human and zebrafish tada3
Conduct co-immunoprecipitation assays to identify binding partners
Use ChIP-seq to identify genomic binding sites
Employ CRISPR-Cas9 system for functional knockdown studies
The conservation between human TADA3 and zebrafish tada3 reflects their evolutionary importance in transcriptional regulation. While the search results don't provide exact conservation percentages, TADA3 functions are generally well-conserved across species . This conservation suggests similar roles in transcriptional activation and histone acetylation.
| Domain/Feature | Human TADA3 | Zebrafish tada3 | Functional Significance |
|---|---|---|---|
| HAT complex binding | Present | Likely present | Essential for chromatin modification |
| p53 interaction motif | Present | Likely conserved | Critical for p53-mediated responses |
| Nuclear localization signal | Present | Predicted present | Required for nuclear function |
Methodology for conservation analysis:
Perform multiple sequence alignment using CLUSTAL or MUSCLE
Identify conserved domains using InterPro or Pfam databases
Generate phylogenetic trees to understand evolutionary relationships
Conduct synteny analysis to examine genomic context conservation
While specific protein interactions of tada3 in zebrafish are not explicitly detailed in the search results, we can infer likely interactions based on human TADA3 studies. In humans, TADA3 interacts with p53 and is required for full p53 activity and p53-mediated apoptosis .
To investigate tada3 protein interactions in zebrafish:
Perform yeast two-hybrid screening using zebrafish tada3 as bait
Conduct co-immunoprecipitation followed by mass spectrometry
Use proximity labeling techniques (BioID or APEX) to identify interaction partners
Employ FRET or BRET assays to confirm direct interactions in vivo
To identify regulatory elements controlling tada3 expression in Danio rerio, researchers can employ several approaches:
CAGE (Cap Analysis Gene Expression) data analysis to precisely identify transcription start sites
Analysis of 3'UTR regulatory elements that may contain upstream sequence elements (USEs) influencing mRNA stability and translation
Promoter analysis using reporter gene assays with GFP fusion constructs
ChIP-seq studies to identify transcription factor binding sites in the tada3 promoter region
When analyzing 3'UTR regulatory elements, researchers should examine regions upstream of polyA signals (PAS) as these can significantly influence gene expression regulation .
For comprehensive analysis of tada3 expression patterns in zebrafish, researchers should employ multiple complementary techniques:
CAGE analysis: This allows precise mapping of transcription start sites (TSSs) and quantification of expression levels . For reproducible results:
RNA-seq: For whole-transcriptome analysis and detection of alternative splicing events.
Whole-mount in situ hybridization: To visualize spatial expression patterns during development.
Transgenic reporter systems: Creating GFP reporters driven by the tada3 promoter can provide live visualization of expression patterns .
Single-cell approaches: For cell-type specific expression profiling.
The p53-tada3 relationship likely plays a significant role in zebrafish transcriptional regulation and DNA damage response. In humans, TADA3 associates with p53 and is required for p53 activity and p53-mediated apoptosis . In zebrafish, p53 recognizes and binds to DNA containing specific p53-responsive elements (REs) to orchestrate genomic response to cellular stress signals .
To investigate this relationship in zebrafish:
Use ChIP-seq to identify overlapping binding sites of p53 and tada3
Perform co-immunoprecipitation to confirm direct interaction
Create tada3 knockdown/knockout zebrafish and assess effects on p53 target gene expression
Use reporter assays with p53-responsive elements to measure transcriptional activity in the presence/absence of tada3
When designing experiments, note that differences in p53 RE sequences can strongly affect p53 transactivation capacity and occur even between closely related species .
To identify tada3 target genes in zebrafish, researchers should employ a multi-faceted approach:
ChIP-seq analysis: To identify genomic regions directly bound by tada3, similar to the approach used to identify p53 target genes in zebrafish .
RNA-seq after tada3 manipulation: Compare transcriptomes after tada3 knockdown/knockout to identify differentially expressed genes.
Combinatorial bioinformatics: Use pattern-matching-based analysis to screen the zebrafish genome for potential regulatory elements, followed by prioritization analysis to identify candidate genes .
Validation experiments: Confirm direct regulation through:
Luciferase reporter assays
Site-directed mutagenesis of binding sites
Electrophoretic mobility shift assays (EMSA)
This approach has proven successful in identifying novel p53 target genes in zebrafish, including runx1, axin1, traf4a, hspa8, col4a5, necab2, and dnajc9 .
CRISPR-based systems offer powerful approaches for studying tada3 function in zebrafish:
Gene knockout: Generate complete or conditional tada3 knockout zebrafish lines to assess developmental and physiological impacts.
Base editing: Introduce specific mutations to study structure-function relationships.
CRISPRi/CRISPRa: For reversible repression or activation of tada3 expression.
Simultaneous mRNA/protein quantification: Develop dual fluorescent reporter systems to monitor both tada3 mRNA and protein levels in single, live cells . This approach:
Allows distinction between transcriptional and post-transcriptional regulation
Can identify trans-acting loci affecting tada3 expression
Provides insight into post-transcriptional modifications
When designing CRISPR experiments, consider potential off-target effects and ensure proper controls, including the use of multiple guide RNAs targeting different regions of the tada3 gene.
The optimal expression system depends on experimental requirements for recombinant zebrafish tada3:
| Expression System | Advantages | Limitations | Best Applications |
|---|---|---|---|
| E. coli | High yield, low cost, rapid | Limited post-translational modifications | Structural studies, antibody production |
| Insect cells | Better folding, some PTMs | Medium cost, moderate yield | Functional studies, protein-protein interactions |
| Mammalian cells | Native-like PTMs, proper folding | Higher cost, lower yield | Complex functional studies, signaling assays |
| Cell-free systems | Rapid, controllable | Limited scale, higher cost | Quick screening, toxic proteins |
Methodological considerations:
For E. coli expression, consider fusion tags to enhance solubility (MBP, SUMO, GST)
For eukaryotic expression, codon optimization may improve yields
Include appropriate purification tags (His, FLAG, Strep) based on downstream applications
Consider inducible expression systems if tada3 overexpression is toxic to host cells
For efficient purification of recombinant zebrafish tada3:
Initial capture: Affinity chromatography based on fusion tags:
Immobilized metal affinity chromatography (IMAC) for His-tagged tada3
Glutathione-agarose for GST-tagged constructs
Amylose resin for MBP fusions
Intermediate purification:
Ion exchange chromatography (IEX) based on tada3's theoretical pI
Hydrophobic interaction chromatography (HIC)
Polishing step:
Size exclusion chromatography (SEC) to remove aggregates and ensure homogeneity
If tada3 functions in a complex, consider co-expression and co-purification with binding partners
Quality control:
SDS-PAGE and Western blotting to confirm purity and identity
Mass spectrometry for exact mass determination
Circular dichroism to verify proper folding
Dynamic light scattering to check homogeneity
Verification of recombinant tada3 functionality should include multiple complementary assays:
Histone acetyltransferase (HAT) activity assays:
In vitro HAT assays using purified histones or nucleosomes as substrates
Measurement of acetylation by Western blotting with acetyl-lysine antibodies
Fluorometric or colorimetric HAT activity assays
Protein-protein interaction assays:
Pull-down assays with known binding partners (p53, other PCAF complex components)
Surface plasmon resonance (SPR) to determine binding kinetics
Isothermal titration calorimetry (ITC) for thermodynamic parameters
Transcriptional activation assays:
Reporter gene assays in cellular contexts
In vitro transcription assays with reconstituted transcription machinery
Structural integrity verification:
Limited proteolysis to confirm proper folding
Thermal shift assays to assess stability
Analytical SEC to confirm absence of aggregation
When designing constructs for recombinant zebrafish tada3 expression:
Domain architecture analysis:
Include all functional domains based on sequence comparison with human TADA3
Consider expressing individual domains for domain-specific studies
Tag selection and placement:
N-terminal vs. C-terminal tags based on known functional domains
Consider TEV or PreScission protease cleavage sites for tag removal
Test multiple tag combinations for optimal expression and functionality
Codon optimization:
Adjust codon usage to match expression host for improved yields
Avoid rare codons that might cause translational pausing
Vector selection:
Choose vectors with appropriate promoters for the expression system
Consider inducible systems for potentially toxic proteins
Include appropriate selection markers for stable expression
Sequence verification:
Confirm sequence fidelity before expression
Consider including mutation-prone regions in sequencing coverage
Recombinant zebrafish tada3 can be utilized in several advanced approaches to investigate chromatin modulation:
Reconstitution of HAT complexes:
Assemble recombinant PCAF/ATAC complexes using purified components
Test activity on various nucleosome substrates with different histone modifications
Investigate the role of tada3 in targeting and regulating HAT activity
Structure-function studies:
Create systematic mutations in tada3 to map domains required for complex assembly
Use cryo-EM or X-ray crystallography to determine structural details of tada3-containing complexes
FRET-based assays to study conformational changes during complex formation
Genome-wide approaches:
Use recombinant tada3 for ChIP-seq experiments to map genome-wide binding sites
CUT&RUN or CUT&Tag with recombinant tada3 for high-resolution mapping
Correlate tada3 binding with histone modification patterns and gene expression
Single-molecule studies:
Real-time observation of HAT complex assembly and activity using labeled recombinant components
TIRF microscopy to visualize tada3-mediated processes on individual nucleosomes
To investigate tada3's role in the p53 regulatory network in zebrafish:
Comparative network analysis:
Identify p53 target genes in zebrafish with and without tada3 function
Compare with known p53 targets in other species to identify conserved and divergent regulation
Map the core p53-dependent transcriptional network in zebrafish
Stress response studies:
Examine how tada3 affects p53-dependent responses to DNA damage
Test if tada3 is required for stabilization and activation of p53 in zebrafish
Investigate the role of tada3 in p53-mediated apoptosis during development
Mechanistic investigation:
Determine if tada3 affects p53 post-translational modifications
Investigate whether tada3 influences p53 binding to target genes
Study how tada3-containing HAT complexes modify chromatin at p53 target genes
Based on human studies, tada3 likely plays a role in p53 stability and activation in zebrafish, affecting cellular responses to DNA damage .
Resolving discrepancies between tada3 mRNA and protein levels requires sophisticated approaches:
Simultaneous quantification system:
Post-transcriptional regulation analysis:
Genetic mapping approaches:
Technical considerations:
Control for different half-lives of mRNA vs. protein
Account for temporal delays between transcription and translation
Consider cell-type specific post-transcriptional regulation
Zebrafish tada3 research offers valuable insights into human disease mechanisms:
Developmental disorders:
Study tada3's role in zebrafish embryogenesis to understand potential developmental impacts
Investigate whether tada3 dysfunction causes specific developmental phenotypes
Create disease-relevant tada3 mutations based on human variants
Cancer research:
Examine how tada3 affects p53-dependent tumor suppression mechanisms
Create zebrafish models with tada3 mutations to assess cancer susceptibility
Screen for compounds that modulate tada3 activity as potential therapeutic leads
Comparative studies:
Therapeutic development:
Use zebrafish tada3 models for high-throughput drug screening
Test gene therapy approaches targeting tada3 dysfunction
Develop small molecules that modulate tada3-containing complex activity
The transparency and rapid development of zebrafish embryos make them ideal for studying transcriptional regulation dynamics in vivo, complementing human disease studies with real-time visualization capabilities .