Recombinant Danio rerio Ubiquitin-like protein 5 (UBL5) is a protein derived from the zebrafish (Danio rerio) and produced through recombinant DNA technology . UBL5 is a member of the ubiquitin-like (UBL) protein family, which are known for their structural similarity to ubiquitin . Unlike ubiquitin, which typically marks proteins for degradation, UBL5 is believed to modulate protein function through binding to target proteins via an isopeptide bond .
Recombinant Danio rerio UBL5 is typically expressed in yeast cells . The yeast protein expression system is considered economical and efficient for both secretion and intracellular expression . Proteins expressed in yeast can undergo modifications like glycosylation, acylation, and phosphorylation, ensuring a conformation close to that of the native protein .
UBL5 is involved in the cellular response to mitochondrial protein misfolding . Research indicates that UBL5 does not degrade proteins but affects their function by binding to target proteins . Furthermore, UBL5 has been studied in the context of metabolic traits and obesity research using Psammomys obesus (fat sand rat) as an animal model .
In zebrafish, UBL5 is part of the peroxisomal protein inventory . Peroxisomes are essential organelles involved in various metabolic pathways .
Studies on ubiquitin-like proteins in Toxoplasma indicate that Urm1, another UBL, is involved in the oxidative stress response . Urmylation of TgURM1 is induced by oxidative stress, suggesting a role in metabolic regulation and antioxidative processes .
Deubiquitinases, like USP5, play a role in controlling cellular processes such as inflammation and carcinogenesis . USP5 is involved in protein metabolism by disassembling polyubiquitin chains . While the relationship between UBL5 and USP5 isn't direct, both are components of the ubiquitin-proteasome system .
Recombinant UBL5 proteins are used in various research applications, including:
KEGG: dre:394116
UniGene: Dr.15659
UBL5 is a highly conserved ubiquitin-like protein that possesses several unique structural features distinguishing it from other members of the UBL family. Unlike typical ubiquitin proteins, UBL5 lacks the C-terminal di-Gly motif necessary for forming covalent conjugates with other proteins. Instead, UBL5 contains a C-terminal dityrosine motif of unknown function . This structural difference suggests that UBL5 functions as a reversible regulator of protein functions rather than a protein degrader . The zebrafish UBL5 shares these distinctive features, making it an interesting model for studying non-canonical ubiquitin-like protein functions.
UBL5 plays multiple critical roles in cellular function across vertebrate systems, which likely extend to Danio rerio:
Pre-mRNA splicing regulation: UBL5 associates primarily with spliceosomal proteins and is essential for efficient pre-mRNA splicing . Depletion of UBL5 leads to decreased splicing efficiency and enhanced intron retention throughout the transcriptome .
Sister chromatid cohesion maintenance: UBL5 is required for proper chromosome alignment and segregation during cell division . Loss of UBL5 function causes premature separation of sister chromatids, indicating its essential role in the fidelity of chromosome segregation .
Endoplasmic reticulum stress response: UBL5 functions as an ER stress-responsive protein that undergoes rapid depletion in response to ER stressors . This depletion occurs through proteasome-dependent yet ubiquitin-independent proteolysis .
Cell survival regulation: UBL5 serves as a physiologically relevant survival regulator. Knockdown of UBL5 activates multiple death pathways and induces severe apoptosis, while overexpression of UBL5 specifically protects against ER stress-induced apoptosis .
UBL5 demonstrates remarkable evolutionary conservation across species. The fundamental functions of UBL5 have been preserved from yeast to humans, particularly its role in pre-mRNA splicing. The yeast ortholog Hub1 promotes spliceosome functions similar to human UBL5, indicating an ancient and essential role in eukaryotic cellular processes . This high degree of conservation suggests that findings from various model organisms, including zebrafish, likely have broad biological relevance. The conservation pattern makes zebrafish UBL5 an excellent model for studying fundamental aspects of UBL5 biology that apply across vertebrate species.
Based on successful approaches with mammalian UBL5, the following methodology is recommended for zebrafish UBL5:
Expression System Selection:
E. coli: BL21(DE3) strain with pET vector systems are suitable for high-yield expression
Consider using a His-tag system for efficient purification as demonstrated in studies of human UBL5
Optimal Expression Conditions:
Induction with 0.5 mM IPTG
Expression at lower temperatures (16-18°C) overnight to enhance soluble protein yield
Supplementation with 0.1 mM ZnCl₂ may improve proper folding
Purification Protocol:
Harvest cells and lyse in buffer containing 50 mM Tris-HCl pH 8.0, 300 mM NaCl, 10 mM imidazole, and protease inhibitors
Purify using nickel affinity chromatography
Apply size exclusion chromatography (Superdex 75) for higher purity
For functional studies, consider tag removal using TEV protease
Quality Control Measures:
SDS-PAGE to confirm purity (expected molecular weight ~8.5 kDa)
Mass spectrometry to verify intact protein sequence
Circular dichroism to assess proper folding
When designing experiments to investigate zebrafish UBL5's role in pre-mRNA splicing, researchers should consider:
Knockdown/Knockout Approaches:
Morpholino antisense oligonucleotides for transient knockdown
CRISPR-Cas9 for stable genetic modifications
Conditional knockout systems to avoid early developmental lethality
Splicing Assessment Methods:
RNA-Seq analysis to measure global intron retention events, as demonstrated in human cell studies
RT-PCR validation of specific candidate transcripts showing altered splicing
Minigene splicing assays to test specific intron processing efficiency
Interaction Studies:
Co-immunoprecipitation followed by mass spectrometry to identify spliceosomal interacting partners in zebrafish, focusing on homologs of known human UBL5 interactors like SART1, PRPC8, and EFTUD2
Proximity ligation assays to confirm interactions in situ
Yeast two-hybrid screening to identify direct binding partners
Rescue Experiments:
Complementation with wild-type versus mutant UBL5 to determine structure-function relationships
Overexpression of downstream splicing factors to identify compensatory mechanisms
Controls:
Include knockdown of known splicing factors for comparison
Use both scrambled morpholinos and uninjected controls
Implement rescue experiments with wild-type UBL5 to confirm specificity
To effectively investigate UBL5's involvement in ER stress response in zebrafish models:
ER Stress Induction Protocols:
Treatment with tunicamycin (1-5 μg/ml) to inhibit N-linked glycosylation
Thapsigargin (0.1-1 μM) to deplete ER calcium stores
Exposure to DTT (1-2 mM) to disrupt disulfide bond formation
UBL5 Stability Assessment:
Western blot analysis to monitor UBL5 protein levels following ER stress induction
Cycloheximide chase assays to determine UBL5 protein half-life under normal versus ER stress conditions
Proteasome inhibitors (MG132) to confirm proteasome-dependent degradation mechanisms
UPR Pathway Analysis:
qRT-PCR for UPR markers (BiP, CHOP, XBP1 splicing)
Phosphorylation status of PERK and eIF2α by western blot
Luciferase reporter assays for ATF6 and XBP1 transcriptional activity
Experimental Design Considerations:
Time-course experiments to capture early versus late UPR events
Dose-response studies to establish appropriate stressor concentrations
Zebrafish developmental stage selection (larvae versus adult tissues)
Transgenic Approaches:
Generate UBL5-GFP fusion lines to monitor subcellular localization changes during ER stress
Create inducible UBL5 overexpression lines to assess protective effects against ER stress-induced apoptosis
Based on successful approaches used with mammalian UBL5, the following comprehensive methodology is recommended:
Quantitative Proteomics Approaches:
SILAC (Stable Isotope Labeling with Amino acids in Cell culture) labeling combined with immunoprecipitation and mass spectrometry analysis, similar to the approach that successfully identified human UBL5 interactions with spliceosomal proteins
BioID proximity labeling: Fusion of zebrafish UBL5 with BirA* biotin ligase to identify proximal proteins in living cells
Crosslinking mass spectrometry (XL-MS) to capture transient interactions
Biochemical Validation Methods:
Co-immunoprecipitation experiments with tagged recombinant UBL5
GST pull-down assays with recombinant proteins to confirm direct interactions
Size exclusion chromatography to identify stable complexes
In Vivo Interaction Detection:
Bimolecular Fluorescence Complementation (BiFC) assays in zebrafish cells
FRET-based approaches to measure protein-protein interactions in zebrafish embryos
PLA (Proximity Ligation Assay) to detect interactions in fixed tissues
Data Analysis Considerations:
Apply stringent statistical filtering (p-value <0.05 and fold change >2)
Validate top candidates by reciprocal pulldowns
Perform Gene Ontology analysis to identify enriched functional categories, similar to the approach that revealed spliceosome and ribonucleoprotein complex enrichment in human UBL5 interactome
For comprehensive analysis of UBL5's effect on splicing in zebrafish:
RNA-Seq Experimental Design:
Use at least 3 biological replicates per condition
Consider developmental time points relevant to the research question
Include multiple UBL5 knockdown/knockout approaches to control for off-target effects
Bioinformatic Analysis Pipeline:
Align RNA-Seq reads to zebrafish genome (GRCz11/danRer11)
Quantify alternative splicing events: exon skipping, intron retention, alternative 5'/3' splice sites
Calculate Percent Spliced In (PSI) values for all annotated exons
Apply specialized splice junction analysis tools (rMATS, MAJIQ, or LeafCutter)
Validation Strategies:
RT-PCR validation of top differentially spliced events
Minigene constructs to test specific splice sites in controlled conditions
Cross-species comparison with known UBL5-dependent splicing events
Functional Impact Assessment:
Conduct GO enrichment analysis on genes with altered splicing
Analyze protein domain impacts of altered splicing
Examine potential introduction of premature termination codons and NMD targets
Data Presentation:
| Analysis Type | Control Samples | UBL5 Knockdown | Significance |
|---|---|---|---|
| Intron retention events | 245 | 1,358 | p < 0.001 |
| Exon skipping events | 178 | 452 | p < 0.001 |
| Alt. 5' splice sites | 112 | 289 | p < 0.01 |
| Alt. 3' splice sites | 98 | 265 | p < 0.01 |
| Transcript isoforms | 1,245 | 2,875 | p < 0.001 |
Note: This table presents hypothetical data based on patterns observed in human cells . Actual numbers would depend on experimental results.
To effectively investigate UBL5's role in cell survival and apoptosis in zebrafish models:
In Vivo Techniques:
TUNEL assays in zebrafish embryos following UBL5 knockdown/knockout
Acridine orange staining to detect apoptotic cells in live embryos
Transgenic zebrafish lines expressing fluorescent apoptosis reporters (secA5-YFP)
Whole-mount immunostaining for cleaved caspase-3
Cellular Assays (using zebrafish cell lines):
Flow cytometry with Annexin V/PI staining to quantify apoptotic populations
Caspase activity assays (caspase-3/7, -8, -9) to delineate apoptotic pathways
Mitochondrial membrane potential measurements (JC-1 or TMRE)
Cell viability/proliferation assays (MTT, BrdU incorporation)
Stress Response Evaluation:
Challenge with ER stressors (tunicamycin, thapsigargin) and measure survival
UBL5 overexpression rescue experiments under stress conditions
Time-course analysis of UBL5 protein levels following stress induction
Mechanistic Investigations:
RNA-Seq analysis to identify death pathways activated upon UBL5 depletion
Western blot analysis for key apoptotic markers (Bcl-2 family, p53)
Co-immunoprecipitation to identify anti-apoptotic binding partners
Quantitative Analysis Framework:
| Apoptotic Marker | Control | UBL5 Morphant | UBL5 Morphant + UBL5 mRNA |
|---|---|---|---|
| TUNEL+ cells | 12 ± 3 | 86 ± 12 | 18 ± 5 |
| Cleaved Caspase-3 | 1.0 ± 0.2 | 4.8 ± 0.6 | 1.3 ± 0.3 |
| p53 activation | 1.0 ± 0.1 | 3.2 ± 0.4 | 1.1 ± 0.2 |
| Cell viability (%) | 94 ± 2 | 45 ± 8 | 90 ± 4 |
Note: This table presents hypothetical data based on patterns observed in mammalian cells . Actual numbers would depend on experimental results.
When encountering discrepancies between zebrafish UBL5 data and results from other models:
Systematic Analysis Framework:
Compare experimental conditions:
Developmental timing differences
Tissue-specific versus whole-organism effects
Acute knockdown versus genetic knockout consequences
Completeness of protein depletion
Consider evolutionary adaptations:
Species-specific UBL5 binding partners
Compensatory mechanisms in different organisms
Paralog redundancy in teleost fish (which underwent genome duplication)
Technical considerations:
Antibody specificity and validation methods
Knockdown/knockout efficiency and verification
Expression system differences in recombinant studies
Reconciliation strategies:
Perform cross-species rescue experiments (e.g., human UBL5 in zebrafish models)
Use domain swapping to identify functional differences
Conduct comparative interactome analysis across species
Biological significance assessment:
Determine if differences reflect fundamental biological divergence or experimental artifacts
Consider whether divergent functions represent species-specific adaptations or core conserved functions
Researchers working with zebrafish UBL5 may encounter several technical challenges:
Antibody Specificity Issues:
Problem: Limited availability of zebrafish-specific UBL5 antibodies
Solution: Generate custom antibodies against zebrafish UBL5; validate using knockout controls; use epitope tagging approaches (HA, FLAG) for recombinant protein detection
Developmental Lethality:
Problem: Complete loss of UBL5 may cause early developmental arrest
Solution: Use conditional knockout systems; temperature-sensitive mutants; tissue-specific CRISPR; dose-dependent morpholinos; study hypomorphic alleles
Protein Stability Challenges:
Solution: Use proteasome inhibitors for stabilization; perform experiments under controlled stress conditions; optimize sample preparation and handling to minimize degradation
Off-Target Effects:
Problem: Morpholino or CRISPR approaches may have unintended consequences
Solution: Use multiple independent knockdown reagents; perform careful rescue experiments; include appropriate controls; validate with genetic mutants
Functional Redundancy:
Problem: Other proteins may compensate for UBL5 loss
Solution: Consider combinatorial knockdown approaches; examine acute versus chronic loss effects; analyze compensatory gene expression changes
RNA-Seq Analysis Challenges:
Problem: Complex splicing patterns can be difficult to interpret
Solution: Use specialized splicing analysis tools; increase sequencing depth; validate key events with RT-PCR; consider longer read sequencing technologies
To establish phenotypic specificity to UBL5 dysfunction:
Essential Validation Criteria:
Evaluation Framework Table:
| Validation Criterion | Strong Evidence | Moderate Evidence | Weak Evidence |
|---|---|---|---|
| Multiple approaches | ≥3 independent methods | 2 independent methods | Single approach |
| Dose-dependency | Clear correlation | Partial correlation | No correlation tested |
| Rescue experiments | Complete rescue with WT | Partial rescue | No rescue attempted |
| Molecular signature | Multiple markers align | Some markers align | No molecular validation |
| Spatiotemporal pattern | Perfect correlation | Partial correlation | No correlation |
| Control experiments | Comprehensive controls | Basic controls | Minimal/no controls |
To identify direct splicing targets of UBL5 in zebrafish:
Integrative Genomics Approaches:
CLIP-seq (Cross-Linking Immunoprecipitation): Map direct RNA-protein interactions by UV-crosslinking UBL5 to its bound RNAs, followed by immunoprecipitation and sequencing
RNA-seq with splice junction analysis: Compare specific splicing events in control versus UBL5-depleted conditions
Targeted intron retention analysis: Focus on introns most affected by UBL5 depletion
Structural and Biochemical Approaches:
In vitro splicing assays: Test candidate pre-mRNAs with purified spliceosomes containing or lacking UBL5
RNA-protein binding assays: Assess direct binding of recombinant UBL5 to candidate RNA sequences
Spliceosome complex purification: Isolate distinct spliceosomal complexes and determine UBL5 association patterns
Genetic Screening Approaches:
Suppressor screens: Identify mutations that rescue UBL5 depletion phenotypes
Synthetic interaction screens: Discover genes that show enhanced phenotypes when partially depleted alongside UBL5
CRISPR-based screens: Target splicing regulators to identify those that phenocopy UBL5 loss
Candidate Gene Testing:
Based on human studies, the cohesion protection factor Sororin represents a high-priority candidate for detailed investigation, as its missplicing appears to be a critical consequence of UBL5 depletion
To explore potential zebrafish-specific functions of UBL5:
Comparative Genomics Strategies:
Sequence analysis: Identify zebrafish-specific motifs or domains within UBL5
Interactome comparison: Perform comparative proteomics across species to identify zebrafish-specific binding partners
Expression pattern mapping: Characterize developmental and tissue-specific expression patterns unique to zebrafish
Specialized Zebrafish Approaches:
Transparent embryo advantage: Utilize live imaging to track UBL5-GFP during development
Tissue regeneration models: Investigate UBL5's role in zebrafish-specific regenerative processes (fin, heart, etc.)
Temperature-sensitive experiments: Exploit poikilothermic nature of zebrafish to study temperature-dependent UBL5 functions
Unique Developmental Contexts:
Maternal-to-zygotic transition: Examine UBL5's role during early developmental processes
Metamorphosis: Investigate potential functions during larval-to-adult transition
Circadian rhythm studies: Analyze UBL5's potential role in day/night-dependent processes
Zebrafish-Specific Applications:
High-throughput screening: Develop zebrafish-based screens for compounds affecting UBL5 function
Behavioral studies: Assess UBL5's impact on zebrafish-specific behaviors
Environmental response: Examine UBL5's role in adaptation to environmental changes
Several cutting-edge technologies are poised to transform zebrafish UBL5 research:
Advanced Genome Editing Technologies:
Prime editing: Enables precise genetic modifications without double-strand breaks
Base editing: Allows specific nucleotide substitutions for subtle UBL5 modifications
Inducible CRISPR systems: Permits temporal and spatial control of UBL5 editing
Single-Cell Technologies:
Single-cell RNA-seq: Reveals cell type-specific UBL5 functions and splicing patterns
Single-cell proteomics: Maps UBL5 protein interactions at cellular resolution
Spatial transcriptomics: Preserves tissue context while analyzing UBL5-dependent gene expression
Advanced Imaging Methodologies:
Super-resolution microscopy: Visualizes UBL5 subcellular localization at nanometer scale
Light-sheet microscopy: Enables whole-organism imaging of UBL5 dynamics
Optogenetic tools: Allows precise spatiotemporal control of UBL5 function
Integrative Multi-Omics Approaches:
Integrated proteogenomics: Correlates UBL5-dependent transcriptome and proteome changes
Metabolomics integration: Links UBL5 function to metabolic pathways
Systems biology modeling: Predicts UBL5's role in complex cellular networks
Novel Protein Interaction Technologies:
Proximity-dependent biotin identification (BioID): Maps UBL5's protein neighborhood in living cells
APEX2 proximity labeling: Provides temporal resolution of UBL5 interactions
MS-CETSA (Cellular Thermal Shift Assay): Assesses UBL5 protein interactions in living cells
The integration of these emerging technologies will likely provide unprecedented insights into the molecular functions and biological significance of UBL5 in zebrafish development, physiology, and disease models.