Recombinant Danio rerio RRP15 is a 249-amino acid protein involved in ribosomal RNA processing. It is known by alternative names including rrp15, zgc:56269, and RRP15-like protein. The protein is primarily localized in the cytoplasm and has been identified as a component critical for nucleolar formation and ribosome biogenesis . The amino acid sequence of Danio rerio RRP15 is:
MAALAVKPHVVVEDGNDDVSEVSISNDEESGADSDHEGADAENSGDEDGKSDEEQNEENPNAGWAEAMAKVLGKKTPDTKPSILLKNKQLDKIKEKEKKERLEKKKQLDKKRAWENICREKPDVVQDREHERNLQRVATRGVVQLFNALKNHQKNVNERIKEVGGSERKKSKILSSVSKKDFIDVLRGTDVAVKKEKEIKAEKSSWSVLKDDFMMGASMKDWDKERDEDGGGGEEDEREPAQEYSSESD .
Unlike the budding yeast Rrp15p, which functions exclusively as a component of pre-60S ribosomal subunits, Danio rerio RRP15 (like human RRP15) is found in both pre-40S and pre-60S subunits. This suggests a broader role in ribosome biogenesis across vertebrates compared to yeast . Methodologically, researchers investigating these functional differences typically employ sucrose gradient centrifugation to separate ribosomal subunits, followed by western blotting or mass spectrometry to identify associated proteins, revealing this distinct evolutionary divergence in function.
Recombinant Danio rerio RRP15 can be successfully expressed in multiple host systems including yeast, E. coli, baculovirus, and mammalian cell expression systems . The choice of expression system depends on your experimental requirements:
| Expression Host | Advantages | Best Applications |
|---|---|---|
| E. coli | High yield, cost-effective | Structural studies, antibody production |
| Yeast | Post-translational modifications | Functional studies requiring minimal modifications |
| Baculovirus | High-level expression, complex modifications | Protein-protein interaction studies |
| Mammalian Cell | Native-like modifications | Studies requiring authentic post-translational modifications |
For most basic research applications, E. coli-expressed protein yields sufficient purity (>85% as determined by SDS-PAGE) and functionality, though researchers should consider mammalian expression systems when studying interactions requiring native post-translational modifications .
Danio rerio RRP15 has emerged as a significant protein in cancer research due to its involvement in nucleolar stress pathways. Studies have demonstrated that RRP15 is frequently upregulated in colorectal cancer (CRC) and hepatocellular carcinoma (HCC), with expression levels correlating with TNM stage and poor survival outcomes .
When designing zebrafish-based cancer models, researchers should consider:
Creating transgenic zebrafish with conditional RRP15 expression to study its oncogenic potential
Employing CRISPR/Cas9-mediated knockdown to assess effects on cancer progression in vivo
Using xenograft models with human cancer cells modified to express zebrafish RRP15 to study cross-species conservation of oncogenic pathways
Experimental data indicates that silencing RRP15 induces ribosome stress, cell cycle arrest, and apoptosis, which collectively suppress cell proliferation and metastasis in cancer models . This makes it a promising target for developing novel cancer therapeutics.
The interaction between RRP15 and LMNA (lamin A) in Danio rerio, identified through affinity capture-mass spectrometry, suggests a potential link between nuclear envelope integrity and ribosome biogenesis . This interaction may provide insight into how nucleolar function is coordinated with nuclear structure.
To investigate this interaction, researchers typically employ:
Proximity-dependent biotinylation techniques (BioID) in zebrafish embryos, which have been adapted for in vivo vertebrate studies
Co-immunoprecipitation assays followed by western blotting
Fluorescence microscopy with co-localization analysis
The LMNA-RRP15 interaction identified through high-throughput screening had a BFDR (Bayesian False Discovery Rate) cutoff of ≤1%, indicating high confidence in this protein-protein association . This interaction opens new avenues for understanding laminopathies and their connection to ribosome biogenesis defects.
RRP15 deficiency induces ribosome stress that suppresses the Wnt/β-catenin signaling pathway through a fascinating molecular mechanism. Research has revealed that ribosome stress caused by RRP15 knockdown facilitates the translation of TOP mRNA LZTS2 (Leucine zipper tumor suppressor 2), which leads to nuclear export and degradation of β-catenin .
The experimental approach to studying this mechanism involves:
RRP15 silencing using siRNA or CRISPR/Cas9 in zebrafish embryos or cell lines
Assessment of ribosome biogenesis using polysome profiling and northern blotting
Analysis of Wnt/β-catenin pathway components through western blotting, immunofluorescence, and reporter assays
Translational efficiency analysis of TOP mRNAs using polysome fractionation and qRT-PCR
These findings suggest RRP15 could be a therapeutic target in cancers with hyperactive Wnt signaling, particularly in high-ribosome biogenesis colorectal cancers .
Recombinant Danio rerio RRP15 is typically supplied as a lyophilized powder and requires specific handling conditions to maintain structural integrity and biological activity:
| Storage Parameter | Recommended Condition | Rationale |
|---|---|---|
| Temperature | -20°C to -80°C | Prevents protein degradation |
| Formulation | PBS with 6% trehalose | Trehalose acts as a cryoprotectant |
| Reconstitution | Sterile PBS or compatible buffer | Maintains protein solubility |
| Freeze-thaw cycles | Avoid multiple cycles | Prevents protein denaturation |
After reconstitution, aliquot the protein to minimize freeze-thaw cycles, as repeated freezing and thawing can significantly reduce protein activity. The presence of 6% trehalose in the formulation helps maintain protein stability during freeze-thaw processes .
To investigate RRP15's critical role in nucleolar formation, researchers should employ a multi-faceted experimental approach:
Immunofluorescence microscopy: Use antibodies against nucleolar markers (fibrillarin, nucleolin) alongside RRP15 to visualize co-localization and structural changes following RRP15 depletion.
Electron microscopy: For ultrastructural analysis of nucleolar morphology changes.
RRP15 knockdown strategies:
Morpholino oligonucleotides in developing zebrafish embryos
siRNA or shRNA in zebrafish cell lines
CRISPR/Cas9-mediated gene editing for stable knockout models
Nucleolar isolation: Differential centrifugation to isolate intact nucleoli, followed by proteomic analysis to identify RRP15-dependent nucleolar composition changes.
Research has demonstrated that RRP15 is essential for nucleolar formation, and its perturbation induces nucleolar stress that activates checkpoint responses, making these methodologies crucial for understanding its fundamental role in cellular homeostasis .
Nucleolar stress induced by RRP15 deficiency can be effectively measured using several complementary approaches in zebrafish models:
rRNA processing analysis:
Northern blotting with probes targeting pre-rRNA intermediates
Pulse-chase labeling with 5-ethynyl uridine (EU) to track newly synthesized rRNA
Checkpoint activation markers:
Western blotting for p53 stabilization and phosphorylation
qRT-PCR analysis of p53 target genes (p21, Bax, PUMA)
Immunostaining for RPL5/RPL11 localization
Cell cycle analysis:
Flow cytometry with propidium iodide staining
EdU incorporation assays to measure S-phase entry
Phospho-histone H3 immunostaining to detect mitotic cells
In vivo assessment:
Transgenic zebrafish lines with fluorescent cell cycle reporters
Whole-mount immunostaining for checkpoint proteins
Live imaging of nucleolar dynamics using fluorescently tagged nucleolar proteins
Research has shown that RRP15 deficiency activates the RPL5/RPL11/5S rRNA-Mdm2-p53 axis in p53-proficient cells, causing G1-G1/S arrest, while p53-deficient cells show S-phase perturbation and ATR-Chk1-γH2AX axis activation .
When encountering low yield of recombinant Danio rerio RRP15, consider the following optimization strategies:
Expression system adjustment:
For E. coli expression: Test multiple strains (BL21, Rosetta, Arctic Express) and optimize induction conditions (temperature, IPTG concentration, induction time)
For yeast expression: Evaluate different promoters and secretion signals
For baculovirus: Optimize MOI and harvest time
Solubility enhancement:
Express as fusion protein with solubility tags (MBP, SUMO, GST)
Include appropriate chaperones to assist folding
Optimize lysis buffer composition with stabilizing agents
Purification optimization:
Test multiple affinity tags for optimal purification efficiency
Implement two-step purification strategy for higher purity
Adjust buffer conditions to maintain protein stability
Protein degradation prevention:
Include protease inhibitors throughout purification
Maintain cold chain during all processing steps
Determine optimal pH and salt concentration for stability
Based on published protocols, E. coli-based expression systems can achieve >85% purity as determined by SDS-PAGE when optimized correctly .
When facing discrepancies between in vitro and in vivo RRP15 studies, implement these systematic approaches:
Validate protein functionality:
Confirm proper folding using circular dichroism or thermal shift assays
Verify activity with functional assays (e.g., RNA binding assays)
Check for proper post-translational modifications
Reconcile experimental conditions:
Match buffer compositions between in vitro and cellular conditions
Consider physiological protein concentrations
Account for cellular compartmentalization effects
Employ complementary approaches:
Perform rescue experiments with wild-type protein
Use domain mapping to identify functional regions
Conduct structure-function relationship studies
Consider context-dependent effects:
Evaluate cell/tissue-specific co-factors
Assess developmental stage-specific functions
Examine microenvironment influences on protein activity
Research has shown that RRP15 functions differently in p53-proficient versus p53-deficient cellular contexts, which may explain some experimental discrepancies . Additionally, since RRP15 functions differ between yeast and vertebrates, evolutionary considerations should be incorporated when interpreting cross-species data .
The differential effects of RRP15 knockdown between p53-proficient and p53-deficient systems represent a crucial consideration for experimental design and data interpretation:
| Cell Type | RRP15 Knockdown Effect | Checkpoint Response | Cell Cycle Impact |
|---|---|---|---|
| p53-proficient cells (e.g., RPE1) | Nucleolar stress | RPL5/RPL11/5S rRNA-Mdm2-p53 axis | G1-G1/S arrest |
| p53-deficient cells (e.g., HeLa, MCF7) | Nucleolar stress | ATR-Chk1-γH2AX axis | S-phase perturbation, delayed S-G2/M progression |
To properly interpret these differential effects:
Always determine p53 status in your experimental system before studying RRP15 function
Include appropriate controls from both p53-proficient and p53-deficient backgrounds
Monitor multiple checkpoint pathways simultaneously (p53 and ATR-Chk1)
Validate findings across different cell types to ensure robustness
This differential response to RRP15 deficiency suggests potential selective targeting of p53-deficient cancer cells, as they undergo cell death rather than cell cycle arrest when RRP15 is depleted .
Based on findings from zebrafish and other vertebrate models, RRP15 shows significant potential as a therapeutic target in cancer treatment through several mechanisms:
Selective cancer cell targeting:
Metastasis inhibition pathways:
Wnt/β-catenin pathway modulation:
Future research should focus on developing zebrafish xenograft models with human cancer cells to test RRP15-targeting compounds, evaluating off-target effects of RRP15 inhibition on normal tissues, and developing small molecule inhibitors specific to RRP15.
Evolutionary studies of RRP15 across vertebrate species, including Danio rerio, can provide significant insights into ribosome biogenesis regulation:
Functional divergence analysis:
Compare RRP15 function between yeast (pre-60S specific) and vertebrates (pre-40S and pre-60S)
Identify conserved domains versus species-specific regions to determine core functionality
Interaction network evolution:
Map RRP15 protein-protein interactions across species using affinity purification-mass spectrometry
Determine which interactions are evolutionarily conserved versus species-specific
Regulatory mechanism comparison:
Analyze promoter regions and transcriptional control of RRP15 across species
Identify conserved regulatory elements that control expression timing
Disease-relevant variations:
Correlate species-specific variations with differential disease susceptibility
Identify potential compensatory mechanisms in different vertebrate models
Understanding the evolutionary trajectory of RRP15 function can provide insights into the fundamentals of ribosome biogenesis regulation and reveal potential species-specific vulnerabilities relevant to disease and therapeutic development.
Advanced imaging techniques offer powerful approaches to elucidate RRP15 dynamics during zebrafish development:
Super-resolution microscopy applications:
Stimulated emission depletion (STED) microscopy to visualize nucleolar subcompartments
Photoactivated localization microscopy (PALM) to track single molecules of RRP15 in live cells
Structured illumination microscopy (SIM) for dynamic nucleolar reorganization
Live imaging in zebrafish embryos:
Generate transgenic zebrafish with fluorescently tagged RRP15
Perform light sheet microscopy for long-term, low-phototoxicity imaging
Use selective plane illumination microscopy (SPIM) for whole-embryo visualization
Multi-modal imaging approaches:
Combine fluorescence imaging with electron microscopy using correlative light and electron microscopy (CLEM)
Implement expansion microscopy for high-resolution imaging of nucleolar components
Utilize lattice light sheet microscopy for high-speed 3D imaging
Functional imaging techniques:
Fluorescence recovery after photobleaching (FRAP) to measure RRP15 mobility
Förster resonance energy transfer (FRET) to detect protein-protein interactions
Fluorescence lifetime imaging microscopy (FLIM) to map RRP15 interaction dynamics