Recombinant Danio rerio Poly (A)-specific ribonuclease PARN (parn), partial, is a recombinant protein derived from the zebrafish (Danio rerio) gene encoding Poly (A)-specific ribonuclease (PARN). PARN is an enzyme involved in the degradation of poly(A) tails of messenger RNA (mRNA), which plays a crucial role in regulating mRNA turnover and gene expression . This recombinant form is often used in research to study the functions of PARN in various biological processes.
Gene Names: The gene encoding PARN in zebrafish is known as parn and zgc:56067.
Other Names: It is also referred to as Poly(A)-specific ribonuclease PARN or poly(A)-specific ribonuclease (deadenylation nuclease) .
Host/Reactivities: The recombinant protein can be expressed in various hosts, including E. coli, yeast, baculovirus, or mammalian cells .
Purity: The purity of the recombinant protein is typically greater than or equal to 85% as determined by SDS-PAGE .
PARN is primarily known for its role in mRNA degradation by shortening the poly(A) tail, which is crucial for mRNA stability and translation efficiency . Beyond mRNA turnover, PARN has been implicated in the processing of non-coding RNAs, including telomerase RNA and ribosomal RNA (rRNA) . In zebrafish, PARN mutations have been associated with hematopoietic defects and developmental abnormalities, highlighting its importance in developmental biology .
Studies using zebrafish models have shown that PARN plays a critical role in hematopoiesis. Mutations in the parn gene lead to a decrease in mature myeloid cell lineages and hemoglobin levels, while hematopoietic stem cell numbers remain unaffected . This suggests that PARN is essential for the differentiation of hematopoietic stem cells into myeloid and erythroid lineages.
PARN has been identified as a factor involved in the maturation of human 18S ribosomal RNA (rRNA), which is crucial for ribosome biogenesis . It participates in the 3' to 5' trimming of the internal transcribed spacer 1 (ITS1) region of pre-rRNA, a step necessary for the production of mature 40S ribosomal subunits .
Mutations in the PARN gene have been linked to human diseases such as dyskeratosis congenita and pulmonary fibrosis . Dyskeratosis congenita is characterized by bone marrow failure, which can be replicated in zebrafish models by disrupting the parn gene .
| Feature | Description |
|---|---|
| Gene Names | parn, zgc:56067 |
| Hosts | E. coli, yeast, baculovirus, mammalian cells |
| Purity | ≥ 85% by SDS-PAGE |
| Function | mRNA degradation, non-coding RNA processing |
| Role | Description |
|---|---|
| mRNA Degradation | Shortens poly(A) tails to regulate mRNA stability |
| Non-coding RNA Processing | Involved in telomerase RNA and rRNA maturation |
| Hematopoiesis | Essential for differentiation of hematopoietic stem cells |
| Ribosome Biogenesis | Participates in 18S rRNA maturation |
STRING: 7955.ENSDARP00000028687
UniGene: Dr.78689
PARN is a 3'-5' exoribonuclease that specifically degrades poly(A) tails of mRNAs, serving as a key regulator of mRNA stability and gene expression in zebrafish. During early embryogenesis, PARN plays a crucial role in the maternal-to-zygotic transition by facilitating the degradation of maternal transcripts, which is essential for proper developmental progression. PARN activity is particularly important during the first 24 hours post-fertilization, coinciding with the clearance of maternal mRNAs to allow zygotic gene expression to take over. The enzyme's deadenylation activity provides temporal control of protein synthesis by affecting mRNA stability and translation efficiency .
Several critical parameters significantly impact recombinant zebrafish PARN activity in vitro:
| Parameter | Optimal Condition | Effect on Activity |
|---|---|---|
| pH | 7.0-7.5 | Activity decreases dramatically below pH 6.5 or above pH 8.0 |
| Divalent cations | 1.5-2.0 mM Mg²⁺ | Essential for catalysis; Mn²⁺ can substitute but alters specificity |
| Monovalent ions | 50-100 mM KCl | Higher concentrations (>150 mM) inhibit activity |
| Temperature | 25-28°C | Reflects physiological temperature in zebrafish |
| Substrate | 5'-capped poly(A) RNA | Cap interaction enhances processivity |
| Reducing agents | 1-5 mM DTT or βME | Maintains enzyme stability and activity |
The adenosine specificity in the active site is particularly affected by the choice of divalent metal ion, with Mg²⁺ promoting adenosine specificity while Mn²⁺ broadens substrate tolerance .
The RRM domain in zebrafish PARN contributes significantly to poly(A) binding, with mutations in this domain reducing poly(A) affinity while preserving catalytic function. This suggests a mechanistic separation between substrate binding and catalysis that may be differentially regulated in zebrafish compared to mammals .
PARN functions as an integral component of miRNA-mediated gene silencing pathways in zebrafish development through several mechanisms:
PARN interacts with components of the miRNA-induced silencing complex (miRISC), including Argonaute proteins, facilitating targeted deadenylation of miRNA-bound transcripts.
During maternal-to-zygotic transition, PARN cooperates with the miR-430 family, which targets hundreds of maternal mRNAs for deadenylation and subsequent degradation.
PARN exhibits a sequential coordination with the CCR4-NOT complex, where PARN initiates poly(A) tail shortening followed by CCR4-NOT completing the deadenylation process.
RNA-immunoprecipitation studies reveal tissue-specific PARN-miRNA target sets, particularly enriched in neural tissues and developing gonads.
This integration of PARN activity with miRNA pathways creates a sophisticated regulatory network essential for proper developmental timing and tissue specification in zebrafish .
Distinguishing direct from indirect effects of PARN activity requires a multi-faceted approach:
Direct target identification: Employ CLIP-seq (Cross-linking immunoprecipitation sequencing) to identify RNA molecules directly bound by PARN in vivo. This approach captures physical interactions between PARN and its target RNAs.
Temporal analysis: Implement time-course experiments after PARN inhibition or knockout. Direct targets typically show immediate changes in poly(A) tail length (detectable within 2-6 hours), while secondary effects emerge later.
Poly(A) tail measurements: Use TAIL-seq or PAL-seq methodologies to directly measure poly(A) tail lengths transcriptome-wide. Direct PARN targets will show increased poly(A) tail lengths shortly after PARN depletion.
Substrate validation: Perform in vitro deadenylation assays with purified recombinant PARN and candidate target RNAs to confirm direct enzymatic activity.
Reporter assays: Construct reporter genes containing 3'UTRs from potential target mRNAs and assess their stability and translation efficiency in PARN-deficient versus wild-type backgrounds.
This integrated approach allows researchers to build a high-confidence set of direct PARN targets distinct from downstream regulatory effects .
Researchers have several complementary options for creating PARN loss-of-function models in zebrafish:
CRISPR/Cas9 gene editing:
Design guide RNAs targeting conserved exons encoding the catalytic domain
Microinject 25-50 pg Cas9 mRNA with 25 pg guide RNA into one-cell embryos
Screen F0 founders for germline transmission of mutations
Establish stable mutant lines through outcrossing and genotyping
Morpholino-mediated knockdown:
Design translation-blocking morpholinos targeting the 5'UTR/start codon region
Optimize injection concentration (typically 0.1-0.3 mM)
Include appropriate controls (mismatch morpholinos, rescue experiments)
Validate knockdown efficiency by Western blot
Dominant-negative approach:
Express catalytically inactive PARN variants (e.g., with mutations in the active site)
Use tissue-specific or inducible promoters for temporal control
Monitor competition with endogenous PARN for substrate binding
Small molecule inhibition:
Treat embryos with deadenylation inhibitors at defined developmental stages
Titrate concentrations to minimize off-target effects
Validate specificity through rescue experiments
Each approach has distinct advantages for addressing different research questions, with CRISPR/Cas9 providing the most specific genetic model for long-term studies, while morpholinos offer rapid assessment of early developmental functions .
For high-quality recombinant zebrafish PARN preparation:
Expression system selection: The yeast Pichia pastoris system provides an excellent balance of proper folding, post-translational modifications, and yield for zebrafish PARN expression .
Expression construct design:
Clone full-length zebrafish parn cDNA into pPICZα vector
Include N-terminal 6×His tag followed by a TEV protease cleavage site
Optimize codon usage for expression host
Verify sequence integrity before transformation
Expression conditions:
Transform P. pastoris strain X-33 or SMD1168
Induce expression with 0.5% methanol at 25°C for 48-72 hours
Monitor expression by small-scale time-course analysis
Purification workflow:
Harvest cells and disrupt using glass beads or mechanical homogenization
Prepare cleared lysate in buffer containing 50 mM Tris-HCl pH 7.5, 300 mM NaCl, 10% glycerol, and 5 mM β-mercaptoethanol
Perform initial capture on Ni-NTA resin
Apply TEV protease to remove His-tag
Perform ion exchange chromatography (MonoQ)
Complete purification with size exclusion chromatography
Concentrate to 1-2 mg/ml and store at -80°C in small aliquots
Quality control:
Assess purity by SDS-PAGE (>95%)
Verify oligomeric state by size exclusion chromatography
Confirm identity by mass spectrometry
Validate activity using standardized poly(A) degradation assays
This protocol yields enzymatically active zebrafish PARN suitable for biochemical and structural studies .
Precise measurement of zebrafish PARN deadenylation activity requires careful assay design:
Substrate preparation:
Synthesize defined-length poly(A) RNA substrates (25-200 adenosines)
Include 5' cap structure (m7GpppG) for physiological relevance
Incorporate fluorescent (5'-Cy5) or radioactive labels for detection
Purify substrates to homogeneity by gel electrophoresis
Reaction conditions:
Buffer: 20 mM Tris-HCl pH 7.5, 50 mM KCl, 1.5 mM MgCl₂, 1 mM DTT
Enzyme concentration: 5-50 nM (titrate to ensure first-order kinetics)
Substrate concentration: 50-100 nM (below Km for initial rate determination)
Temperature: 25°C (zebrafish physiological temperature)
Time points: 0, 5, 10, 20, 30, 60 minutes
Analysis methods:
Denaturing PAGE for substrate length distribution
Phosphorimager or fluorescence scanning for quantification
Plot remaining substrate versus time for rate determination
Calculate processivity (adenosines removed per binding event)
Critical controls:
No-enzyme control
Heat-inactivated enzyme
EDTA-containing reaction (chelates metal ions)
Non-adenylated substrate to confirm specificity
Data analysis:
First-order rate constant determination
Comparative analysis across conditions
Michaelis-Menten parameters (Km, kcat, kcat/Km)
This methodology provides quantitative measurements of PARN activity suitable for comparing wild-type and mutant enzymes or assessing activity across developmental stages .
To elucidate stage-specific PARN functions in zebrafish development:
Conditional knockdown/knockout strategies:
Heat-shock inducible CRISPR systems
Photoactivatable morpholinos
Chemical-inducible gene expression systems (e.g., Tet-On/Off)
Tissue-specific promoters driving Cre recombinase in floxed parn lines
Temporal expression profiling:
Quantitative RT-PCR of parn across developmental timepoints
In situ hybridization to map spatial expression patterns
Transgenic reporters (parn promoter driving fluorescent proteins)
Western blotting to track protein levels
Stage-specific rescue experiments:
Introduce wild-type PARN at defined developmental timepoints in mutant backgrounds
Use photoactivatable or caged mRNAs for temporal control
Assess phenotypic rescue to define critical periods for PARN function
Transcript-specific analyses:
Perform RNA-seq at multiple developmental stages in PARN-deficient embryos
Focus on maternal transcript clearance during early development
Analyze zygotic transcript stability during organogenesis
Examine tissue-specific effects during later development
Integrative analysis:
Correlate PARN expression/activity with target mRNA stability
Map developmental phenotypes to molecular changes
Create temporal activity maps across development
This multi-faceted approach reveals how PARN function transitions from maternal mRNA clearance to tissue-specific roles during later development .
Comprehensive phenotypic analysis of zebrafish PARN mutants requires attention to several critical factors:
Developmental staging:
Use standardized staging criteria (hours post-fertilization, morphological landmarks)
Account for potential developmental delays when comparing to wild-type
Document development at multiple timepoints to capture progressive phenotypes
Genetic background considerations:
Maintain consistent genetic backgrounds when comparing mutants
Generate and analyze multiple independent mutant alleles
Create maternal-zygotic mutants to assess complete loss of function
Phenotypic characterization hierarchy:
Morphological assessment (body axis, organ formation, pigmentation)
Behavioral analysis (swimming patterns, responses to stimuli)
Cellular defects (using tissue-specific markers)
Molecular signatures (transcriptome, poly(A) tail dynamics)
Quantitative phenotyping approaches:
Develop objective scoring systems for morphological traits
Employ automated behavioral tracking systems
Use image analysis software for consistent measurement
Apply appropriate statistical methods for categorical and continuous data
Distinguishing primary from secondary effects:
Conduct time-course analyses to identify earliest defects
Perform tissue-specific rescue experiments
Use transcriptomics to identify direct PARN targets
Compare with phenotypes of other deadenylase mutants
This framework enables comprehensive characterization of how PARN dysfunction affects zebrafish development at organismal, cellular, and molecular levels .
Understanding the distinct contributions of maternal and zygotic PARN is crucial for accurate experimental interpretation:
Distinguishing maternal from zygotic effects:
Generate maternal-zygotic mutants (MZ, from homozygous mutant females)
Compare with zygotic-only mutants (Z, from heterozygous females)
The difference between MZ and Z phenotypes represents the maternal contribution
Developmental implications:
Maternal PARN primarily affects early development (first 6-10 hours)
Zygotic PARN becomes critical during and after maternal-to-zygotic transition
Some phenotypes may only appear in maternal-zygotic mutants
Experimental design adaptations:
Use maternal-specific knockdown techniques for early effects
Employ heat-shock inducible systems to bypass early requirements
Perform tissue-specific manipulations to isolate later functions
Molecular profiling strategies:
Analyze poly(A) tail lengths of known maternal transcripts
Perform ribosome profiling to assess translation regulation
Compare transcriptomes between MZ and Z mutants at multiple stages
Rescue experiment design:
Inject wild-type parn mRNA at the one-cell stage to rescue maternal contribution
Use transgenic expression to rescue zygotic functions
Apply tissue-specific rescue to map requirement domains
This approach creates a comprehensive understanding of PARN function throughout development and prevents misattribution of phenotypes to the wrong developmental program .
Comprehensive identification of PARN targets in zebrafish requires integration of multiple high-throughput methodologies:
CLIP-seq (Cross-linking immunoprecipitation sequencing):
UV-crosslink PARN to its RNA targets in vivo
Immunoprecipitate PARN-RNA complexes
Sequence captured RNA fragments
Identify direct binding sites with nucleotide resolution
TAIL-seq (Poly(A) tail sequencing):
Compare wild-type and PARN-deficient samples
Measure poly(A) tail lengths transcriptome-wide
Identify mRNAs with extended poly(A) tails in PARN mutants
Correlate with expression changes
Ribosome profiling:
Assess translational efficiency changes upon PARN manipulation
Correlate with poly(A) tail length alterations
Identify functional consequences of PARN-mediated deadenylation
Degradome sequencing:
Capture and sequence RNA decay intermediates
Identify transcripts undergoing active degradation
Compare degradation patterns between wild-type and PARN mutants
Data integration framework:
Develop computational pipelines to integrate multiple data types
Apply machine learning to predict high-confidence PARN targets
Validate top candidates with reporter assays
Categorize targets by developmental stage and tissue specificity
This multi-omics approach creates a comprehensive map of PARN targets and their regulatory dynamics throughout zebrafish development .
For precise quantification of PARN enzymatic parameters in developmental contexts:
Developmental stage-specific extract preparation:
Collect synchronously developing embryos at defined stages
Generate cytoplasmic extracts under native conditions
Normalize protein concentration across samples
Verify PARN levels by immunoblotting
Activity measurement approaches:
In-extract deadenylation assays using defined substrates
Immunodepletion controls to confirm PARN-specific activity
Comparison with recombinant enzyme under matched conditions
Inclusion of physiological competitors and regulators
Kinetic parameter determination:
Measure initial rates across substrate concentrations
Determine apparent Km and Vmax at each developmental stage
Assess processivity through single-molecule approaches
Quantify cap-dependence through comparative kinetics
Physiological substrate testing:
Isolate native mRNPs from staged embryos
Measure deadenylation rates on these physiological substrates
Compare with synthetic substrate behavior
Assess influence of associated proteins
Quantitative analysis framework:
Apply appropriate enzyme kinetic models
Account for competing activities in extracts
Normalize for PARN concentration differences
Create developmental enzyme activity profiles
This methodology reveals how PARN kinetic parameters evolve through development and respond to changing cellular environments .
When confronting discrepancies between in vitro and in vivo PARN activity:
Identify potential sources of discrepancy:
Missing cofactors or regulators in vitro
Compartmentalization effects in vivo
Post-translational modifications
Competing or cooperative factors in cells
Substrate accessibility differences
Bridging experimental approaches:
Supplement in vitro reactions with cellular fractions
Perform activity assays in semi-permeabilized cells
Immunoprecipitate native PARN complexes for activity testing
Compare recombinant and endogenous PARN directly
Systematic variant testing:
Test PARN variants lacking specific domains
Create phosphomimetic mutations to simulate PTMs
Assess activity with and without potential regulators
Vary reaction conditions to better match cellular environment
Advanced imaging approaches:
Develop FRET-based PARN activity reporters
Visualize deadenylation in living cells
Track substrate and enzyme dynamics simultaneously
Correlate spatial organization with activity
Mathematical modeling:
Develop kinetic models incorporating cellular factors
Fit experimental data from both in vitro and in vivo sources
Identify parameters that explain discrepancies
Use models to predict conditions that reconcile differences
This systematic approach identifies the cellular factors and conditions that modify PARN activity in vivo, explaining apparent contradictions and providing a more complete understanding of physiological regulation .