Recombinant Neurospora crassa ATP-dependent rRNA helicase rrp-3 (rrp-3), partial, refers to a genetically engineered version of the rrp-3 protein, which is involved in RNA processing and ribosome biogenesis. This enzyme plays a crucial role in unwinding RNA structures, a process essential for the proper functioning of ribosomes. The term "partial" indicates that the recombinant protein might not include the full-length sequence of the native rrp-3 protein.
rRNA helicases are enzymes that utilize ATP hydrolysis to unwind double-stranded RNA structures. These enzymes are vital for various cellular processes, including ribosome assembly, translation initiation, and RNA degradation. In the context of Neurospora crassa, a model organism for studying fungal genetics and molecular biology, rRNA helicases like rrp-3 are essential for maintaining cellular homeostasis and ensuring proper RNA metabolism.
Neurospora crassa offers a robust system for expressing and purifying recombinant proteins. Techniques such as the Strep-tag® system have been successfully employed for protein purification, as seen in the expression of nitrate reductase . This system allows for efficient purification of recombinant proteins, which is crucial for studying their biochemical properties.
The study of recombinant rRNA helicases like rrp-3 can provide insights into RNA metabolism and ribosome assembly. Understanding the biochemical properties and regulation of these enzymes can have implications for biotechnology and medical research, particularly in areas related to RNA processing and ribosome function.
While specific data tables for the recombinant Neurospora crassa ATP-dependent rRNA helicase rrp-3 (rrp-3), partial, are not available, the following table summarizes key aspects of RNA helicases and Neurospora crassa research:
| Aspect | Description | Relevance to rrp-3 |
|---|---|---|
| RNA Helicase Function | Unwinds RNA structures using ATP hydrolysis | Essential for ribosome assembly and RNA metabolism |
| Neurospora crassa | Model organism for genetics and molecular biology | Facilitates studies on RNA processing and helicase function |
| Recombinant Protein Expression | Utilizes systems like Strep-tag® for purification | Enables biochemical characterization of recombinant proteins |
Enzymatic Characterization of Recombinant Nitrate Reductase: This study demonstrates the use of Neurospora crassa for recombinant protein expression and purification, highlighting the potential for similar approaches with rRNA helicases .
Global Gene Expression in Neurospora crassa: Provides insights into gene regulation and RNA silencing pathways in Neurospora crassa, which are relevant to understanding the broader context of RNA helicase function .
RNA Polymerase III in Neurospora crassa: Shows the role of RNA Polymerase III in transcribing small RNAs, which can be relevant to understanding RNA processing mechanisms .
Neurospora crassa as a Model Organism: Highlights the organism's utility in studying host-virus interactions and RNAi-mediated responses, which can inform research on RNA helicases .
This protein is essential for pre-ribosomal RNA processing. It plays a role in 35S pre-rRNA maturation and its cleavage into mature 18S rRNA.
KEGG: ncr:NCU04504
How does RRP-3 contribute to the antiviral defense mechanism in Neurospora crassa?
Recent research has established N. crassa as a model organism for studying host-virus interactions and identified RRP-3 as a component of antiviral defense mechanisms . Upon viral infection with different RNA viruses (NcFV1, NcPV1, or RnPV2), RRP-3 protein levels are strikingly elevated . While the exact mechanism remains to be fully elucidated, several lines of evidence suggest RRP-3's role in antiviral defense:
RRP-3 upregulation coincides with activation of RNA interference (RNAi) components like DCL-2 (Dicer-like protein) upon viral infection.
As an ATP-dependent RNA helicase, RRP-3 may unwind viral RNA structures, potentially exposing them to degradation by RNAi machinery.
It may function in coordination with other components of the exosome complex to degrade viral RNAs through 3'→5' exonucleolytic activity, similar to the role of its yeast homolog Dob1p (Mtr4p) in RNA degradation .
Methodologically, the antiviral function of RRP-3 can be studied by comparing viral replication efficiency in wild-type versus rrp-3 mutant strains, analyzing RRP-3's association with viral RNAs through RNA immunoprecipitation, and identifying protein interaction partners during viral infection.
What structural characteristics enable the ATP-dependent helicase activity of RRP-3?
While the specific crystal structure of N. crassa RRP-3 has not been reported, insights can be drawn from other DEAD-box helicases. These proteins typically contain:
Two RecA-like domains that form a cleft for ATP binding and hydrolysis
Conserved sequence motifs, including:
The Walker A motif (P-loop) for binding ATP
The DEAD box (Asp-Glu-Ala-Asp) for ATP hydrolysis coordination
The SAT motif for coupling ATP hydrolysis to RNA unwinding
RNA-binding motifs (e.g., motifs Ia, Ib, IV, and V)
Based on molecular dynamics simulations of related helicases, the mechanism likely follows an inchworm-like translocation model where ATP binding and hydrolysis induce conformational changes between open and closed states . During the ATP cycle:
ATP binding stabilizes the closed conformation, bringing the two RecA-like domains together
This creates high-affinity RNA binding and destabilizes RNA duplexes
ATP hydrolysis and phosphate release reset the enzyme to the open conformation
These structural transitions are essential for processivity and can be studied using ATP analogs that arrest the helicase at different stages of the catalytic cycle.
How does RRP-3 coordinate with the exosome complex in RNA processing pathways?
By analogy to the yeast homolog Dob1p (Mtr4p), RRP-3 likely functions as a cofactor for the exosome complex, a multi-subunit assembly of 3'→5' exonucleases . This coordination is thought to occur through several mechanisms:
Substrate preparation: RRP-3 unwinds secondary structures in RNA substrates that would otherwise block progression of the exosome's exonucleases
Recruitment function: RRP-3 may help target the exosome to specific RNA substrates through protein-protein interactions
Processivity enhancement: The helicase activity likely facilitates continuous degradation by preventing stalling at structured regions
Evidence from yeast supports this model, as mutations in both Dob1p and Rrp4p (an exosome component) show strong synergistic growth inhibition, suggesting they function in the same pathway . Experimental approaches to investigate this coordination include:
Co-immunoprecipitation to identify physical interactions between RRP-3 and exosome components
In vitro reconstitution of RRP-3 with exosome complexes to measure enhanced RNA degradation activity
Pulse-chase labeling of pre-rRNA to track processing defects in rrp-3 mutants
Specific RNA processing events that likely require RRP-3 include 3' processing of 5.8S rRNA from its 7S precursor and degradation of the 5' external transcribed spacer (ETS) region of pre-rRNA .
What is the relationship between RRP-3 and other RNA surveillance mechanisms in Neurospora crassa?
RRP-3 is likely integrated with multiple RNA surveillance mechanisms in N. crassa:
Nonsense-mediated decay (NMD): RRP-3 may help unwind structured regions to facilitate degradation of transcripts with premature termination codons
Nuclear RNA quality control: As a putative component of TRAMP (Trf4/Air2/Mtr4 polyadenylation) complex based on homology to yeast Mtr4p, RRP-3 could target aberrant RNAs for degradation
Antiviral RNAi pathways: RRP-3 shows coordinated upregulation with Dicer proteins (DCL-1, DCL-2) and Argonaute (QDE-2) during viral infection
This integration creates a comprehensive RNA surveillance network that protects genome integrity. Notably, N. crassa has advanced genome defense mechanisms like Repeat-Induced Point mutation (RIP) that successfully prevents selfish DNA from gaining a foothold . RRP-3 may complement these DNA-level defenses by providing RNA-level surveillance.
Methodologically, the relationship between these mechanisms can be studied by creating double mutants (e.g., rrp-3Δ with qde-2Δ) and assessing synthetic phenotypes, or by analyzing changes in the small RNA profile in rrp-3 mutants to identify specific RNA species affected by RRP-3 deficiency.
How can molecular dynamics simulations enhance our understanding of RRP-3 activity?
Molecular dynamics (MD) simulations offer powerful tools for investigating RRP-3's mechanism of action at atomic resolution. Based on approaches used for related helicases , the following methodology can be applied:
Structural modeling: Generate a homology model of RRP-3 based on crystal structures of related DEAD-box helicases
Energy minimization: Refine the model through energy minimization in explicit solvent
Equilibrium simulations: Perform microsecond-scale MD simulations of RRP-3 in different states:
Apo (unbound) state
ATP-bound state
ADP-bound state
RNA-bound state with different substrates
Key analyses should include:
Conformational changes between open and closed states
Hydrogen bond networks stabilizing nucleotide binding
RNA binding cleft dynamics
Water and ion interactions at the active site
Recent studies on similar helicases have employed sophisticated MD techniques on the microsecond timescale, revealing that "ATP hydrolysis reaction was not explicitly analyzed here, but has been addressed in the literature for the PcrA helicase and for other motor proteins. The present work is not aimed at giving a mechanistic understanding of hydrolysis itself, but rather at providing an insight on the system's behavior after the hydrolysis reaction has occurred."
These simulations can guide experimental design by identifying key residues for mutagenesis and predicting how specific mutations might affect helicase activity.
What are the optimal conditions for expressing and purifying recombinant RRP-3?
Based on standard protocols for related DEAD-box helicases and commercial availability information , optimal expression and purification of recombinant RRP-3 typically involves:
Expression system options:
E. coli BL21(DE3) with pET-based vectors for high-yield bacterial expression
Baculovirus-infected insect cells for eukaryotic post-translational modifications
Native expression in N. crassa with epitope tags for authentic processing
Purification protocol:
Cell lysis under native conditions using buffer containing:
50 mM Tris-HCl (pH 7.5-8.0)
300-500 mM NaCl
5-10% glycerol
1-5 mM DTT or β-mercaptoethanol
Protease inhibitor cocktail
Initial capture using affinity chromatography:
His-tag: Ni-NTA or TALON resin
GST-tag: Glutathione-Sepharose
FLAG-tag: Anti-FLAG M2 affinity gel
Secondary purification:
Ion exchange chromatography (typically Q-Sepharose)
Size exclusion chromatography (Superdex 200)
Quality control:
For functional studies, it's crucial to verify that the recombinant protein retains ATPase activity, which can be measured using colorimetric assays that detect inorganic phosphate release.
What assays can be used to measure RRP-3 helicase activity in vitro?
Several robust assays can quantitatively measure the RNA helicase activity of purified RRP-3:
1. Fluorescence-based unwinding assays:
Substrate: RNA duplex with fluorophore-quencher pair
Principle: Fluorescence increases upon strand separation
Advantages: Real-time monitoring, high sensitivity
Protocol outline:
Prepare RNA substrate with 5'-fluorescein and 3'-quencher
Mix with RRP-3 in helicase buffer (typically 20 mM HEPES pH 7.5, 50 mM KCl, 5 mM MgCl₂, 1 mM DTT)
Initiate reaction by adding ATP (1-2 mM)
Monitor fluorescence increase (excitation: 495 nm, emission: 520 nm)
Calculate unwinding rates from initial velocity
2. Gel-based unwinding assays:
Substrate: ³²P-labeled RNA duplex
Principle: Separation of single-stranded from double-stranded RNA on native PAGE
Protocol outline:
Incubate labeled duplex with RRP-3 and ATP
Stop reaction at various timepoints with proteinase K and EDTA
Resolve products on 12% native PAGE
Quantify bands by phosphorimaging
3. ATPase activity assays (coupled to helicase function):
Malachite green assay for inorganic phosphate
NADH-coupled spectrophotometric assay
[γ-³²P]ATP hydrolysis with thin-layer chromatography
4. RNA binding assays:
Electrophoretic mobility shift assay (EMSA)
Fluorescence anisotropy
Surface plasmon resonance (SPR)
When designing these assays, it's important to include proper controls:
No-ATP control to confirm ATP dependence
Heat-denatured RRP-3 control
Non-hydrolyzable ATP analogs (e.g., AMP-PNP) to distinguish binding from unwinding
How can CRISPR-Cas9 genome editing be optimized for studying rrp-3 function in Neurospora crassa?
CRISPR-Cas9 genome editing in N. crassa requires specialized protocols due to this organism's unique biology, including its genome defense mechanisms like RIP (Repeat-Induced Point mutation) . An optimized workflow includes:
1. sgRNA design considerations:
Target sequence must contain PAM site (NGG for SpCas9)
Avoid sequences with homology to other regions (to prevent off-target effects)
Design sgRNAs targeting both 5' and 3' regions of rrp-3 coding sequence
Use N. crassa codon optimization for guide expression
2. Delivery methods:
Transformation of protoplasts with RNP complexes (pre-assembled Cas9 protein + sgRNA)
Plasmid-based delivery using N. crassa-compatible promoters (e.g., trpC promoter)
Homology-directed repair templates for precise modifications:
Knock-in of fluorescent tags (e.g., GFP, mCherry)
Introduction of point mutations to study specific domains
Conditional expression systems (e.g., Qa-2 promoter for inducible expression)
3. Screening strategies:
PCR-based genotyping with primers flanking the target site
Restriction fragment length polymorphism (RFLP) analysis if edit creates/destroys restriction site
Sanger sequencing of PCR products
Western blotting to confirm protein modification/knockout
4. Phenotypic analysis:
Growth rate measurements on different media
Microscopic analysis of developmental stages
RNA processing defects using Northern blotting
Efficiency enhancement strategies:
Co-transformation with selectable markers (e.g., hygromycin resistance)
Use of N. crassa strains deficient in non-homologous end joining (e.g., Δmus-51 or Δmus-52)
Optimization of homology arm length (typically 500-1000 bp for efficient homologous recombination)
What approaches can be used to investigate the role of RRP-3 in ribosome biogenesis in Neurospora crassa?
Investigating RRP-3's role in ribosome biogenesis requires a combination of molecular, biochemical, and microscopic techniques:
1. Analysis of pre-rRNA processing:
Northern blotting with probes specific for different pre-rRNA regions
Primer extension analysis to map precise 5' ends of processing intermediates
Pulse-chase labeling with [³H]uridine or [³²P]orthophosphate followed by gel electrophoresis
qRT-PCR to quantify specific pre-rRNA species
2. Polysome profiling:
Ultracentrifugation of cell lysates through sucrose gradients
Monitoring A254 profiles to detect:
Free 40S and 60S ribosomal subunits
80S monosomes
Polysome fractions
Northern blotting of gradient fractions to track specific rRNAs
Western blotting to detect RRP-3 association with specific fractions
3. Protein-RNA interactions:
RNA immunoprecipitation (RIP) using epitope-tagged RRP-3
Cross-linking and immunoprecipitation (CLIP) to identify direct binding sites
In vitro binding assays with purified components
4. Co-localization studies:
Fluorescence microscopy of GFP-tagged RRP-3
Co-staining for nucleolar markers
Electron microscopy to visualize ribosome biogenesis compartments
5. Genetic interaction analysis:
Creation of double mutants with other pre-rRNA processing factors
Synthetic genetic array analysis to identify genetic interactions
Complementation tests with known rRNA processing mutants
Based on yeast studies, one would expect rrp-3 mutants to show specific defects in 18S rRNA synthesis , which can be detected as an accumulation of precursor rRNAs and a reduction in mature 18S rRNA levels. The precise role can be further elucidated by examining which pre-rRNA processing steps are blocked in rrp-3 mutants compared to wild-type strains.