Recombinant Sabia virus RNA-directed RNA polymerase L (L), partial

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Sabia Virus RNA-Directed RNA Polymerase L (L), Partial

The Recombinant Sabia virus RNA-directed RNA polymerase L (L), partial refers to a genetically engineered version of the L protein from the Sabia virus, which is a critical component in the replication and transcription of the viral genome. The L protein, also known as the RNA-dependent RNA polymerase (RdRp), is essential for the lifecycle of arenaviruses, including the Sabia virus. This enzyme catalyzes the synthesis of new viral RNA strands from an RNA template, making it a crucial target for antiviral drug development.

Structure and Function of the L Protein

The L protein of the Sabia virus is a large protein consisting of approximately 2212 amino acids with a molecular weight of about 253 kDa. It contains two major domains: the RNA-dependent RNA polymerase and the endonuclease domains . The RNA-dependent RNA polymerase domain is responsible for replicating the viral genome and transcribing viral genes, while the endonuclease domain may play a role in processing viral RNA.

DomainFunction
RNA-dependent RNA polymeraseReplication and transcription of viral RNA
EndonucleaseProcessing of viral RNA

Role in Viral Replication

During the replication cycle of the Sabia virus, the L protein interacts with other viral proteins, such as the nucleoprotein (NP) and the matrix protein Z. The NP helps in protecting the viral RNA from host RNases and facilitates its interaction with the L protein for replication . The Z protein regulates the activity of the L protein by binding at the product exit site, controlling the initiation and termination of RNA synthesis .

Antiviral Targeting

The L protein is a promising target for antiviral therapies due to its highly conserved structure across arenaviruses. Targeting the L protein could lead to broad-spectrum antiviral activity with a reduced risk of drug resistance . Additionally, since vertebrate cells lack RNA-dependent RNA polymerase activity, drugs targeting this enzyme are less likely to cause off-target effects .

Research Findings and Implications

Recent studies have highlighted the potential of targeting RNA-dependent RNA polymerases in various RNA viruses, including arenaviruses. The development of inhibitors for RdRp has shown promise in treating diseases caused by RNA viruses, such as COVID-19 and Hepatitis C . For the Sabia virus, understanding the structure and function of the L protein can guide the design of specific antiviral compounds.

Viral TargetAntiviral StrategyPotential Outcome
L Protein (RdRp)Inhibition of RdRp activityReduced viral replication and disease severity
Z Protein InteractionDisruption of Z-L interactionEnhanced antiviral efficacy by promoting continuous RNA synthesis

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to settle the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, provided as a guideline for your use.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
LRNA-directed RNA polymerase L; Protein L; EC 2.7.7.48; EC 3.1.-.-; Large structural protein; Replicase; Transcriptase
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Protein Length
Partial
Purity
>85% (SDS-PAGE)
Species
Sabia mammarenavirus (isolate Human/Brasil/SPH114202/1990) (SABV) (Sabi mammarenavirus)
Target Names
L
Uniprot No.

Target Background

Function
This RNA-dependent RNA polymerase is responsible for the replication and transcription of the viral RNA genome. During transcription, it synthesizes four subgenomic RNAs and ensures their capping through a cap-snatching mechanism utilizing cellular capped pre-mRNAs as primers. The 3'-ends of subgenomic mRNA molecules are heterogeneous and lack polyadenylation. The replicase function directs the synthesis of antigenomic and genomic RNA, which are encapsidated and non-capped. The use of the same enzyme for both transcription and replication necessitates precise coordination, potentially regulated by proteins N and Z in a dose-dependent manner.
Database Links

KEGG: vg:3077250

Protein Families
Arenaviridae RNA polymerase family
Subcellular Location
Virion. Host cytoplasm.

Q&A

What is the structure and function of Sabia virus RNA-dependent RNA polymerase L?

The Sabia virus RNA-dependent RNA polymerase L (L protein) is a large multifunctional protein that serves as the catalytic core of the viral replication complex. Structurally, like other viral RdRps, the L protein adopts a characteristic "right hand" configuration consisting of fingers, palm, and thumb subdomains that form a cupped shape essential for its catalytic activity . The palm subdomain contains the catalytic site with conserved aspartate residues that coordinate divalent metal ions (typically Mg²⁺) required for nucleotide addition during RNA synthesis .

The L protein of arenaviruses, including SABV, contains highly conserved catalytic domains that are responsible for multiple enzymatic activities beyond RNA polymerization. These include an endonuclease domain in the N-terminal region and a cap-binding domain, which together enable cap-snatching mechanisms for viral mRNA synthesis . The central RdRp domain catalyzes both genomic replication and viral transcription, while additional domains may be involved in protein-protein interactions with other viral components .

Functionally, the L protein is indispensable for viral replication, as it catalyzes the synthesis of both genomic and antigenomic viral RNA, as well as viral mRNAs. Its error-prone nature contributes to the genetic variability of RNA viruses, which is a key factor in their adaptability and evolution . Understanding the structure-function relationships of SABV L protein provides critical insights for the development of targeted antiviral strategies.

How do the conserved motifs in SABV L protein contribute to its catalytic activity?

The SABV L protein, like other viral RdRps, contains seven conserved structural motifs (designated A through G) that play critical roles in catalysis despite sequence divergence across viral families . Motifs A to E are located within the palm subdomain, while motifs F and G reside in the fingers subdomain . These motifs are spatially arranged to form the active site and coordinate essential functions during RNA synthesis.

Motif A contains the conserved DX₂₋₄D sequence that binds divalent metal ions essential for catalysis . Motif C houses the highly conserved GDD motif (or variant forms in some viruses) that is crucial for positioning the metal ions at the active site . These two motifs align spatially to form the RNA recognition motif (RRM) that coordinates the catalytic reaction . Motif B contributes to nucleotide selection and binding, while motifs D and E are involved in structural integrity and proper positioning of the template RNA .

In arenaviruses including SABV, an additional motif H may be present in the thumb subdomain, contributing to template binding and positioning . Mutations in these conserved motifs typically result in significant reductions in polymerase activity or altered fidelity, highlighting their critical importance in the catalytic mechanism. Experimental approaches to study these motifs typically involve site-directed mutagenesis followed by enzymatic activity assays using purified recombinant protein .

What expression systems are most effective for producing functional recombinant SABV L protein?

Expression of functional recombinant SABV L protein presents significant challenges due to its large size (approximately 250 kDa), potential toxicity to host cells, and complex folding requirements. Several expression systems have been utilized for arenavirus L proteins, each with distinct advantages and limitations for research applications.

Bacterial expression systems (typically E. coli) offer cost-effectiveness and high protein yields but often result in insoluble or improperly folded L protein due to the lack of eukaryotic chaperones and post-translational modifications. To overcome these limitations, researchers frequently express functional domains separately rather than the full-length protein when using bacterial systems . Addition of solubility tags (such as MBP, SUMO, or GST) can improve solubility, though activity may still be compromised.

Insect cell expression systems (such as Sf9 or High Five cells with baculovirus vectors) represent a preferred approach for full-length L protein expression, as they provide eukaryotic folding machinery while maintaining high protein yields . Mammalian cell expression systems (typically HEK293T or BHK cells) offer the most authentic environment for proper folding and potential post-translational modifications but generally yield lower protein amounts. These systems are particularly valuable when studying protein-protein interactions with other viral or host factors .

Cell-free expression systems have emerged as an alternative approach that can circumvent potential toxicity issues. Regardless of the expression system chosen, purification typically involves affinity chromatography (using His, FLAG, or other tags), followed by size exclusion chromatography to obtain homogeneous protein preparations suitable for enzymatic and structural studies .

What are the key protein-protein interactions between SABV L protein and other viral components?

The SABV L protein engages in critical protein-protein interactions that regulate viral RNA synthesis and replication complex assembly. Among the most significant interactions is that between the L protein and the viral matrix protein Z, which has evolved specifically in New World arenaviruses like SABV . This interaction involves a distinct interface that differs from Old World arenaviruses, suggesting virus-specific regulatory mechanisms that could be selectively targeted for antiviral development.

The L protein also forms essential interactions with the viral nucleoprotein (NP), which serves multiple functions in the replication cycle. The NP-L interaction facilitates the recognition of viral RNA templates and the assembly of functional replication complexes . The C-terminal domain of NP exhibits exonuclease activity that may influence RNA synthesis fidelity and potentially counteract cellular antiviral responses by degrading immunostimulatory RNAs . Methodologically, these interactions can be studied through co-immunoprecipitation assays, yeast two-hybrid systems, or bimolecular fluorescence complementation in living cells.

Additionally, similar to other viral RdRps, the SABV L protein likely interacts with viral RNA structures, including promoter elements at the genomic termini that direct replication initiation. Structure prediction methods and sequence alignments suggest that these interactions may involve specific regions within the fingers and thumb domains of the L protein . Minigenome systems, in which reporter genes are flanked by viral regulatory sequences, provide valuable tools for dissecting the functional significance of these RNA-protein interactions in a cellular context without requiring work with infectious virus .

How can structural insights into SABV L protein be leveraged for rational antiviral drug design?

Structural analysis of the SABV L protein offers significant opportunities for rational antiviral drug design targeting specific functional domains. The highly conserved catalytic core of the RdRp domain presents an attractive target for broad-spectrum antivirals that could inhibit multiple arenaviruses . Computational approaches such as structure-based virtual screening can identify small molecules that bind to the active site or allosteric regulatory sites of the L protein.

The template channel of the L protein represents another promising target for inhibitor development, as demonstrated for other pathogenic viruses from families such as Flaviviridae . This channel, formed by residues from multiple subdomains, guides the template RNA to the active site during replication. Small molecules designed to occlude this channel could block template binding and effectively inhibit viral replication. Crystallographic or cryo-EM studies of L protein in complex with potential inhibitors provide crucial insights into binding modes and guide medicinal chemistry optimization efforts .

Protein-protein interaction surfaces, particularly those between L and Z proteins or L and NP, offer targets for virus-specific inhibition strategies . Since these interactions have evolved differently in New World arenaviruses compared to Old World arenaviruses, compounds targeting these interfaces might show selectivity for SABV and related viruses. High-throughput screening assays based on fluorescence resonance energy transfer (FRET) or alpha-screen technology can identify compounds that disrupt these critical protein-protein interactions.

What methodological approaches are most effective for studying SABV L protein enzymatic activities in vitro?

Investigating the enzymatic activities of recombinant SABV L protein requires sophisticated biochemical assays that can monitor various aspects of polymerase function. RNA synthesis activity can be assessed using template-dependent polymerase assays with radiolabeled or fluorescently labeled nucleotides, allowing quantification of both de novo initiation and elongation activities . Gel-based assays and scintillation counting provide standard readouts, while more advanced techniques such as real-time monitoring using fluorescence polarization offer kinetic insights.

Studying the cap-snatching mechanism of the L protein endonuclease domain requires specialized assays that monitor the cleavage of capped RNA substrates . These typically involve synthetic capped RNA oligonucleotides and detection of cleavage products by gel electrophoresis or HPLC analysis. The protein-primed initiation mechanism, potentially involving VPg-like proteins as observed in some RNA viruses, can be assessed through nucleotidylation assays that monitor the covalent addition of nucleotides to protein primers .

For high-resolution mechanistic studies, pre-steady-state kinetic approaches using rapid quench-flow or stopped-flow techniques allow dissection of individual steps in the catalytic cycle . Single-molecule approaches, though technically challenging, provide unique insights into polymerase dynamics and processivity that are masked in bulk assays. Additionally, thermal shift assays and isothermal titration calorimetry can evaluate inhibitor binding and aid in characterizing structure-activity relationships for potential antiviral compounds .

How does the SABV L protein participate in viral immune evasion strategies?

The SABV L protein likely contributes to viral immune evasion through multiple mechanisms that extend beyond its primary role in viral replication. Like other viral RdRps, the L protein may interfere with cellular antiviral pathways through direct protein-protein interactions with host factors . For example, some viral polymerases can enter the nucleus and disrupt pre-mRNA splicing by targeting central processing factors such as Prp8, thereby inhibiting host gene expression without shutting off cellular transcription and translation machinery entirely .

The L protein's enzymatic activities may also contribute to immune evasion by limiting the production of viral pathogen-associated molecular patterns (PAMPs) that trigger innate immune responses. The cap-snatching mechanism not only enables viral mRNA translation but also prevents the accumulation of uncapped viral RNAs that could be detected by pattern recognition receptors like RIG-I . Additionally, the L protein works in concert with the viral nucleoprotein, which possesses exonuclease activity in its C-terminal domain that may degrade immunostimulatory double-stranded RNA intermediates produced during replication .

Research approaches to study these immune evasion mechanisms include co-immunoprecipitation followed by mass spectrometry to identify host interaction partners, reporter assays measuring interferon pathway activation in the presence of wild-type or mutant L proteins, and transcriptome analysis to assess global changes in host gene expression during infection . Comparative studies between SABV L protein and other arenavirus polymerases can highlight conserved and unique strategies employed by different viruses to counteract host defenses .

What biosafety considerations are essential when working with recombinant SABV L protein?

Standard laboratory safety practices should include the use of biological safety cabinets, appropriate personal protective equipment, and validated decontamination procedures. Institutional biosafety committee approval is mandatory before initiating work with SABV components. Additionally, researchers should be familiar with dual-use research of concern (DURC) guidelines, as studies enhancing polymerase activity or fidelity could potentially increase viral fitness . Documentation, training, and regular safety audits are essential components of a comprehensive biosafety program for work with recombinant viral proteins from high-containment pathogens .

What are the most effective strategies for developing minigenome systems to study SABV L protein function?

Minigenome systems represent powerful tools for investigating SABV L protein function in a cellular context without requiring work with infectious virus. Development of an effective SABV minigenome system involves several key components and methodological considerations that enable precise analysis of polymerase activity and regulation.

The core components of a SABV minigenome system include: (1) a reporter construct containing a reporter gene (typically luciferase or fluorescent protein) flanked by the viral untranslated regions (UTRs) that contain promoter elements recognized by the L protein; (2) expression plasmids for the L protein and nucleoprotein (NP), which together form the minimal replication complex; and (3) optionally, plasmids expressing additional viral proteins such as Z to study regulatory interactions . The reporter construct is typically designed in either a genomic (negative-sense) or antigenomic (positive-sense) orientation, with the latter often yielding higher activity due to direct transcription of the reporter gene upon transfection.

Methodologically, minigenome systems are established through transfection of the component plasmids into appropriate cell lines (typically human or rodent cells permissive for arenavirus replication), followed by measurement of reporter activity as a readout of L protein-mediated transcription . Time-course experiments can distinguish between transcription of the input plasmid and authentic L protein-dependent RNA synthesis. Mutations in the L protein can be readily introduced to assess their impact on polymerase function, making minigenome systems invaluable for structure-function analyses .

Advanced applications include the incorporation of viral UTR mutations to map sequence elements required for L protein recognition, competitive inhibition assays to screen for polymerase inhibitors, and adaptation for high-throughput screening platforms to identify host factors that influence L protein activity .

How might cryo-EM and AI-based structure prediction advance our understanding of SABV L protein function?

Recent advances in cryo-electron microscopy (cryo-EM) and artificial intelligence-based structure prediction methods (such as AlphaFold2 and RoseTTAFold) are revolutionizing our ability to study complex viral proteins like the SABV L polymerase. These technological developments offer unprecedented opportunities to elucidate structural details that have remained elusive due to difficulties in crystallizing large, flexible viral proteins.

Cryo-EM has emerged as a powerful tool for resolving structures of viral polymerases in different functional states, including in complex with RNA templates, nucleotides, and other viral proteins . This technique can capture conformational changes associated with the catalytic cycle and identify potential allosteric sites for drug targeting. For SABV L protein, cryo-EM could reveal the spatial arrangement of its multiple functional domains and how they coordinate during viral RNA synthesis, providing insights not attainable through crystallography alone .

AI-based structure prediction methods offer complementary approaches that can rapidly generate structural models even in the absence of experimental data. For SABV L protein, these methods can predict structures of individual domains and their interfaces, guide experimental design, and facilitate virtual screening for potential inhibitors . The integration of predicted structures with limited experimental data (such as cross-linking mass spectrometry or hydrogen-deuterium exchange) can yield comprehensive structural models that inform rational drug design efforts .

What are the prospects for developing broad-spectrum antivirals targeting conserved features of arenavirus L proteins?

The development of broad-spectrum antivirals targeting conserved features of arenavirus L proteins represents a promising approach to address the threat posed by SABV and related pathogens. The highly conserved catalytic core of the RdRp domain, particularly motifs A and C containing the catalytic aspartate residues, presents an attractive target for inhibitors that could act against multiple arenaviruses .

Nucleoside analogs that are incorporated into the growing RNA chain and cause chain termination or lethal mutagenesis have shown efficacy against various RNA viruses and could potentially inhibit SABV L protein . Non-nucleoside inhibitors targeting allosteric sites that affect conformational dynamics required for catalysis also offer promising avenues for broad-spectrum activity . The conserved template channel structure, which has been successfully targeted in flaviviruses, represents another potential site for developing inhibitors that block RNA binding .

Structure-based drug design approaches can identify compounds that interact with conserved pockets within the L protein. Virtual screening of compound libraries against structural models, followed by biochemical and cell-based validation assays, provides an efficient pipeline for inhibitor discovery . Additionally, repurposing of existing antivirals targeting related viral polymerases could accelerate the development timeline for anti-SABV therapeutics .

Challenges in this approach include the potential for viral resistance through mutations in the polymerase gene and the need to balance broad-spectrum activity with sufficient potency against specific viral targets. Combination therapies targeting multiple viral functions or different regions of the L protein may mitigate resistance concerns and enhance therapeutic efficacy .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.