Recombinant Satsuma dwarf virus RNA1 polyprotein

Shipped with Ice Packs
In Stock

Description

Introduction to Recombinant Satsuma Dwarf Virus RNA1 Polyprotein

Recombinant Satsuma dwarf virus (SDV) RNA1 polyprotein refers to a genetically engineered form of the polyprotein encoded by the RNA1 segment of the Satsuma dwarf virus. SDV is a plant virus that affects citrus plants, causing dwarfism and other growth abnormalities. The RNA1 segment of SDV encodes a large polyprotein that is crucial for viral replication and other functions.

Structure and Function of SDV RNA1 Polyprotein

The RNA1 of SDV encodes a polyprotein that is likely cleaved into several functional proteins involved in replication. These proteins include a putative protease cofactor, helicase, and RNA-dependent RNA polymerase (RdRp) . The RdRp is essential for the replication of RNA viruses, as it catalyzes the synthesis of new viral RNA strands.

Key Features of SDV RNA1 Polyprotein:

  • Length and Composition: The RNA1 segment is approximately 7,581 nucleotides long, excluding the poly(A) tail, and encodes a large polyprotein .

  • Functional Domains: The polyprotein contains domains for protease, helicase, and RdRp activities, which are crucial for viral replication .

  • Homology: The RdRp domain shows homology with other viral RdRps, particularly those of comoviruses and nepoviruses, although SDV does not cluster with these groups phylogenetically .

Recombinant Expression and Applications

Recombinant expression of the SDV RNA1 polyprotein involves using genetic engineering techniques to produce this protein in a host system, such as bacteria or insect cells. This approach allows for the study of the protein's structure, function, and interactions in a controlled environment.

Potential Applications:

  • Vaccine Development: Understanding the structure and function of viral proteins can aid in the development of vaccines or therapeutic agents against SDV.

  • Biotechnology Research: Recombinant proteins can be used to study viral replication mechanisms and to develop new biotechnological tools.

Sequence Analysis:

  • Nucleotide Sequence: The 3'-terminal region of SDV RNA1 contains a single open reading frame (ORF) of 2868 nucleotides and a non-coding region of 248 nucleotides upstream of the poly(A) tail .

  • Amino Acid Sequence Homology: The RdRp domain shows significant homology with RdRps from other viruses, such as comoviruses and nepoviruses, but SDV does not cluster with these groups .

Phylogenetic Analysis:

  • SDV is distinct from comoviruses and nepoviruses, despite sharing some sequence homologies with these groups .

Table: Sequence Homologies of SDV RdRp with Other Viruses

VirusSequence Homology (%)
CPMV28
RCMV27
CPSMV26
GFLV25
TBRV24
GCMV24
HPV20

References:

  • Iwanami, T., et al. (1996). Nucleotide sequence of the 3'-terminal region of RNA1 of Satsuma dwarf virus. Annals of the Phytopathological Society of Japan, 62(1), 4–10.

  • Nakazono-Nagaoka, E., et al. (2004). A new virus related to Satsuma dwarf virus: the nucleotide sequence of the 3'-terminal regions of RNAs 1 and 2 of Hyuganatsu virus. Archives of Virology, 149(11), 2241–2253.

  • Martin, R. R., et al. (2007). Identification, characterization, and detection of Black raspberry necrosis virus. Phytopathology, 97(1), 44–53.

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your preferred format in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires advance notice 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 collect 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%, which can serve as a reference.
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 formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The specific tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its inclusion.
Synonyms
RNA1 polyprotein; P1
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1401-2081
Protein Length
full length protein
Species
Satsuma dwarf virus (isolate Satsuma mandarin/Japan/S-58/1977) (SDV)
Target Protein Sequence
AELDAAILDRFSIPISECRKELTPMTTRLGYVVGQYPRALRKTSIVPSIIHDNLWRKPET EPTILGKIDDRSPFPYDPYATIGEKFVQEVGPIDLSVGSDASLVVANIGSSWKAVGKPQC PTVLTWEVAINGDAAIPYCERLPLSTSEGYPDSIQRNFGEKGKKRFFDLKGENVRVPTPA LMEELEVLERELQKEEVCLTCINTACAKDEKTAPKKVRVQPKTRIFEILPFQINIIIRRY LMFWMQLLMVAHDELPSKVGINVYSESWDRLLGRHTRLANHFTGDYSGFDTSTPRVLVYA IIDKINELADDGEVNQRTRRNIIRFVLNRYLISDGVLYEIHGGTPSGFAPTVMINSVVNE FYLKWSWIGLLKEAGYANQATLYAFHEATEISLYGDDNFVSVATPVASVYNLTTISNFLG RIGVKLGDGAKTGTIKPFIPLEEVDFLKRQFVADSGSTAILCPLKKISIEERLFYVRGGQ DEIAALELNIATALCEAFFHGKEYFSFLEGKIIEAMRKSGVALSRPLPTMESVRAWYMSQ RGNTKIRSPSFEGLGTMSGILNIGLAEARSVGGVACFSGIEFRGRSDDHLMVIPTYIPGG WRTKQQQTYISFVRDSEKMAQVIKRVAHFSTVVATDKSMAYLVAICIAYSRGSISRMEVR CHVQNLKVAEMLLCNQICNFL
Uniprot No.

Target Background

Function

Picornain 3C-like protease is a thiol protease that likely cleaves the polyprotein.

Database Links

KEGG: vg:993327

Subcellular Location
Host membrane; Multi-pass membrane protein.

Q&A

What is the genomic organization of Satsuma dwarf virus?

Satsuma dwarf virus possesses a bipartite genome consisting of RNA1 (6795 nucleotides) and RNA2 (5345 nucleotides). Each RNA segment encodes a polyprotein that undergoes post-translational processing. The genome organization shows similarities to members of the como-, faba-, and nepoviruses, but with distinct features that set SDV apart taxonomically. RNA1 encodes proteins primarily involved in viral replication, while RNA2 encodes structural proteins including two coat proteins, which is atypical compared to nepoviruses that typically have a single coat protein . The genomic organization reflects evolutionary adaptations specific to the virus's replication strategy and host interactions, making it an interesting model for studying viral evolution and host adaptation mechanisms.

What functional domains are present in the SDV RNA1 polyprotein?

The SDV RNA1 polyprotein contains multiple functional domains that are essential for viral replication and infection. Based on sequence analyses and comparison with related viruses, the RNA1 polyprotein contains domains for helicase activity, a viral genome-linked protein (VPg), protease, and RNA-dependent RNA polymerase (RdRp) . These domains work in concert to facilitate viral RNA replication, with the helicase unwinding double-stranded RNA intermediates, the protease processing the polyprotein into functional units, and the RdRp synthesizing new viral RNA strands. Understanding these domains and their interactions is crucial for developing targeted antiviral strategies and for gaining insights into the molecular mechanisms underlying viral replication cycles.

How does SDV RNA1 polyprotein compare to similar proteins in related viruses?

Comparative analyses reveal both similarities and significant differences between SDV RNA1 polyprotein and those of related viruses. There is extensive amino acid sequence similarity in the N-terminal regions of proteins encoded by RNA1 and RNA2 of SDV, a feature previously reported only for tomato ringspot nepovirus . Phylogenetic analysis of the RNA polymerase region places SDV apart from como-, faba-, and nepoviruses, suggesting a distinct evolutionary lineage. When compared with Hyuganatsu virus (HV), the partial RNA-dependent RNA polymerase region in RNA1 shows 78.3-84.0% amino acid sequence identity . Similarly, the Strawberry mottle virus (SMoV) polyprotein of RNA1 shares its closest similarity with RNA1 of SDV, particularly in regions with identities to helicase, viral genome-linked protein, protease and polymerase (RdRp) . These comparative analyses are essential for understanding the evolutionary relationships among plant viruses and for refining viral taxonomy.

What experimental approaches are most effective for expressing recombinant SDV RNA1 polyprotein?

Recombinant expression of SDV RNA1 polyprotein presents several technical challenges due to its large size and multiple functional domains. Current successful approaches include expression in E. coli systems using His-tag fusion constructs, as demonstrated with the commercially available recombinant protein (residues 1401-2081) . For optimal expression, several methodological considerations are critical: (1) Codon optimization based on the expression host is essential for improving translation efficiency; (2) Expression of individual functional domains rather than the entire polyprotein often yields better results for structural and functional studies; (3) Lowering induction temperature (16-18°C) can enhance proper folding; and (4) Addition of solubility-enhancing fusion tags such as MBP (maltose-binding protein) or SUMO can improve protein solubility and yield. Depending on the research objectives, baculovirus-insect cell expression systems may provide better post-translational modifications for certain structural or functional studies of viral polyproteins.

How can researchers effectively study the proteolytic processing of SDV RNA1 polyprotein?

Studying the proteolytic processing of SDV RNA1 polyprotein requires a multi-faceted approach. A time-course analysis using pulse-chase labeling with radioactive amino acids followed by immunoprecipitation can reveal the sequential processing events and intermediate products. To identify specific cleavage sites, site-directed mutagenesis of predicted protease recognition sequences can be performed, followed by in vitro translation assays to observe changes in processing patterns. Mass spectrometry analysis of purified viral proteins from infected plant material provides precise identification of cleavage sites and post-translational modifications. For in vitro studies, recombinant virus-encoded protease can be expressed and purified to test its activity on synthetic peptides representing potential cleavage sites. Additionally, time-resolved structural studies using X-ray crystallography or cryo-electron microscopy of the polyprotein at various stages of processing can provide insights into conformational changes during maturation. These combined approaches allow for comprehensive characterization of the proteolytic processing pathway.

What are the challenges in determining the three-dimensional structure of SDV RNA1 polyprotein domains?

Structural determination of SDV RNA1 polyprotein domains faces several significant challenges. The polyprotein's large size (derived from a 6795 nt RNA1) makes crystallization of the full-length protein extremely difficult. Individual domains often require specific buffer conditions and stabilizing agents to maintain their native conformation when expressed separately. Some domains, particularly the RdRp, may require cofactors or substrates for stabilization during structural studies. Membrane-associated domains present additional purification challenges and may require specialized approaches such as lipid nanodiscs or detergent micelles for structural studies. Intrinsically disordered regions within the polyprotein contribute to conformational heterogeneity, complicating both crystallization and cryo-EM approaches. To overcome these challenges, researchers should consider a divide-and-conquer approach, focusing on well-defined functional domains separately, employing a combination of X-ray crystallography, nuclear magnetic resonance (NMR) spectroscopy for smaller domains, and cryo-electron microscopy for larger assemblies. Computational approaches including homology modeling based on related viral proteins can provide preliminary structural insights to guide experimental designs.

What techniques are most effective for studying SDV RNA1 polyprotein interactions with host factors?

Studying SDV RNA1 polyprotein interactions with host factors requires a combination of biochemical, molecular, and imaging techniques. Yeast two-hybrid (Y2H) screening provides an initial high-throughput approach to identify potential protein-protein interactions between viral polyprotein domains and host proteins. Results from Y2H should be validated using co-immunoprecipitation (Co-IP) assays with either antibodies against the viral protein or epitope-tagged versions expressed in plant cells. For in vivo verification, bimolecular fluorescence complementation (BiFC) allows visualization of protein interactions within living cells, providing spatial information about where these interactions occur. Proximity ligation assays (PLA) offer high sensitivity for detecting protein interactions with minimal disruption to cellular structures. For a comprehensive understanding of the virus-host interactome, immunoprecipitation followed by mass spectrometry (IP-MS) can identify multiple interaction partners simultaneously. To study the functional consequences of these interactions, virus-induced gene silencing (VIGS) or CRISPR-Cas9-mediated knockout of identified host factors can reveal their role in viral replication. Complementing these approaches with confocal microscopy using fluorescently tagged proteins helps track the temporal and spatial dynamics of these interactions during infection.

How can researchers effectively analyze the RNA-dependent RNA polymerase activity of SDV RNA1 polyprotein?

Analyzing the RNA-dependent RNA polymerase (RdRp) activity of SDV RNA1 polyprotein requires specialized assays to measure nucleic acid synthesis. In vitro RdRp assays using purified recombinant polymerase domain and template RNA provide a direct measure of polymerase activity. The reaction typically contains the purified RdRp, template RNA, nucleotides (including radiolabeled or modified nucleotides for detection), and appropriate buffers with magnesium or manganese ions as cofactors. Products can be analyzed by denaturing polyacrylamide gel electrophoresis followed by autoradiography or phosphorimaging. To study template specificity, researchers should test various RNA templates, including viral and non-viral sequences, to determine if the RdRp recognizes specific structural elements or sequences. Real-time monitoring of polymerase activity can be achieved using fluorescence-based assays with dye-labeled nucleotides. For in vivo studies, replicon systems based on SDV sequences can be developed, where replication is measured through reporter gene expression or quantification of replicon RNA levels. Site-directed mutagenesis of conserved motifs in the polymerase domain can identify residues critical for catalytic activity or template binding. Additionally, inhibitor screening assays can identify compounds that specifically target the viral RdRp, potentially leading to antiviral development.

What approaches are recommended for studying the role of SDV RNA1 polyprotein in viral pathogenesis?

Investigating the role of SDV RNA1 polyprotein in viral pathogenesis requires an integrated approach combining molecular virology, cell biology, and host response analysis. Infectious clones representing wild-type and mutant versions of SDV allow for precise manipulation of viral sequences. Mutations in specific domains of the RNA1 polyprotein can help identify regions critical for replication versus those involved in pathogenesis or host interactions. Differential transcriptomics and proteomics comparing host responses to wild-type versus mutant viruses can reveal pathways specifically triggered by functional RNA1 polyprotein. Subcellular localization studies using confocal microscopy with fluorescently tagged viral proteins help identify replication sites and virus-induced cellular remodeling. To understand tissue specificity, in situ hybridization and immunohistochemistry can track viral RNA and protein distribution throughout the infected plant. For host range studies, inoculation of various citrus varieties and related species with wild-type and mutant viruses can correlate RNA1 polyprotein features with host adaptation. Transgenic expression of individual domains of the polyprotein can determine if specific regions trigger pathogenesis-related responses independent of viral replication. Time-course studies monitoring viral accumulation, movement, and symptom development provide insights into the dynamic role of RNA1 polyprotein throughout the infection cycle.

How can structural information about SDV RNA1 polyprotein inform antiviral development?

Detailed structural characterization of SDV RNA1 polyprotein domains provides valuable templates for structure-based antiviral design. The RdRp domain represents a primary target for antiviral development, as inhibition of viral replication directly impacts virus propagation. Structural data allows identification of active sites and potential allosteric binding pockets that could be targeted by small-molecule inhibitors. Computational approaches such as molecular docking and virtual screening can utilize RdRp structures to identify compounds predicted to bind and inhibit polymerase activity. Crystal structures of the viral protease domain enable design of peptidomimetic inhibitors that block polyprotein processing, a strategy successfully employed against other viral proteases. Structure-activity relationship studies guided by solved protein structures can optimize lead compounds for improved binding affinity, specificity, and pharmacokinetic properties. Beyond direct antiviral development, structural knowledge of viral protein-host protein interfaces can inform the design of compounds that disrupt these essential interactions while minimizing effects on normal host cell functions. Although SDV primarily affects citrus crops rather than humans, the methodological approaches developed through studying its polyprotein structure can be applied to related human pathogens within the picorna-like virus superfamily .

What is the current understanding of the evolutionary relationships between SDV RNA1 polyprotein and other viral polyproteins?

Phylogenetic analysis places SDV in a unique position relative to other plant viruses, suggesting it represents a distinct evolutionary lineage. SDV RNA1 polyprotein shows sequence and organizational similarities to members of the Comoviridae family but has sufficient unique features to warrant classification in a separate genus . The RdRp domain of SDV clusters with a group containing other agriculturally important viruses including Apple latent spherical virus, Naval orange infectious mottling virus, and Rice tungro spherical virus . This grouping suggests a common ancestral origin for these viruses despite their diverse host ranges. The unique arrangement of functional domains within the RNA1 polyprotein and the presence of two coat proteins instead of one (encoded by RNA2) differentiate SDV from typical nepoviruses . Interestingly, the extensive amino acid sequence similarity between the N-terminal regions of RNA1 and RNA2 polyproteins is a feature previously observed only in tomato ringspot nepovirus, suggesting potential recombination events or duplication during evolution . Comparative genomics approaches examining conserved protein domains, non-coding regulatory elements, and RNA secondary structures across related viruses are helping reconstruct the evolutionary history of SDV and its relationship to other members of the picorna-like superfamily.

What emerging technologies hold promise for advancing research on SDV RNA1 polyprotein?

Several cutting-edge technologies are poised to significantly advance our understanding of SDV RNA1 polyprotein structure, function, and interactions. Cryo-electron microscopy (cryo-EM) has revolutionized structural biology of large protein complexes and could enable visualization of the polyprotein in different states of processing without the need for crystallization. AlphaFold2 and other AI-based protein structure prediction tools can provide increasingly accurate models of viral protein structures, particularly useful for domains resistant to experimental structural determination. Single-molecule techniques, including FRET (Förster Resonance Energy Transfer) and optical tweezers, allow real-time observation of polyprotein folding, processing, and RdRp activity at unprecedented resolution. CRISPR-Cas13 systems for RNA targeting offer new approaches for studying viral RNA-protein interactions in vivo. Nanopore direct RNA sequencing enables analysis of viral RNA modifications and structures, potentially revealing regulatory elements that interact with the polyprotein. Proteomics approaches using crosslinking mass spectrometry (XL-MS) can map intramolecular interactions within the polyprotein and intermolecular interactions with host factors. Plant virus-based expression systems derived from SDV could be developed as biotechnology tools for protein expression in plants. Advances in microfluidics and lab-on-a-chip technologies facilitate high-throughput screening for inhibitors of polyprotein processing or RdRp activity. Integration of these emerging technologies with established approaches will significantly accelerate research on SDV RNA1 polyprotein and related viral systems.

How does the SDV taxonomy inform research approaches to studying related viruses?

The unique taxonomic position of SDV has significant implications for research strategies targeting related viruses. Based on genome organization and phylogenetic analysis, SDV displays features distinct from the Comovirus, Fabavirus, and Nepovirus genera, suggesting the need for a separate genus within the Comoviridae family . This taxonomic understanding guides researchers in selecting appropriate comparative models and experimental systems. For example, when studying a newly identified citrus virus, comparing its genome organization to SDV and related viruses like Hyuganatsu virus can help predict functional domains within polyproteins and potential host interactions . The classification of Strawberry mottle virus as a member of an SDV-like lineage of picorna-like viruses demonstrates how SDV serves as a reference point for taxonomic placement of other viruses . When designing diagnostic tests, knowledge of conserved and variable regions across the SDV-related virus group enables development of broad-spectrum assays or type-specific detection methods. For evolutionary studies, the unique features of SDV, such as the N-terminal similarity between RNA1 and RNA2 polyproteins, provide important markers for tracing recombination events and evolutionary history across the picorna-like virus superfamily. These taxonomic insights ultimately inform sampling strategies, comparative genomics approaches, and experimental design for the broader field of plant virology.

VirusGenome StructureRNA1 LengthRNA2 LengthCoat Protein FeaturesTaxonomic ClassificationReference
Satsuma dwarf virus (SDV)Bipartite RNA6795 nt5345 ntTwo coat proteinsProposed new genus in Comoviridae
Hyuganatsu virus (HV)Bipartite RNAPartial sequencing (3'-terminal 1306 nt)Partial sequencing (3'-terminal 2160 nt)Similar to SDVNew SDV-related virus species
Strawberry mottle virus (SMoV)Bipartite RNA7036 nt5619 ntSimilar to SDV large CPSDV-like lineage of picorna-like viruses

What are the implications of SDV RNA1 polyprotein research for understanding other plant viral diseases?

Research on SDV RNA1 polyprotein has broader implications for understanding and managing other plant viral diseases. The insights gained from studying SDV polyprotein processing and functional domains can be applied to related viruses affecting important crops beyond citrus, such as strawberries (Strawberry mottle virus) and various other fruit crops affected by members of the Comoviridae family . The identification of conserved RdRp motifs across SDV and related viruses provides potential targets for broad-spectrum antiviral compounds that could be effective against multiple plant pathogens. Understanding the structural basis of host specificity and adaptation can inform breeding programs for viral resistance in crops, even those affected by viruses only distantly related to SDV. The mechanisms by which SDV RNA1 polyprotein interacts with host factors to establish replication complexes likely shares commonalities with other positive-strand RNA viruses, potentially revealing universal aspects of plant virus replication. Diagnostic approaches developed for SDV detection and characterization can serve as methodological templates for other emerging plant viral diseases. Evolutionary studies of SDV and related viruses help predict how plant viruses might adapt to new hosts or environmental conditions, informing surveillance and management strategies. By establishing SDV as a model system, researchers can more efficiently investigate fundamental aspects of plant virus-host interactions that have applications across multiple pathosystems.

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 2024 Thebiotek. All Rights Reserved.