H1N1 Puerto Rico Recombinant

H1N1 Influenza A- Virus Puerto Rico 08/1934 Recombinant
Shipped with Ice Packs
In Stock

Description

Viral Replication and NS1 Mutations

The nonstructural protein 1 (NS1) of H1N1 Puerto Rico Recombinant influences viral replication and host immune evasion. A key study identified the G45R mutation in NS1, which enhances replication without altering dsRNA-binding or type I interferon (IFN) suppression .

Mechanistic Insights

  • NS1 Function: NS1 antagonizes host antiviral responses via dsRNA binding and RIG-I inhibition .

  • G45R Mutation: Increases replication efficiency in A549 cells and murine lung epithelial cells (Let1), independent of IFN pathways .

Vaccine Applications

Recombinant HA proteins from this strain serve as vaccine antigens. A DSF-guided refolding protocol optimized HA production for immunogenicity:

Vaccine Efficacy in Mice

Vaccine FormulationDose (μg)Protection Against Lethal ChallengeAntibody Response
HA 18–528 (Refolded)3100% survival High IgG1/IgG2a
Denatured HA-PR83No protection Minimal response

Key Observations

  • Refolded HA 18–528 induced neutralizing antibodies and balanced Th1/Th2 responses .

  • Denatured HA lacked structural integrity, failing to elicit protective immunity .

Host-Specific Fitness Landscapes

Using H1N1 Puerto Rico Recombinant as a backbone, researchers analyzed NS1 variants from diverse influenza strains to map viral fitness in murine lungs, MDCK cells, and embryonated eggs .

Fitness Drivers

  • Host Environment: Host factors (e.g., interferon signaling) dominate replication efficiency over viral strain background .

  • NS1 Diversity: Allele B NS1 variants showed superior fitness in murine lungs, correlating with conserved amino acid residues .

High-Yield Vaccine Backbones

Recombinant H1N1 Puerto Rico viruses with optimized internal gene segments (e.g., NS1, PB2) improve titers in cell culture and embryonated eggs, enabling scalable vaccine production .

Performance Metrics

Vaccine StrainPropagation SystemTiter Improvement
PR8 BackboneMDCK cells2–5× higher
PR8 BackboneEmbryonated eggs3–10× higher

Unresolved NS1 Functions

While the G45R mutation enhances replication, its mechanism remains unclear. Further studies are needed to identify NS1’s non-IFN-related roles, such as interactions with host proteins or viral RNA .

Universal Vaccine Potential

The HA protein from H1N1 Puerto Rico Recombinant is being explored in novel vaccine platforms (e.g., novirhabdoviruses) to induce cross-protective immunity against diverse influenza subtypes .

Product Specs

Introduction
H1N1, a subtype of the Influenza A virus, has mutated into various strains, including the Spanish Flu strain, mild human flu strains, endemic pig strains, and strains found in birds. The Influenza A virus is roughly 100nm in diameter and spherical. It is enclosed in a lipid bilayer derived from its host's plasma membrane. Embedded within this bilayer are approximately 500 molecules of hemagglutinin ("H") and 100 molecules of neuraminidase ("N"). Internally, the virus contains 3000 molecules of matrix protein, 8 RNA segments, and numerous nucleoproteins. Each RNA segment is linked with multiple copies of a nucleoprotein, several RNA polymerase subunits, and unidentified "non-structural" protein molecules.
Description
Recombinant Full-Length H1N1 Puerto Rico 08/1934, with a molecular weight of approximately 62 kDa, is glycosylated with N-linked sugars. It is produced using baculovirus vectors in insect cells.
Physical Appearance
A sterile, colorless solution.
Formulation
The Recombinant H1N1 A/Puerto Rico 08/1934 solution consists of 10mM Sodium phosphate (pH 7.2), 150mM NaCl, and 0.005% Tween-20.
Stability
Recombinant H1N1 A/Puerto Rico 08/1934 should be stored at 4°C. Freezing is not recommended.
Purity
Purity exceeds 90.0% as determined by SDS-PAGE analysis.
Source
Baculovirus Insect Cells.

Q&A

What is the molecular structure of H1N1 A/Puerto Rico/8/1934 hemagglutinin protein?

The hemagglutinin (HA) protein from A/Puerto Rico/8/1934 (H1N1) consists of 528 amino acids (Met1-Gln528) and is a glycoprotein with a molecular weight of approximately 62 kDa when fully glycosylated. The protein sequence is available under accession number ABD77675.1. In recombinant form, it can be produced with various tags such as C-terminal polyhistidine for purification purposes. The HA protein is one of the two major surface proteins of influenza viruses (alongside neuraminidase) and appears as a spike-like structure on the viral surface with approximately 500 molecules per virion. It plays critical roles in viral attachment to host cells and membrane fusion during the infection process .

How does the H1N1 A/Puerto Rico/8/1934 strain differ from other historical and modern H1N1 strains?

The A/Puerto Rico/8/1934 strain is one of the earliest isolated influenza strains and serves as a reference laboratory strain. It differs from modern H1N1 strains in several ways, including antigenicity and genetic composition. Notably, antibodies raised against this historical strain show limited cross-reactivity with contemporary H1N1 viruses. Specificity testing shows that antibodies against A/Puerto Rico/8/1934 HA do not cross-react with modern strains such as A/Victoria/4897/2022, A/Wisconsin/588/2019, and A/Wisconsin/67/2022 in ELISA assays . This antigenic drift illustrates the evolutionary distance between the 1934 strain and current circulating viruses. The strain has been extensively adapted to laboratory use and may contain specific mutations that enhance its replication in experimental settings, such as the G45R mutation in the nonstructural protein 1 (NS1) .

What are the optimal storage conditions for maintaining viability of recombinant H1N1 A/Puerto Rico/8/1934 proteins?

Recombinant H1N1 A/Puerto Rico/8/1934 proteins should be stored at 4°C and should NOT be frozen to maintain structural integrity and biological activity. The formulation typically contains buffer components such as 10mM sodium phosphate at pH 7.2, 150mM NaCl, and 0.005% Tween-20 that help stabilize the protein . For long-term storage considerations, protein solutions should be kept in aliquots to avoid repeated freeze-thaw cycles which can lead to protein denaturation. When handling these proteins, researchers should minimize exposure to room temperature and follow specific manufacturer guidance for each product preparation. Documentation of storage conditions and expiration dates is essential for experimental reproducibility and validity of research findings.

What are the recommended protocols for using H1N1 A/Puerto Rico/8/1934 recombinant proteins in vaccine development research?

When using H1N1 A/Puerto Rico/8/1934 recombinant proteins for vaccine research, researchers should implement a multifaceted approach:

  • Immunogenicity Assessment: Begin with dose-response studies using purified recombinant proteins (particularly hemagglutinin) with various adjuvants to determine optimal formulations. The recombinant protein should be characterized for purity (>90% as determined by SDS-PAGE) and properly folded structure before immunization studies .

  • Animal Model Selection: Select appropriate animal models based on research objectives. For basic immunogenicity, mice are suitable, while ferrets provide better models for transmission and pathogenesis studies.

  • Adjuvant Screening: Test multiple adjuvant formulations to enhance immune responses, as recombinant proteins alone often show limited immunogenicity.

  • Immunization Schedule: Implement prime-boost strategies with appropriate intervals between doses (typically 2-4 weeks) to maximize antibody titers and affinity maturation.

  • Immune Response Analysis: Evaluate both humoral (neutralizing antibodies by hemagglutination inhibition and microneutralization assays) and cellular immune responses (T-cell responses via ELISpot or flow cytometry).

  • Challenge Studies: For protective efficacy assessment, conduct viral challenge studies using either wild-type A/Puerto Rico/8/1934 or reassortant viruses carrying the homologous HA protein.

The experimental design should include appropriate controls such as adjuvant-only groups and comparative analysis with current standard vaccines when applicable.

How can researchers effectively use antibodies against H1N1 A/Puerto Rico/8/1934 in immunological assays?

To effectively use antibodies against H1N1 A/Puerto Rico/8/1934 in immunological assays, researchers should follow these methodological considerations:

  • ELISA Applications: When developing sandwich ELISA assays, pair compatible antibodies such as monoclonal antibody clone #05 as a capture antibody with appropriate detection antibodies. Optimize antibody concentrations through titration experiments to determine the optimal working dilution range .

  • Specificity Verification: Always confirm antibody specificity against the target strain (A/Puerto Rico/8/1934) and test for potential cross-reactivity with other influenza strains. Commercial antibodies have been tested against various H1N1 and H3N2 strains and have shown high specificity for the A/Puerto Rico/8/1934 strain .

  • Immunofluorescence Optimization: For immunofluorescence applications, use appropriate fixation methods (4% paraformaldehyde is commonly used) and permeabilization agents (0.1-0.5% Triton X-100) to ensure access to viral antigens. Include appropriate blocking steps (5% BSA or serum) to minimize non-specific binding.

  • Western Blotting Protocol: For western blot analysis of viral proteins, use optimized protein extraction methods and appropriate loading controls. Protein A purified monoclonal antibodies typically perform well in western blot applications when used at experimentally determined concentrations .

  • Controls: Always include positive controls (purified recombinant proteins), negative controls (uninfected cells/irrelevant antigens), and isotype controls to validate assay specificity and sensitivity.

  • Signal Development and Quantification: Select appropriate detection systems (colorimetric, chemiluminescent, or fluorescent) based on sensitivity requirements and available instrumentation.

What methods are recommended for generating and validating influenza reassortant viruses using A/Puerto Rico/8/1934 backbone?

Generation and validation of influenza reassortant viruses using the A/Puerto Rico/8/1934 backbone require careful experimental design and rigorous validation:

  • Reverse Genetics System Setup:

    • Utilize the 8-plasmid reverse genetics system where each genomic segment is cloned into appropriate expression vectors

    • Maintain a complete set of plasmids for all eight segments of A/Puerto Rico/8/1934

    • For reassortment, replace specific segments (commonly HA and NA) with corresponding segments from other influenza strains

  • Transfection and Rescue:

    • Co-transfect the plasmid mixture into a compatible cell line (typically HEK293T cells)

    • Overlay with MDCK cells after 24 hours for efficient virus amplification

    • Harvest supernatant containing rescued viruses 48-72 hours post-transfection

  • Virus Isolation and Amplification:

    • Perform plaque purification to isolate clonal viral populations

    • Expand virus in embryonated chicken eggs or MDCK cells

    • Create working stocks and determine viral titers using plaque assays or TCID50 methods

  • Genetic Validation:

    • Confirm genomic composition by segment-specific RT-PCR

    • Sequence the complete viral genome to verify correct reassortment and absence of unwanted mutations

    • For studies involving specific mutations like G45R in NS1, perform targeted sequencing to confirm the mutation's presence

  • Phenotypic Characterization:

    • Assess growth kinetics in relevant cell lines (MDCK, A549, Let1 cells)

    • Compare replication efficiency with parental strains

    • Evaluate virus morphology by electron microscopy if changes in structural proteins were introduced

  • Functional Assays:

    • For reassortants with modified NS1 (such as G45R mutation), perform dsRNA-binding assays and IFN antagonism assays to characterize functional changes

    • For HA/NA reassortants, conduct hemagglutination and neuraminidase activity assays

Proper biosafety practices should be followed throughout the process, with appropriate containment levels based on the reassortant's anticipated phenotype.

How does the G45R mutation in NS1 of A/Puerto Rico/8/1934 influence viral replication mechanisms?

The G45R mutation in the nonstructural protein 1 (NS1) of A/Puerto Rico/8/1934 enhances viral replication through several mechanisms that appear to be independent of the protein's dsRNA-binding activity and direct type I interferon antagonism:

  • Effect on Replication Efficiency: Viruses carrying the G45R mutation in NS1 demonstrate accelerated replication kinetics compared to wild-type viruses in various cell lines, including A549 and murine lung epithelial type I (Let1) cells. This enhanced replication occurs without corresponding increases in dsRNA binding capability .

  • Structural Implications: The mutation occurs in the RNA-binding domain (RBD) of NS1, but contrary to computational predictions, experimental dsRNA-NS1 pull-down assays revealed that G45R/NS1 does not show increased dsRNA-binding compared to wild-type NS1. This suggests that the mutation enhances viral replication through alternative mechanisms beyond the canonical dsRNA sequestration pathway .

  • Interferon Antagonism Independence: While NS1 is known to inhibit IFNα/β induction and response, the G45R mutation appears to enhance viral replication through pathways that are not directly linked to suppression of RIG-I mediated IFNβ-promoter activity. This represents a novel function of NS1 that contributes to viral fitness .

  • Evolutionary Significance: The G45R mutation has been observed in pandemic H1N1 2009 viruses, suggesting potential evolutionary advantages. Studies propose that this mutation might be associated with increased cytokine production during infections, potentially contributing to disease severity through immunopathological mechanisms rather than direct viral cytopathic effects .

  • Strain-Specific Functions: The enhanced replication mediated by G45R appears to be strain-specific, as similar effects have been observed when comparing A/swine/IA/15/1930 (IA30) NS with PR8 NS. This highlights the importance of genetic background in determining the functional impact of specific mutations .

These findings suggest that researchers investigating influenza pathogenicity should consider the complex, multifunctional nature of NS1 beyond its established role in interferon antagonism.

What are the key considerations when comparing immunogenicity of different recombinant hemagglutinin protein preparations from A/Puerto Rico/8/1934?

When comparing immunogenicity of different recombinant hemagglutinin (HA) preparations from A/Puerto Rico/8/1934, researchers should consider several critical factors that can significantly impact experimental outcomes:

  • Expression System Influence:
    Different expression platforms produce proteins with varying post-translational modifications:

    Expression SystemGlycosylation PatternProtein FoldingTypical Applications
    HEK293 CellsComplex, human-like glycansNative-likeStructural studies, neutralizing antibody generation
    Insect CellsSimpler glycans, mannose-richGenerally correctHigh-yield production, functional studies
    Bacterial SystemsNo glycosylationMay require refoldingEpitope mapping, linear epitope antibodies
  • Protein Domain Considerations:

    • Full-length HA vs. HA ectodomain (ECD): Full-length includes transmembrane domain while ECD is more soluble

    • HA0 vs. HA1/HA2: Uncleaved precursor vs. proteolytically processed form affects receptor binding and fusion activity

    • Presence of trimerization domains may influence proper oligomeric assembly and stability

  • Tag Effects on Structure and Function:

    • C-terminal polyhistidine tags (as in the commercially available products) generally have minimal impact on HA functionality

    • Location of tags (N-terminal vs. C-terminal) can affect folding and epitope accessibility

    • Size and type of tags should be considered when interpreting immunological outcomes

  • Protein Quality Assessment:

    • Purity levels (>90% by SDS-PAGE is standard for research-grade preparations)

    • Aggregation state (monitored by size exclusion chromatography)

    • Biological activity (hemagglutination assay, receptor binding)

    • Antigenicity (reactivity with conformation-dependent antibodies)

  • Immunization Protocol Variables:

    • Adjuvant selection drastically affects quality and quantity of immune response

    • Dosing schedule (prime-boost intervals)

    • Administration route (intramuscular vs. intranasal vs. subcutaneous)

    • Protein dose standardization (by mass or activity units)

  • Immune Response Analysis Standardization:

    • Use consistent antibody panels for comparative studies

    • Standardize ELISA protocols across experiments

    • Include reference standards for neutralization assays

    • Evaluate both binding and functional antibodies

What approaches are recommended for resolving discrepancies in experimental data when working with A/Puerto Rico/8/1934 recombinant proteins?

When researchers encounter discrepancies in experimental data while working with A/Puerto Rico/8/1934 recombinant proteins, a systematic troubleshooting approach is essential. The following methodological framework can help identify and resolve inconsistencies:

  • Protein Quality Assessment:

    • Perform fresh analysis of protein integrity using SDS-PAGE and western blotting

    • Verify glycosylation status using glycan-specific stains or mass spectrometry

    • Assess aggregation state through dynamic light scattering or size exclusion chromatography

    • Confirm biological activity through functional assays specific to the protein (e.g., hemagglutination for HA proteins)

  • Reagent and Reference Standard Verification:

    • Use validated antibodies with confirmed specificity against A/Puerto Rico/8/1934 proteins

    • Include well-characterized reference standards in each experiment

    • Create internal control pools that can be used across multiple experiments

    • Verify the authenticity of cell lines using STR profiling

  • Protocol Standardization:

    • Document detailed protocols including buffer compositions, incubation times, and temperatures

    • Standardize protein quantification methods (BCA vs. Bradford vs. UV absorbance)

    • Control for lot-to-lot variations in critical reagents

    • Implement automated systems where possible to reduce operator variability

  • Statistical Analysis and Data Integration:

    • Apply appropriate statistical tests based on data distribution and experimental design

    • Use multivariate analysis to identify patterns in complex datasets

    • Consider meta-analysis approaches when comparing across multiple experiments

    • Implement blind analysis protocols for subjective measurements

  • Experimental Design Refinement:

    • Increase biological and technical replicates

    • Include appropriate positive and negative controls

    • Design factorial experiments to identify interaction effects

    • Consider time-course studies to identify temporal variables

  • Cross-Validation Strategies:

    • Verify key findings using orthogonal methods

    • Collaborate with independent laboratories for validation

    • Compare results with published literature while accounting for methodological differences

    • Consider interlaboratory studies for highly variable assays

  • Documentation and Data Sharing:

    • Maintain comprehensive records of all experimental conditions

    • Share raw data alongside processed results

    • Document all deviations from standard protocols

    • Implement electronic laboratory notebooks for improved traceability

By systematically addressing these aspects, researchers can identify sources of experimental variability and develop robust, reproducible protocols for working with A/Puerto Rico/8/1934 recombinant proteins.

How are researchers using A/Puerto Rico/8/1934 recombinant proteins in development of universal influenza vaccines?

Researchers are leveraging A/Puerto Rico/8/1934 recombinant proteins in several innovative approaches toward universal influenza vaccine development:

  • Conserved Epitope Targeting:
    The A/Puerto Rico/8/1934 strain serves as an important historical reference point for identifying conserved epitopes that have remained stable over decades of viral evolution. Researchers engineer recombinant HA proteins from this strain to selectively display conserved regions while masking variable domains. This approach helps focus immune responses on broadly protective epitopes rather than strain-specific regions.

  • Structure-Based Immunogen Design:
    Using the well-characterized structure of A/Puerto Rico/8/1934 HA as a scaffold, researchers create chimeric proteins that incorporate conserved elements from multiple influenza subtypes. Advanced protein engineering techniques allow precise manipulation of antigenic sites to elicit broadly neutralizing antibodies against the conserved stem region of HA.

  • Reassortant Platform Development:
    The genetic backbone of A/Puerto Rico/8/1934 provides a stable foundation for creating reassortant viruses that express HA proteins from different strains while maintaining consistent internal proteins. This approach enables systematic comparison of immune responses to different HA variants in a controlled genetic background . Mutations like G45R in NS1 can be incorporated to enhance vaccine virus replication efficiency without increasing pathogenicity.

  • Historical Immunological Imprinting Studies:
    Comparing immune responses to historical strains like A/Puerto Rico/8/1934 with contemporary isolates helps researchers understand how prior exposures shape subsequent immunity—a concept known as original antigenic sin or imprinting. This knowledge guides vaccine designs that can overcome potential immune focusing effects.

  • Novel Adjuvant Development and Testing:
    The well-characterized nature of A/Puerto Rico/8/1934 recombinant proteins makes them excellent candidates for evaluating novel adjuvant formulations. Researchers can directly compare how different adjuvants influence the breadth, magnitude, and durability of immune responses to the same antigenic target.

  • Multivalent Vaccine Approaches:
    By including A/Puerto Rico/8/1934 components alongside proteins from other influenza strains, researchers develop multivalent formulations that provide broader coverage against diverse influenza viruses while maintaining production efficiency.

These approaches collectively contribute to the goal of developing vaccines that protect against a wider range of influenza viruses, including potential pandemic strains, without requiring annual reformulation.

What new methodologies are emerging for studying protein-protein interactions involving A/Puerto Rico/8/1934 viral proteins?

Emerging methodologies for studying protein-protein interactions (PPIs) involving A/Puerto Rico/8/1934 viral proteins are advancing our understanding of influenza biology through several cutting-edge approaches:

  • Proximity Labeling Techniques:

    • BioID and TurboID methods are being applied to viral proteins like NS1 with mutations such as G45R to identify proximal interaction partners in living cells

    • APEX2-based proximity labeling allows temporal resolution of dynamic interactions during different stages of viral replication

    • These approaches help identify transient interactions that may be missed by traditional co-immunoprecipitation methods

  • Advanced Structural Biology Approaches:

    • Cryo-electron microscopy (cryo-EM) with improved resolution now enables visualization of complex assemblies involving hemagglutinin and other viral proteins

    • Hydrogen-deuterium exchange mass spectrometry (HDX-MS) provides insights into conformational changes upon protein binding

    • Integrative structural biology combining multiple techniques (X-ray crystallography, NMR, SAXS, cryo-EM) creates comprehensive models of viral protein complexes

  • Protein-Protein Interaction Screening:

    • CRISPR-based genetic screens identify host factors that interact with specific viral proteins

    • Protein microarrays containing human proteome allow systematic identification of virus-host interactions

    • Split protein complementation assays (such as split luciferase) enable high-throughput screening of potential interaction partners

  • Live-Cell Imaging Techniques:

    • Förster resonance energy transfer (FRET) sensors designed for specific viral proteins allow real-time monitoring of interactions

    • Lattice light-sheet microscopy provides improved spatiotemporal resolution for tracking viral protein complexes during infection

    • Super-resolution microscopy techniques overcome the diffraction limit to visualize nanoscale organization of viral assemblies

  • Computational Prediction and Validation:

    • Machine learning algorithms trained on known influenza protein interactions predict novel interaction partners

    • Molecular dynamics simulations reveal binding mechanisms and conformational changes

    • These computational predictions guide targeted experimental validation studies

  • Cross-linking Mass Spectrometry (XL-MS):

    • Chemical cross-linking followed by mass spectrometry identifies interaction interfaces between viral proteins and host factors

    • MS-cleavable cross-linkers improve identification of cross-linked peptides

    • This approach is particularly valuable for studying the interfaces of the NS1 protein with its various binding partners

These methodologies collectively provide unprecedented insights into the molecular mechanisms of influenza virus pathogenesis and host interactions, offering new avenues for therapeutic intervention.

What challenges remain in translating findings from A/Puerto Rico/8/1934 research to contemporary pandemic preparedness?

Despite the extensive use of A/Puerto Rico/8/1934 in influenza research, significant challenges remain in translating findings to contemporary pandemic preparedness:

  • Evolutionary Distance and Antigenic Drift:
    The substantial genetic and antigenic differences between A/Puerto Rico/8/1934 and contemporary circulating strains limit direct extrapolation of findings. Antibodies against A/Puerto Rico/8/1934 HA show minimal cross-reactivity with modern H1N1 strains, as demonstrated in specificity testing of commercial antibodies . This evolutionary gap requires careful validation of findings in contemporary strain contexts before application to pandemic planning.

  • Laboratory Adaptation Effects:
    A/Puerto Rico/8/1934 has undergone extensive laboratory adaptation, potentially introducing mutations that enhance replication in experimental systems but may not reflect natural viral behavior. The G45R mutation in NS1, which enhances viral replication, exemplifies how laboratory-adapted strains may contain features that alter their biology relative to clinical isolates . Research findings must account for these adaptation effects when developing pandemic countermeasures.

  • Host Range Considerations:
    Most A/Puerto Rico/8/1934 research occurs in laboratory cell lines or mouse models, which may not accurately represent human infection dynamics. Contemporary pandemic planning requires translation to models that better reflect human respiratory physiology and immune responses, such as human airway epithelial cultures, organoids, or humanized mouse models.

  • Technological Platform Differences:
    Modern vaccine platforms (mRNA, viral vectors) differ substantially from the experimental systems often used with A/Puerto Rico/8/1934. Translating findings from traditional protein-based studies to these newer technologies requires additional validation and potentially modified approaches.

  • Complex Immune History in Human Populations:
    Unlike laboratory models using naïve animals, human populations have complex influenza exposure histories that shape immune responses through mechanisms like original antigenic sin and imprinting. A/Puerto Rico/8/1934 research often fails to account for these pre-existing immunity effects that significantly impact pandemic vaccine effectiveness.

  • Changing Ecological Context:
    Modern pandemic threats occur in a different ecological context than existed in 1934, with altered patterns of human-animal contact, population density, and global travel. Research using historical strains must be contextualized within current epidemiological landscapes.

  • Data Integration Challenges:
    Integrating findings from A/Puerto Rico/8/1934 research with genomic surveillance, computational modeling, and clinical studies of contemporary strains requires sophisticated bioinformatic approaches and standardized reporting frameworks that are still evolving.

Addressing these challenges requires interdisciplinary collaboration, improved animal models, comparative studies between historical and contemporary strains, and systems biology approaches that can bridge the gap between laboratory findings and real-world pandemic preparedness.

What are the most reliable sources for obtaining standardized A/Puerto Rico/8/1934 recombinant proteins and related research materials?

For researchers requiring standardized A/Puerto Rico/8/1934 recombinant proteins and related materials, several reliable sources provide well-characterized reagents:

  • Commercial Protein Suppliers:

    • Sino Biological offers recombinant hemagglutinin (HA) proteins from A/Puerto Rico/8/1934 with C-terminal His tags produced in HEK293 cells, suitable for functional and structural studies

    • ProSpec Bio provides recombinant full-length H1N1 A/Puerto Rico/8/1934 produced using baculovirus vectors in insect cells, with documented stability parameters and formulation details

    • Additional suppliers include major life sciences companies that produce standardized preparations with extensive quality control documentation

  • Antibody Resources:

    • Bio-Techne/Novus Biologicals offers monoclonal antibodies specific to A/Puerto Rico/8/1934 hemagglutinin with validated applications in ELISA and other immunological techniques

    • Antibodies against other viral proteins including PA (polymerase acidic protein) are available with specified applications in Western blotting and immunofluorescence

    • These resources typically provide detailed validation data including specificity testing against multiple influenza strains

  • Academic Repositories:

    • BEI Resources (managed by ATCC) maintains a collection of influenza reagents including viral stocks, proteins, and plasmids

    • The International Reagent Resource (IRR) provides authenticated influenza reagents to qualified researchers

    • These repositories often provide materials at lower cost than commercial sources but may have longer procurement timelines

  • Collaborative Research Networks:

    • Centers of Excellence for Influenza Research and Surveillance (CEIRS) network laboratories can provide specialized reagents and technical expertise

    • International collaborations like GISAID facilitate sharing of sequence data and sometimes physical materials

  • Quality Considerations When Selecting Sources:

    • Look for suppliers providing comprehensive quality control data (purity measurements, functional validation)

    • Consider the expression system used and its implications for downstream applications

    • Verify that suppliers provide lot-specific analysis certificates

    • Check for detailed handling and storage recommendations to maintain reagent stability

When obtaining these materials, researchers should carefully document specific catalog numbers, lot numbers, and any supplier-specified handling protocols to ensure experimental reproducibility. For recombinant proteins, storage at 4°C rather than freezing is recommended for A/Puerto Rico/8/1934 hemagglutinin preparations .

What emerging technologies might transform our understanding of A/Puerto Rico/8/1934 and its applications in the next decade?

Several cutting-edge technologies are poised to revolutionize our understanding of A/Puerto Rico/8/1934 and expand its research applications in the coming decade:

  • Single-Cell Omics Integration:
    The integration of single-cell transcriptomics, proteomics, and metabolomics will provide unprecedented insights into host-pathogen interactions at the individual cell level. This approach will reveal how A/Puerto Rico/8/1934 infection creates heterogeneous responses across cell populations and identify previously unrecognized cellular subsets that may serve as key viral reservoirs or immune coordinators.

  • Cryo-Electron Tomography (Cryo-ET):
    Advances in cryo-ET will allow visualization of viral structures within their native cellular environment at near-atomic resolution. This technology will bridge the gap between in vitro structural studies of isolated recombinant proteins and their actual conformation and interactions within infected cells, potentially revealing new therapeutic targets.

  • AI-Driven Protein Design:
    Machine learning approaches like AlphaFold have transformed protein structure prediction. Future applications will extend to designing optimized versions of A/Puerto Rico/8/1934 proteins with enhanced stability, immunogenicity, or specific functional properties. This could lead to next-generation immunogens based on the well-characterized PR8, but with significantly improved properties for vaccine applications.

  • Organoid and Microphysiological Systems:
    Human respiratory tract organoids and lung-on-chip technologies will provide more physiologically relevant models for studying A/Puerto Rico/8/1934 infection. These systems recapitulate the complex cellular architecture and functions of human respiratory tissues, allowing more accurate assessment of viral tropism, pathogenesis, and immune responses in a human-relevant context.

  • CRISPR-Based Viral Engineering:
    Beyond traditional reverse genetics, CRISPR technologies will enable more precise and efficient modification of viral genomes. This will facilitate high-throughput functional genomics studies to comprehensively map how each amino acid in viral proteins contributes to replication, immune evasion, and pathogenesis, using A/Puerto Rico/8/1934 as a model system.

  • In Situ Structural Biology:
    Emerging techniques like cryo-electron microscopy of vitreous sections (CEMOVIS) and correlative light and electron microscopy (CLEM) will allow researchers to visualize viral components within infected cells at unprecedented resolution. Applied to A/Puerto Rico/8/1934, these approaches will reveal how viral proteins like NS1 with the G45R mutation interact with host components in their native environment.

  • Systems Vaccinology and Immunomics: Multi-omics approaches applied to vaccine studies will identify molecular signatures associated with protective immunity. When applied to A/Puerto Rico/8/1934-based vaccines or challenges, these technologies will help decipher the complex networks of immune responses and guide rational design of improved vaccines with broader protection.

Product Science Overview

Introduction

The H1N1 Influenza A virus, specifically the Puerto Rico 08/1934 strain, is a significant historical and scientific entity in the study of influenza viruses. This strain has been extensively used in research due to its well-documented genetic and antigenic properties. The recombinant form of this virus, particularly the hemagglutinin (HA) protein, has been instrumental in understanding viral mechanisms and developing vaccines.

Historical Context

The Puerto Rico 08/1934 strain of H1N1 Influenza A virus was first isolated in 1934. This strain is one of the earliest and most well-characterized influenza viruses. It has served as a reference strain in numerous studies and has been pivotal in the development of influenza vaccines. The 1934 strain is particularly notable for its role in the 1918 influenza pandemic, which caused widespread morbidity and mortality globally.

Genetic and Antigenic Properties

The H1N1 Influenza A virus has a segmented RNA genome, which allows for genetic reassortment and the emergence of new viral strains. The Puerto Rico 08/1934 strain is characterized by its hemagglutinin (HA) and neuraminidase (NA) surface proteins. The HA protein facilitates viral entry into host cells by binding to sialic acid receptors, while the NA protein aids in the release of newly formed viral particles from infected cells.

The recombinant form of the Puerto Rico 08/1934 strain typically involves the expression of the HA protein in a host system, such as HEK293 cells. This recombinant HA protein is often tagged with a His-tag to facilitate purification and detection. The recombinant HA protein retains the antigenic properties of the native virus, making it a valuable tool for immunological studies and vaccine development .

Applications in Research

The recombinant HA protein from the Puerto Rico 08/1934 strain has been widely used in various research applications, including:

  • Vaccine Development: The HA protein is a primary target for influenza vaccines. Recombinant HA proteins are used to generate immune responses in vaccine candidates.
  • Immunological Studies: Researchers use recombinant HA proteins to study the immune response to influenza infection and to identify potential therapeutic targets.
  • Diagnostic Assays: Recombinant HA proteins are used in diagnostic assays to detect antibodies against influenza viruses in clinical samples.

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.