Dengue NS1 ST1, Insect

Dengue Virus NS1 Subtype 1 Recombinant, Insect Cells
Shipped with Ice Packs
In Stock

Description

Definition and Biological Context

Dengue NS1 is a glycoprotein essential for viral replication and immune modulation. The "ST1" designation specifies serotype 1, one of four dengue virus serotypes. The "Insect" label indicates production in insect cell lines (e.g., Spodoptera frugiperda), which ensures proper post-translational modifications while maintaining cost-effectiveness .

Expression System

  • Host Cells: Produced in insect cell lines (e.g., Sf9 or Sf21) using baculovirus vectors .

  • Amino Acid Range: Comprises residues 777–1131 of the dengue polyprotein .

  • Post-Translational Modifications:

    • Contains high-mannose glycans, unlike mammalian cell-derived NS1, which includes complex glycans .

    • Forms hexamers, the biologically active conformation involved in immune evasion and vascular leakage .

Physical and Functional Characteristics

PropertyDetails
Molecular Weight~50 kDa (SDS-PAGE)
Purity>95% (chromatography)
Formulation1 mg/mL in PBS, pH 7.4, with EDTA and protease inhibitors
StabilityStable at 4°C for 1 week; long-term storage at <-18°C

Diagnostic Development

  • Used as an antigen in ELISA and lateral flow assays to detect dengue NS1 in patient sera .

  • Exhibits high specificity for dengue serotype 1, with minimal cross-reactivity to Zika or West Nile NS1 .

Vaccine and Therapeutic Studies

  • Serves as an immunogen to generate neutralizing antibodies .

  • Studies highlight its role in immune evasion by binding host proteins like mannose-binding lectin (MBL), shielding the virus from complement-mediated neutralization .

Pathogenesis Research

  • Insect-derived NS1 retains functionality in inducing endothelial hyperpermeability, a hallmark of severe dengue .

  • Triggers degradation of the endothelial glycocalyx via sialidases and heparanase, contributing to vascular leakage .

Key Research Findings

  • Secretion Mechanism: NS1 from insect cells follows an unconventional secretory pathway dependent on caveolin-1 and chaperone complexes, distinct from mammalian systems .

  • Glycan Impact: High-mannose glycans enable binding to MBL, which protects dengue virus from lectin pathway neutralization .

  • Structural Stability: Hexameric conformation critical for interactions with host factors like TLR4 and complement proteins .

Comparative Advantages

  • Cost-Effectiveness: Insect cell systems offer scalable production with lower costs than mammalian systems .

  • Antigenicity: Retains native epitopes due to proper folding and glycosylation, enhancing utility in immunoassays .

Limitations and Considerations

  • Glycosylation Differences: Lack of complex glycans may alter immune recognition compared to mammalian-derived NS1 .

  • Functional Variability: Serotype-specific effects necessitate parallel testing with NS1 from other dengue serotypes .

Future Directions

  • Therapeutic Targeting: NS1’s role in vascular leak makes it a candidate for monoclonal antibody therapies .

  • Cross-Reactivity Studies: Investigating interactions with emerging flaviviruses (e.g., Zika) to improve diagnostic specificity .

Product Specs

Introduction
Dengue fever is caused by four closely related virus serotypes of the genus Flavivirus, family Flaviviridae. Each serotype is distinct enough that infection with one does not confer immunity to the others. This means that an individual can experience multiple dengue infections, and regions can experience epidemics involving multiple serotypes (hyperendemicity). Research has shown that Morpholino antisense oligos can effectively target and inhibit Dengue virus replication in cell cultures and mice models.
Description
This product consists of the recombinant Dengue Virus Nonstructural protein 1 (NS1) Subtype 1, expressed in insect cells. It encompasses amino acids 777 to 1131 of the NS1 protein and has a molecular weight of approximately 50 kDa. The protein is purified to a high degree using a proprietary chromatographic method.
Physical Appearance
A clear solution without any particles, sterilized by filtration.
Formulation
The Dengue NS1 ST1 protein is supplied in a solution containing 1mg/ml protein in a buffer consisting of 1xPBS (pH 7.4), 0.1% Thimerosal, 5mM EDTA, and protease inhibitors (1µg/ml each of Leupeptin, Aprotinin, and Pepstatin A).
Stability
For short-term storage (up to 4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. Adding a carrier protein like HSA or BSA to a final concentration of 0.1% is advisable for long-term storage. Avoid repeated freezing and thawing.
Purity
The purity of the Dengue NS1 ST1 protein is greater than 95% as determined by SDS-PAGE analysis using a 12.5% gel.
Applications
This product can be used as a control in antigen detection tests.
Source
Insect cells.

Q&A

What is the molecular structure and basic function of dengue virus NS1?

Dengue virus NS1 is a conserved glycoprotein of 46-50 kDa that exists in multiple oligomeric forms within infected cells and in secreted form. In infected cells, NS1 associates with cell membranes as a dimer, while it is secreted into the extracellular environment as a hexamer. This protein is conserved across flaviviruses but exhibits specific characteristics in dengue virus infections . The molecular structure of NS1 includes glycosylation sites that contribute to its functional properties, though the glycosylation status can vary depending on the secretory pathway and cell type involved in its production. The basic functions of NS1 include roles in viral replication and immune evasion, while secreted NS1 contributes to pathogenesis .

Methodologically, researchers investigating NS1 structure often employ techniques such as crystallography, cryo-electron microscopy, and biochemical analyses of purified protein. Comparative studies between NS1 from different flaviviruses (such as dengue and West Nile virus) have revealed important structural differences that correlate with functional specificity .

How can NS1 be detected during dengue virus infection?

NS1 antigen testing serves as a valuable diagnostic tool during the acute phase of dengue virus infection. The protein is detectable in serum primarily during the first 7 days of illness, making NS1 tests particularly useful for early diagnosis . These tests utilize synthetically labeled antibodies that specifically recognize the dengue NS1 protein in patient samples.

Methodologically, NS1 detection is typically performed using serum samples, although blood and plasma can also be used. The tests can be as sensitive as molecular detection methods during the early symptomatic period. While a positive NS1 test confirms dengue virus infection, it does not provide information about the specific dengue serotype (DENV1-4) . For researchers requiring serotype information for surveillance purposes, nucleic acid amplification tests such as RT-PCR must be employed as a complementary approach. In clinical research settings, combining NS1 and IgM antibody tests provides comprehensive diagnostic coverage during the first week of illness .

What is the significance of NS1 secretion in the mosquito vector?

NS1 is efficiently secreted from infected mosquito cells, representing a critical aspect of the virus-vector interaction. The secretion of NS1 from mosquito cells follows distinctive pathways compared to vertebrate cells, suggesting evolutionary adaptations specific to the arthropod vector . This differential secretion mechanism may contribute to successful viral replication and transmission within the mosquito.

Methodologically, researchers investigating NS1 secretion in mosquito cells employ cell culture systems using established mosquito cell lines (such as C6/36 from Aedes albopictus). Experimental approaches include the use of pharmacological inhibitors of secretory pathways, protein trafficking analysis through subcellular fractionation, and immunofluorescence microscopy to track NS1 localization . Understanding NS1 secretion in mosquito cells provides insights into vector competence and potential targets for interrupting the transmission cycle.

How does the secretory pathway for NS1 differ between mosquito and vertebrate host cells?

NS1 follows distinctly different secretory pathways in mosquito versus vertebrate cells, representing a fundamental difference in virus-host interactions across the transmission cycle. In vertebrate cells, NS1 secretion follows the classical secretory pathway through the Golgi complex . In contrast, NS1 secretion in mosquito cells occurs via an unconventional route that bypasses the Golgi complex .

Methodologically, this difference can be demonstrated experimentally through the use of Brefeldin A (BFA), which inhibits protein transport from the endoplasmic reticulum to the Golgi apparatus. Studies have shown that BFA treatment does not significantly inhibit NS1 secretion in mosquito cells, while methyl-β-cyclodextrin (MβCD, which depletes cholesterol) does reduce NS1 release . This suggests involvement of a cholesterol-dependent, Golgi-independent pathway in mosquito cells.

The mosquito cell secretory pathway for NS1 is dependent on caveolin-1 (CAV1), a key component of the caveolar system responsible for cholesterol transport. Experimental silencing of CAV1 expression significantly reduces NS1 secretion, whereas silencing SAR1 (a GTPase involved in the classical secretory pathway) does not affect NS1 release in mosquito cells . More specifically, NS1 secretion in mosquito cells occurs in association with the chaperone caveolin complex, comprised of CAV1 and the chaperones FKBP52, CyA, and Cy40, which regulate cholesterol transport within the cell .

Proximity ligation assays have demonstrated direct interaction between NS1 and CAV1 in infected mosquito cells, and computational simulations suggest favorable interactions between the caveolin-binding domain in NS1 and the scaffolding domain of CAV1 . These findings align with the lipoprotein characteristics of secreted NS1 and provide important insights into vector-specific viral adaptations.

What molecular mechanisms underlie NS1-induced vascular leakage in severe dengue?

NS1 contributes to dengue pathogenesis through its ability to induce endothelial hyperpermeability, which can lead to the plasma leakage characteristic of severe dengue disease. NS1 disrupts the endothelial glycocalyx layer (EGL), a network of membrane-bound proteoglycans and glycoproteins that lines the vascular endothelium and regulates endothelial barrier function .

Methodologically, researchers have elucidated several specific mechanisms by which NS1 damages the EGL:

  • NS1 induces expression of sialidases, leading to degradation of sialic acid components of the EGL

  • NS1 triggers expression of heparanase, resulting in shedding of heparan sulfate proteoglycans

  • NS1 activates cathepsin L, a lysosomal cysteine proteinase that enzymatically cleaves and activates heparanase

These effects have been demonstrated experimentally using human pulmonary microvascular endothelial cells treated with purified NS1 protein. Importantly, specific inhibitors of sialidases, heparanase, and cathepsin L have been shown to prevent both EGL disruption and endothelial hyperpermeability induced by NS1 .

The specificity of these pathogenic effects is notable - they are observed with NS1 from all four dengue virus serotypes (DENV1-4) but not with NS1 from the related West Nile virus . This suggests unique structural features of dengue NS1 that confer this pathogenic capability and may explain certain aspects of dengue-specific disease manifestations.

What experimental approaches can reveal the NS1 trafficking mechanisms in mosquito cells?

Investigating NS1 trafficking in mosquito cells requires specialized experimental approaches that can elucidate unconventional secretory pathways. Researchers employ a combination of biochemical, genetic, and microscopy-based techniques to characterize these mechanisms.

Methodologically, key experimental approaches include:

  • Pharmacological inhibition studies: Using inhibitors such as Brefeldin A (which blocks the classical ER-to-Golgi pathway) and methyl-β-cyclodextrin (which depletes cellular cholesterol) to identify pathway dependencies. The differential effects of these compounds provide evidence for an unconventional, cholesterol-dependent secretory pathway for NS1 in mosquito cells .

  • RNA interference (RNAi): Silencing genes involved in different secretory pathways (e.g., CAV1 for caveolae-dependent transport, SAR1 for classical secretion) to determine their role in NS1 secretion. Quantification of secreted NS1 after gene silencing helps identify essential pathway components .

  • Proximity ligation assays (PLA): This technique allows visualization of protein-protein interactions within cells with high specificity and sensitivity. PLA has demonstrated direct interaction between NS1 and CAV1 in infected mosquito cells, providing evidence for caveolin-dependent trafficking .

  • Glycosylation analysis: Comparing glycosylation patterns between secreted NS1 and other viral proteins (such as E protein) can reveal differences in secretory route. Different glycosylation profiles support the hypothesis that NS1 bypasses the Golgi complex during secretion in mosquito cells .

  • Computational modeling: Classical mechanics and docking simulations have suggested favorable interactions between the caveolin-binding domain in NS1 and the scaffolding domain of CAV1, providing theoretical support for experimental findings .

How might the differential secretion of NS1 in mosquito cells be targeted for vector control strategies?

The unique secretory pathway of NS1 in mosquito cells presents a potential target for novel vector control strategies aimed at interrupting dengue virus transmission. Understanding the mosquito-specific, caveolin-dependent secretion mechanism could lead to interventions that specifically target viral replication or transmission in the insect vector.

Methodologically, researchers could pursue several approaches:

  • Small molecule inhibitors: Developing compounds that specifically inhibit the chaperone caveolin complex (CAV1, FKBP52, CyA, and Cy40) in mosquito cells could potentially block NS1 secretion and disrupt the viral life cycle . This approach would require initial high-throughput screening of compound libraries followed by validation in mosquito cell cultures.

  • Genetic modification strategies: CRISPR-Cas9 or RNAi-based approaches could be used to modify or suppress expression of caveolin complex components in mosquito populations. Transgenic mosquitoes with reduced ability to support NS1 secretion might have decreased vector competence .

  • Peptide-based interventions: Designing peptides that mimic the interaction interface between NS1 and CAV1 could competitively inhibit this interaction and block the secretory pathway. Structural data from docking simulations would inform peptide design .

  • Cholesterol metabolism modulation: Since the unconventional secretory pathway in mosquito cells is cholesterol-dependent, strategies targeting cholesterol transport or metabolism specifically in mosquito tissues could indirectly impact NS1 secretion .

The development of such vector-targeted approaches would require careful consideration of specificity to avoid effects on non-target organisms, as well as strategies for delivery and persistence in mosquito populations. Experimental evaluation would initially focus on laboratory-controlled studies before moving to contained field trials.

What are the optimal methods for purifying functional NS1 for experimental studies?

Obtaining pure, functional NS1 protein is essential for studying its biological activities and interactions. Researchers utilize several approaches to purify NS1, each with advantages for specific experimental applications.

Methodologically, the following approaches are commonly employed:

  • Recombinant expression systems: NS1 can be expressed in various systems including E. coli, yeast, insect cells (using baculovirus), and mammalian cells. For studies requiring proper protein folding and post-translational modifications, eukaryotic expression systems are preferred. Insect cell expression (particularly using Sf9 or High Five cells) often provides a good balance between yield and proper protein processing.

  • Affinity tags: Engineering NS1 with affinity tags (His-tag, FLAG-tag, or GST) facilitates purification through affinity chromatography. The tag placement (N-terminal vs. C-terminal) should be optimized to minimize interference with protein function.

  • Secretion-based purification: For studies requiring naturally secreted NS1 hexamers, collection from the supernatant of infected or transfected cells provides material with authentic oligomeric structure. This approach is particularly valuable for studies on NS1-induced endothelial dysfunction.

  • Denaturation-renaturation protocols: When inclusion body formation occurs (particularly in bacterial systems), protocols involving denaturation followed by controlled refolding can recover functional protein.

  • Quality control: Critical validation steps include Western blotting, size exclusion chromatography to confirm oligomeric state, glycosylation analysis, and functional assays (such as lipoprotein binding or EGL disruption assays).

When studying NS1 from different flaviviruses (e.g., comparing dengue and West Nile virus NS1), consistent purification methodology is essential to ensure that observed functional differences are not artifacts of the purification process .

What cell culture models best represent in vivo NS1 interactions in mosquito vectors?

Selecting appropriate cell culture models is crucial for accurately representing the biological context of NS1 interactions in mosquito vectors. Different experimental questions may require specific cell types or culture conditions.

Methodologically, researchers should consider:

  • Mosquito cell lines: C6/36 (derived from Aedes albopictus larvae) is commonly used, but other options include Aag2 (from Aedes aegypti) and AP-61. These cell lines vary in their immune responses and virus susceptibility, so selection should align with research questions.

  • Primary cell cultures: While more challenging to establish, primary cultures from dissected mosquito tissues (midgut, salivary glands) may better represent the natural environment for virus-vector interactions.

  • 3D culture systems: Advanced models incorporating extracellular matrix components can better mimic tissue architecture and cell-cell interactions compared to traditional monolayer cultures.

  • Co-culture systems: Combining mosquito cells with vertebrate endothelial cells can model the transmission interface and allow study of NS1 effects across both host systems.

  • Physiologically relevant conditions: Maintaining cultures at temperatures that mimic the mosquito environment (typically 28°C rather than 37°C) and using appropriate media formulations improves model fidelity.

When studying NS1 secretion specifically, researchers should include appropriate controls for monitoring the classical secretory pathway (e.g., a protein known to follow the Golgi-dependent route) alongside NS1 to confirm pathway specificity .

How might comparative studies of NS1 across different flaviviruses inform therapeutic strategies?

Comparative analysis of NS1 proteins from different flaviviruses provides valuable insights into conserved and divergent features that could inform therapeutic development. Research has demonstrated that NS1 from dengue virus serotypes 1-4 induces endothelial glycocalyx disruption, while NS1 from West Nile virus does not produce this effect .

Methodologically, a comprehensive research approach would include:

  • Structural comparison: High-resolution structural analysis of NS1 from multiple flaviviruses (dengue, Zika, West Nile, yellow fever) to identify unique structural elements that correlate with pathogenic functions.

  • Domain swapping experiments: Creating chimeric NS1 proteins with domains from different flaviviruses to map the specific regions responsible for endothelial disruption or differential secretion pathways.

  • Evolutionary analysis: Phylogenetic studies combined with functional assays to understand how NS1 functions evolved across the flavivirus family and correlate with vector usage and disease manifestations.

  • Cross-reactivity studies: Investigating whether antibodies or small molecule inhibitors targeting NS1 from one flavivirus have activity against NS1 from other family members.

  • Conserved pathway targeting: Identifying whether there are common mechanisms or interaction partners that could serve as broad-spectrum therapeutic targets across multiple flavivirus infections.

This comparative approach could lead to the development of pan-flavivirus interventions or help explain the virus-specific pathologies observed in different flavivirus infections .

What are the emerging techniques for studying NS1 trafficking in live mosquitoes?

Advancing beyond cell culture models to study NS1 trafficking in intact mosquitoes represents an important frontier in understanding vector-pathogen interactions. New methodological approaches are enabling researchers to visualize and manipulate NS1 dynamics in living insects.

Methodologically, emerging techniques include:

  • Fluorescently tagged NS1: Genetically engineering dengue virus strains expressing fluorescent protein-tagged NS1 allows real-time visualization of protein trafficking in infected mosquitoes using confocal microscopy.

  • Intravital imaging: Adapting techniques from vertebrate models to visualize protein dynamics in living mosquito tissues through microscopic access windows or transparent cuticle areas.

  • Tissue-specific gene manipulation: Using mosquito-optimized CRISPR-Cas9 systems or conditional knockdown approaches to modulate caveolin complex components in specific tissues relevant to viral transmission.

  • Nanobody-based detection: Developing small antibody fragments (nanobodies) that can bind NS1 in living cells without interfering with function, potentially coupled with fluorescent reporters.

  • Artificial blood meal systems: Creating ex vivo systems that allow controlled delivery of virus or purified NS1 to mosquito midguts while maintaining the ability to monitor subsequent protein trafficking.

Product Science Overview

Introduction

Dengue virus (DENV) is a mosquito-borne flavivirus that causes dengue fever, a significant public health concern in tropical and subtropical regions. The virus has four distinct serotypes (DENV-1 to DENV-4), and infection with one serotype does not confer immunity against the others. The nonstructural protein 1 (NS1) is a multifunctional glycoprotein that plays a crucial role in the viral life cycle and pathogenesis.

Structure and Function of NS1

NS1 is a 46-55 kDa glycoprotein that exists in multiple oligomeric forms, including monomers, dimers, and hexamers. It is involved in various functions, such as viral replication, immune evasion, and pathogenesis. NS1 is secreted from infected cells and can be found in the bloodstream of infected individuals. The secreted form of NS1 (sNS1) is predominantly dimeric and plays a role in immune modulation and vascular leakage, a hallmark of severe dengue disease .

Recombinant NS1 Production in Insect Cells

Recombinant NS1 proteins are produced using various expression systems, including insect cells. Insect cell expression systems, such as Drosophila S2 cells, are commonly used due to their ability to perform post-translational modifications similar to those in mammalian cells. The production of recombinant NS1 in insect cells involves transfecting the cells with a plasmid encoding the NS1 gene, followed by protein expression and purification .

Applications of Recombinant NS1

Recombinant NS1 proteins are used in various research and diagnostic applications. They are employed in the development of diagnostic assays for dengue infection, as NS1 is a biomarker for early detection of the disease. Additionally, recombinant NS1 is used in vaccine development and studies investigating the immune response to dengue virus .

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.