Recombinant Viridovipera stejnegeri Stejaggregin-A subunit beta-2

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder

Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement. We will accommodate your request whenever possible.

Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.

Note: All proteins are shipped with standard blue ice packs unless dry ice shipping is specifically requested in advance. Additional fees apply for dry ice shipping.

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%, but this can be adjusted per customer requirements.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and the protein's inherent 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. Aliquoting is recommended for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.

The tag type is determined during production. If you require a specific tag type, please inform us, and we will prioritize its development.

Synonyms
Snaclec stejaggregin-A subunit beta-2
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
24-148
Protein Length
Full Length of Mature Protein
Purity
>85% (SDS-PAGE)
Species
Trimeresurus stejnegeri (Chinese green tree viper) (Viridovipera stejnegeri)
Target Protein Sequence
GFCCPLG WSSYDLYCYK VFKQQMNWTD AEKFCTEQHT GSHLVSFHSS EEADFVVNMT YPILKLDFVW IGLSNVWNQC NSEWSDGTKL DYKDWSGESE CIASKTVENQ WWTKSCSRTH YVVCKFQA
Uniprot No.

Target Background

Function

Interferes with a step in hemostasis, such as modulation of platelet aggregation or the coagulation cascade.

Protein Families
Snaclec family
Subcellular Location
Secreted.
Tissue Specificity
Expressed by the venom gland.

Q&A

What is Recombinant Viridovipera stejnegeri Stejaggregin-A subunit beta-2 and what are its structural characteristics?

Recombinant Viridovipera stejnegeri Stejaggregin-A subunit beta-2 is a protein subunit derived from the venom of the bamboo pitviper (Viridovipera stejnegeri). It is produced through recombinant technology rather than direct extraction from the snake . The protein belongs to the C-type lectin-like protein family commonly found in snake venoms and consists of a beta subunit that plays a role in various biological activities. The structure typically features conserved cysteine residues that form disulfide bridges critical for maintaining the protein's tertiary structure and function. Researchers should note that while the native protein exists as a heterodimeric complex, the recombinant beta-2 subunit is often expressed independently for specific research applications.

What expression systems are optimal for producing Recombinant Viridovipera stejnegeri Stejaggregin-A subunit beta-2?

Several expression systems can be utilized for the production of Recombinant Viridovipera stejnegeri Stejaggregin-A subunit beta-2, each with distinct advantages. Bacterial systems like E. coli offer high yield and cost-effectiveness but may struggle with proper disulfide bond formation. Mammalian expression systems, particularly HEK 293 cells, provide superior post-translational modifications and proper protein folding. Evidence from similar recombinant protein studies indicates that viral vector systems, such as adeno-associated viral (AAV) vectors, can achieve efficient expression with proper protein functionality . For instance, AAV-vector expression systems have demonstrated a 13- to 77-fold increase in receptor protein density compared to controls, with proper trafficking and signaling capabilities, suggesting similar approaches may be valuable for Stejaggregin-A subunit beta-2 expression .

How can researchers validate the biological activity of recombinant Stejaggregin-A subunit beta-2?

Validating biological activity requires multiple complementary approaches. Binding assays using radiolabeled ligands similar to the [3H]DHA binding technique used for beta2AR can determine if the recombinant protein maintains appropriate binding kinetics . Functional assays should examine the protein's effects on platelet aggregation, as C-type lectin proteins from snake venoms often target platelet receptors. Cell-based assays measuring downstream signaling pathways provide further validation. Competition assays with known ligands can establish binding specificity and rank order of potency, as demonstrated in beta2AR research where isoproterenol>epinephrine>norepinephrine potency was established . Importantly, researchers should include appropriate positive and negative controls and perform dose-response experiments to fully characterize the recombinant protein's activity profile.

What experimental design approaches are most effective for investigating Stejaggregin-A subunit beta-2 interactions with cellular targets?

The most effective experimental designs for investigating Stejaggregin-A subunit beta-2 interactions utilize multi-factorial approaches. Two-level factorial experiments are particularly valuable in the early stages of research when investigating multiple potential factors to identify the "vital few" that significantly impact protein-target interactions . This approach allows researchers to quickly filter out insignificant effects while focusing on important variables such as pH, temperature, ionic strength, and cofactor requirements. For instance, a 2^k design (where k represents the number of factors) enables systematic evaluation of all possible combinations of experimental factors at high and low levels . When studying Stejaggregin-A subunit beta-2, researchers should consider factors including protein concentration, target cell type, incubation time, and presence of competing ligands. This design approach minimizes experimental runs while maximizing information yield about interaction parameters.

How do researchers address discrepancies between in vitro and in vivo findings when studying Recombinant Viridovipera stejnegeri Stejaggregin-A subunit beta-2?

Addressing discrepancies between in vitro and in vivo findings requires a systematic approach to identify factors that differ between systems. First, researchers should examine protein stability and degradation kinetics in both environments. The protein may have different half-lives or undergo different post-translational modifications in vivo. Second, tissue-specific effects should be considered – data from studies on sVEGFR-2 demonstrate that tissue-specific loss of function can induce spontaneous lymphatic development in some tissues without affecting blood vasculature . This suggests microenvironment-specific effects that may not be replicated in simplified in vitro systems. Third, researchers should consider biological complexity factors such as the presence of endogenous inhibitors or enhancers in vivo that are absent in vitro. Finally, dosing and pharmacokinetic considerations significantly impact in vivo outcomes. A structured approach using paired in vitro and in vivo experiments with identical readouts can help identify specific variables responsible for discrepancies.

What methodological approaches can distinguish between specific and non-specific effects of Stejaggregin-A subunit beta-2 in experimental systems?

Distinguishing between specific and non-specific effects requires rigorous experimental controls and validation techniques. First, dose-response relationships should be established – specific effects typically show saturable dose-dependent responses, while non-specific effects often exhibit linear relationships. Second, competitive binding assays with structurally related proteins can demonstrate binding specificity. Third, researchers should employ molecular approaches such as site-directed mutagenesis of key binding residues, which can confirm whether observed effects depend on specific structural elements. A powerful approach is to use receptor knockdown/knockout systems alongside wild-type comparisons, similar to methods used in VEGFR-2 splice variant research . Additionally, researchers can utilize domain-specific antibodies to block particular regions of the protein and observe whether specific activities are inhibited. Finally, cross-validation across multiple experimental systems (different cell types, in vitro vs. ex vivo models) can help establish consistency of specific effects.

How should researchers design experiments to investigate the molecular mechanisms of Stejaggregin-A subunit beta-2?

When designing experiments to investigate molecular mechanisms, researchers should employ a systematic approach that combines structural analysis, functional assays, and mechanistic studies. Begin with computational predictions of protein structure and potential binding sites, followed by experimental validation through techniques like circular dichroism and X-ray crystallography. For functional characterization, employ surface plasmon resonance to measure binding kinetics to potential targets. Cellular assays should include both target cells (e.g., platelets) and negative control cells lacking the suspected receptor. Signal transduction studies should examine changes in second messengers (similar to cAMP measurements used in beta2AR research where isoproterenol-stimulated cAMP levels were 5-fold higher in infected cells compared to controls ). Protein trafficking studies using fluorescently-tagged constructs can reveal internalization dynamics, comparable to the receptor trafficking demonstrated for beta2AR . For all experiments, proper data collection requires table formats that clearly distinguish independent variables (left column) from dependent variables (subsequent columns) with appropriate units and statistical treatments .

What approaches help resolve contradictory data when studying Recombinant Viridovipera stejnegeri Stejaggregin-A subunit beta-2?

Resolving contradictory data requires a systematic troubleshooting approach. First, researchers should verify protein quality using techniques such as mass spectrometry, circular dichroism, and size-exclusion chromatography to confirm proper folding and absence of aggregation. Second, experimental conditions should be standardized across laboratories, including buffer composition, temperature, and incubation times. Third, researchers should consider biological variability – for example, studies on Viridovipera stejnegeri gut microbiota have shown significant differences in microbial composition across populations despite no significant variance in α-diversity , suggesting that geographical source of the original protein sequence could impact results. Fourth, method validation should ensure that assays are measuring what they claim to measure through positive and negative controls. Finally, researchers should employ meta-analysis techniques to identify patterns across multiple studies that may explain apparent contradictions. All of these approaches should be documented in a standardized format to facilitate comparison between experiments.

How can researchers optimize assay conditions for studying Stejaggregin-A subunit beta-2 interactions?

Optimization of assay conditions requires systematic evaluation of multiple parameters. The table below outlines a methodical approach to optimizing key variables:

ParameterRange to TestMeasurement MethodOptimization Goal
pH5.0-9.0 (0.5 increments)Activity assay & binding affinityMaximum signal-to-noise ratio
Temperature4°C, 25°C, 37°CBinding kinetics & stability measurementsBalance between stability and activity
Ionic strength50-300 mM NaClProtein solubility & target bindingMinimal non-specific binding
Divalent cations0-10 mM Ca²⁺, Mg²⁺Activity assays with/without EDTADetermine cofactor requirements
Incubation time5 min to 24 hoursTime-course of binding/activityDetermine equilibrium conditions

Researchers should employ a two-level factorial design approach initially to identify significant factors , followed by response surface methodology to find optimal conditions. For each condition, researchers should measure both protein stability (using techniques like differential scanning fluorimetry) and functional activity to identify conditions that maintain the protein in its native conformation while allowing optimal target interaction. Control experiments using heat-denatured protein should be included to establish baselines for non-specific effects. Optimized conditions should be validated across multiple protein batches to ensure reproducibility.

What methodologies are most effective for studying potential therapeutic applications of Stejaggregin-A subunit beta-2?

The most effective methodologies for studying therapeutic applications combine in vitro mechanistic studies with appropriate disease models. Initially, researchers should establish mechanism of action through target identification (e.g., using affinity chromatography or proteomics approaches) and characterize dose-response relationships in relevant cell types. For vascular applications, drawing from VEGFR research methodologies, researchers should examine effects on endothelial cell proliferation, migration, and tube formation . Moving to in vivo models, researchers can employ approaches similar to those used in VEGFR-2 splice variant studies, which demonstrated that tissue-specific protein expression affected lymphatic vessel development without altering blood vasculature . Disease-specific models should be selected based on the protein's mechanism of action – for example, thrombosis models if the protein affects platelet aggregation. For all in vivo studies, researchers must carefully document administration routes, dosing, pharmacokinetics, and potential immunogenicity. Finally, safety assessment should include examination of off-target effects through comprehensive tissue analysis and biomarker measurements.

How should researchers approach comparative studies between native and recombinant Stejaggregin-A?

Comparative studies between native and recombinant Stejaggregin-A require carefully designed experiments addressing multiple parameters. Begin with structural comparisons using techniques such as circular dichroism, mass spectrometry, and peptide mapping to identify any differences in post-translational modifications or disulfide bond arrangements. Functional comparisons should employ identical assay conditions for both proteins, measuring dose-response relationships rather than single-point comparisons. When discrepancies are observed, researchers should systematically investigate potential causes, including differences in purity, stability, or conformational states. To ensure comparable activity measurements, standard reference materials should be established and shared between laboratories. One effective approach is to create a table comparing multiple parameters between native and recombinant forms:

ParameterNative Stejaggregin-ARecombinant Stejaggregin-A subunit beta-2Analytical Method
Molecular weightExpected complex massSubunit massMass spectrometry
Glycosylation profileNative patternExpression system-dependentGlycan analysis
Binding affinityBaseline KdComparative KdSurface plasmon resonance
EC50 in functional assaysBaseline potencyComparative potencyCell-based assays
Stability (t1/2)Native half-lifeRecombinant half-lifeAccelerated stability testing

This systematic comparison helps researchers understand the strengths and limitations of using the recombinant protein as a research tool or potential therapeutic.

What are the key challenges in maintaining structural integrity of Recombinant Viridovipera stejnegeri Stejaggregin-A subunit beta-2 during experimentation?

Maintaining structural integrity presents several challenges that researchers must address methodically. First, disulfide bond integrity is critical – C-type lectin proteins typically contain multiple disulfide bridges essential for proper folding. Researchers should avoid reducing agents in buffers and consider adding stabilizing agents like calcium ions. Second, aggregation during storage and freeze-thaw cycles can compromise activity; researchers should validate protein behavior using dynamic light scattering and size-exclusion chromatography after various storage conditions. Third, proteolytic degradation can occur during expression and purification; adding protease inhibitors and optimizing purification strategies can minimize this issue. Fourth, adsorption to surfaces can reduce effective concentration; pre-treating containers with blocking proteins or using low-binding materials can address this challenge. Finally, batch-to-batch variation can introduce inconsistencies; implementing rigorous quality control protocols that assess multiple parameters (not just total protein concentration) helps ensure experimental reproducibility.

How can researchers address the challenge of target specificity when studying Stejaggregin-A subunit beta-2 interactions?

Addressing target specificity challenges requires multiple complementary approaches. First, researchers should employ binding studies with purified potential receptors to establish direct interactions. Surface plasmon resonance can provide quantitative binding parameters and help distinguish specific from non-specific binding. Second, competition assays with structurally related proteins can reveal binding site specificity, similar to the agonist competition assays performed with beta2AR that established rank order potency . Third, cell-based assays comparing responses in cells expressing or lacking putative targets can confirm receptor-dependent effects. Researchers can use CRISPR-Cas9 technology to generate knockout cell lines for this purpose. Fourth, structure-function studies using truncated or mutated versions of both Stejaggregin-A subunit beta-2 and its putative targets can map interaction domains. Finally, in vivo studies examining tissue-specific effects can reveal physiological relevance, as demonstrated in VEGFR-2 splice variant research where tissue-specific protein expression affected specific vascular beds . Addressing target specificity is crucial for understanding molecular mechanisms and developing potential therapeutic applications.

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.