SDF2 Human, sf9

Stromal Cell-Derived Factor 2, Sf9 Human Recombinant
Shipped with Ice Packs
In Stock

Description

SDF2 produced in Sf9 Baculovirus cells is a single, glycosylated polypeptide chain containing 202 amino acids (19-211a.a.) and having a molecular mass of 22.3kDa. (Molecular size on SDS-PAGE will appear at approximately 18-28kDa). SDF2 is expressed with an 9 amino acid His tag at C-Terminus and purified by proprietary chromatographic techniques.

Product Specs

Introduction
Stromal Cell-Derived Factor 2 (SDF2) is a secretory protein that shares partial similarity with the hydrophilic segments of yeast mannosyltransferases. SDF2 protein is ubiquitously expressed, and the corresponding gene exhibits significant conservation among mammals. SDF2 is a protein-coding gene with alternative splicing, resulting in both coding and non-coding variants.
Description
SDF2, produced in Sf9 Baculovirus cells, is a single, glycosylated polypeptide chain comprising 202 amino acids (19-211a.a.) with a molecular mass of 22.3 kDa. On SDS-PAGE, the molecular size appears approximately between 18-28 kDa. SDF2 is expressed with a 9 amino acid His tag at the C-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered colorless solution.
Formulation
SDF2 protein solution (0.25 mg/ml) is formulated in 50 mM Tris-HCl (pH 8.0), 10% glycerol, 0.1 M NaCl, 0.1 mM PMSF, and 0.5 mM EDTA.
Stability
For short-term storage (2-4 weeks), store at 4°C. For long-term storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Greater than 95.0% purity as determined by SDS-PAGE.
Synonyms

Stromal Cell Derived Factor 2, Stromal Cell-Derived Factor 2, SDF-2 

Source
Sf9, Baculovirus cells.
Amino Acid Sequence

ADPSSLGVVT CGSVVKLLNT RHNVRLHSHD VRYGSGSGQQ SVTGVTSVDD SNSYWRIRGK SATVCERGTP IKCGQPIRLT HVNTGRNLHS HHFTSPLSGN QEVSAFGEEG EGDYLDDWTV LCNGPYWVRD GEVRFKHSST EVLLSVTGEQ YGRPISGQKE VHGMAQPSQN NYWKAMEGIF MKPSELLKAE AHHAELHHHH HH

Q&A

What is SDF2 Human, sf9 and what are its key structural characteristics?

SDF2 Human produced in Sf9 baculovirus cells is a single, glycosylated polypeptide chain containing 202 amino acids (positions 19-211) with a molecular mass of 22.3kDa. When analyzed via SDS-PAGE, it typically appears at approximately 18-28kDa due to glycosylation patterns. The protein is expressed with a 9-amino acid His tag at the C-terminus to facilitate purification .

SDF2 is a secretory protein that shares structural similarities with hydrophilic segments of yeast mannosyltransferases. Its expression is ubiquitous throughout human tissues, and the gene sequence is highly conserved among mammals, suggesting evolutionary importance . The protein undergoes alternative splicing, resulting in both coding and non-coding variants that may serve different biological functions.

How does the Sf9 expression system compare to other protein expression platforms for human SDF2?

The Sf9 insect cell system offers several advantages for SDF2 expression compared to bacterial or mammalian cell systems. Unlike bacterial expression systems such as E. coli, Sf9 cells can perform complex post-translational modifications including glycosylation, which is crucial for maintaining SDF2's native structure and function .

While mammalian cell lines might offer more human-like glycosylation patterns, the Sf9 baculovirus system provides higher protein yields and scalability for research purposes. The BEVS (Baculovirus Expression Vector System) in Sf9 cells has been optimized through experimental design approaches such as Placket-Burman design and Box-Behnken methods to identify critical parameters affecting protein expression, including feed percentage, cell count, and multiplicity of infection . These optimizations make Sf9 an efficient system for producing research-grade SDF2 with consistent quality.

What purification methods are most effective for SDF2 Human expressed in Sf9 cells?

Purification of His-tagged SDF2 from Sf9 cell culture typically employs a multi-step chromatographic approach. The initial capture step usually utilizes immobilized metal affinity chromatography (IMAC) with Ni-NTA or similar matrices that bind the C-terminal His tag . This is often followed by additional purification steps such as ion exchange chromatography or size exclusion chromatography to achieve higher purity.

For optimal results, researchers should consider:

  • Maintaining reducing conditions throughout purification to prevent disulfide bond formation

  • Using protease inhibitors in lysis buffers to prevent degradation

  • Optimizing buffer conditions (pH, salt concentration) at each purification step

  • Performing quality control via SDS-PAGE and Western blotting to confirm identity and purity

  • Considering the removal of the His tag using specific proteases if the tag might interfere with downstream applications

The purification strategy should be tailored to the specific research application, with additional considerations for maintaining protein stability and activity throughout the process.

What are the optimal storage conditions for maintaining SDF2 Human, sf9 stability?

Based on manufacturer recommendations, purified SDF2 Human, sf9 should be stored at either -20°C for long-term storage or at 4°C for short-term use . To maintain protein stability and activity, consider the following guidelines:

  • Store the protein in small aliquots to avoid repeated freeze-thaw cycles

  • Include stabilizing agents such as glycerol (10-25%) for frozen storage

  • Use appropriate buffer systems (typically phosphate or Tris-based) at physiological pH

  • Consider adding reducing agents if the protein contains cysteine residues

  • For extended storage periods, lyophilization may be considered, though refolding efficiency should be tested

Stability studies should be performed to determine the specific shelf-life under your laboratory's storage conditions, with periodic activity assays to confirm protein functionality over time.

How can I optimize the expression of SDF2 Human in Sf9 cells for specific research applications?

Optimizing SDF2 expression in Sf9 cells requires systematic evaluation of multiple parameters. Research indicates that a design of experiments (DOE) approach can significantly improve protein yield and quality . Key parameters to consider include:

  • Cell culture conditions:

    • Cell density at infection (typically 1-2×10^6 cells/mL)

    • Culture medium composition and supplementation

    • Temperature (usually 27-28°C for Sf9 cells)

    • Dissolved oxygen and pH levels

  • Infection parameters:

    • Multiplicity of infection (MOI) - the ratio of virus particles to cells

    • Time of harvest post-infection (typically 48-72 hours)

    • Feed percentage and feeding strategy for high-density cultures

  • Vector design:

    • Promoter selection (polyhedrin or p10 promoters are commonly used)

    • Signal peptide optimization for secreted expression

    • Codon optimization for insect cell expression

Statistical experimental designs such as Box-Behnken can help identify interactions between parameters that might not be apparent in one-factor-at-a-time approaches. For example, studies with similar proteins have shown that feed percentage, cell count, and MOI significantly affect recombinant protein expression levels and bioactivity .

What analytical methods can determine the structural integrity and functionality of purified SDF2 Human, sf9?

Comprehensive characterization of SDF2 Human requires multiple analytical approaches:

  • Structural analysis:

    • Circular dichroism (CD) spectroscopy to assess secondary structure

    • Fourier-transform infrared spectroscopy (FTIR) for complementary structural information

    • Mass spectrometry to confirm molecular weight and identify post-translational modifications

    • Differential scanning calorimetry (DSC) to evaluate thermal stability

    • Size exclusion chromatography with multi-angle light scattering (SEC-MALS) to assess oligomeric state

  • Functional analysis:

    • Surface plasmon resonance (SPR) to quantify binding interactions with known partners

    • Enzyme-linked immunosorbent assay (ELISA) for detecting specific interactions

    • Cell-based assays relevant to SDF2's biological functions

    • Glycoanalysis to characterize and compare glycosylation patterns with native human SDF2

  • Quality control:

    • Endotoxin testing to ensure preparation is suitable for cell culture applications

    • Aggregation analysis via dynamic light scattering (DLS)

    • Stability testing under various storage conditions

By combining these approaches, researchers can develop a comprehensive profile of the recombinant SDF2 and determine whether it faithfully represents the native human protein in terms of structure and function.

How does glycosylation of SDF2 in Sf9 cells differ from native human glycosylation patterns, and what implications does this have for research?

Glycosylation patterns in Sf9 insect cells differ significantly from human cells, which has important implications for SDF2 research:

Sf9 cells typically produce simpler, high-mannose type N-glycans lacking the complex terminal sialylation seen in human cells. For SDF2, which is known to be glycosylated, these differences may affect:

  • Protein folding and stability - glycosylation contributes to proper folding and can protect against proteolytic degradation

  • Biological activity - if glycans are involved in receptor recognition or binding

  • Immunogenicity - altered glycans may elicit different immune responses

  • Pharmacokinetics - clearance rates may differ for proteins with insect vs. human glycosylation

Researchers working with SDF2 Human, sf9 should consider these limitations, particularly when:

  • Studying receptor-ligand interactions

  • Performing in vivo studies where glycosylation affects circulation half-life

  • Investigating structure-function relationships

For applications where native human glycosylation is critical, alternative expression systems such as human cell lines (HEK293, CHO) or glycoengineered insect cell lines should be considered, despite potentially lower yields compared to standard Sf9 systems .

What approaches can be used to investigate SDF2's role in protein quality control and ER stress pathways?

SDF2 has been implicated in endoplasmic reticulum (ER) quality control and stress response pathways. To investigate these functions using recombinant SDF2 Human, sf9, consider the following methodological approaches:

  • Protein interaction studies:

    • Co-immunoprecipitation with known ER chaperones and folding machinery

    • Proximity labeling techniques (BioID, APEX) to identify novel interaction partners

    • Yeast two-hybrid screening using SDF2 as bait

  • Cellular stress response assays:

    • Measure unfolded protein response (UPR) markers after SDF2 knockdown/overexpression

    • Quantify ER stress-induced apoptosis in presence/absence of functional SDF2

    • Monitor calcium homeostasis as SDF2 may influence ER calcium storage

  • Structural analysis of SDF2-substrate complexes:

    • Cryo-electron microscopy of SDF2 in complex with client proteins

    • Hydrogen-deuterium exchange mass spectrometry to map interaction surfaces

    • FRET-based assays to detect conformational changes during substrate binding

  • In vitro reconstitution:

    • Reconstitute minimal protein folding systems with purified components

    • Assess SDF2's ability to prevent aggregation of model substrates

    • Determine whether SDF2 acts as a holdase or foldase chaperone

These approaches should be complemented with appropriate controls, including comparison to native human SDF2 where possible, to account for any functional differences due to the Sf9 expression system.

How can SDF2 Human, sf9 be used in drug discovery and therapeutic development research?

SDF2 Human, sf9 can serve as a valuable tool in several aspects of drug discovery:

  • Target validation:

    • Use purified SDF2 in binding assays to confirm interaction with potential drug candidates

    • Develop SDF2-based cellular assays for high-throughput screening

    • Structure-based drug design leveraging the purified protein for crystallization studies

  • Therapeutic protein development:

    • Engineer SDF2 variants with enhanced stability or novel functions

    • Study structure-function relationships to identify critical domains

    • Develop SDF2-based fusion proteins or conjugates with specific targeting properties

  • Mechanism of action studies:

    • Investigate SDF2's role in cellular stress pathways and protein homeostasis

    • Explore connections between SDF2 dysfunction and disease states

    • Identify potential intervention points in SDF2-related pathways

  • Biomarker development:

    • Generate antibodies against SDF2 using the purified protein as an immunogen

    • Develop quantitative assays for SDF2 detection in biological samples

    • Correlate SDF2 levels or modifications with disease progression

For these applications, researchers should carefully consider the limitations of Sf9-expressed SDF2, particularly regarding glycosylation differences compared to the native human protein.

What experimental considerations are important when using antibodies to detect SDF2 Human in different experimental contexts?

When developing or using antibodies against SDF2 Human, researchers should consider:

  • Epitope selection and antibody specificity:

    • Determine whether antibodies recognize linear or conformational epitopes

    • Verify cross-reactivity with native human SDF2 vs. Sf9-expressed SDF2

    • Test for potential cross-reactivity with related proteins

    • Validate antibody performance in multiple applications (WB, IP, IHC, Flow)

  • Detection strategies:

    • For Western blotting, SDF2 typically appears at 18-28kDa due to glycosylation

    • For immunoprecipitation, consider whether the His-tag might interfere with antibody binding

    • For immunohistochemistry, optimize fixation methods to preserve epitope accessibility

    • For ELISA, determine optimal coating concentration and blocking conditions

  • Controls and validation:

    • Include recombinant SDF2 as a positive control

    • Use SDF2 knockout or knockdown samples as negative controls

    • Perform peptide competition assays to confirm specificity

    • Validate with multiple antibodies targeting different regions of SDF2

  • Quantification considerations:

    • Establish standard curves using purified SDF2 Human, sf9

    • Consider the impact of post-translational modifications on antibody recognition

    • Evaluate detection sensitivity and dynamic range for your specific application

Properly validated antibodies are essential for reliable SDF2 detection and quantification in research applications.

How can SDF2 Human, sf9 be incorporated into co-immunoprecipitation studies to identify novel protein interactions?

Co-immunoprecipitation (Co-IP) is a powerful technique for studying protein-protein interactions. When using SDF2 Human, sf9 in Co-IP experiments, consider the following methodological approach:

  • Experimental design:

    • Forward approach: Immobilize anti-SDF2 antibodies to pull down SDF2 and its binding partners

    • Reverse approach: Use antibodies against suspected interaction partners to co-precipitate SDF2

    • Direct pull-down: Utilize the His-tag on recombinant SDF2 for direct capture via Ni-NTA beads

  • Sample preparation:

    • Cell lysate preparation with appropriate buffers that preserve protein-protein interactions

    • Consider crosslinking to stabilize transient interactions

    • Include protease and phosphatase inhibitors to prevent degradation

    • Test different detergent conditions to optimize solubilization while maintaining interactions

  • Controls and validation:

    • Input controls to verify protein expression levels

    • IgG or other irrelevant antibody controls to identify non-specific binding

    • Competitive elution with excess antigen to confirm specificity

    • Reciprocal Co-IPs to verify interactions from both directions

  • Detection methods:

    • Western blotting with specific antibodies for suspected interaction partners

    • Mass spectrometry for unbiased identification of the complete interactome

    • Proximity ligation assays to confirm interactions in situ

Using these approaches, researchers can systematically map the SDF2 interactome and identify potential functional relationships in various cellular contexts.

What are common challenges in achieving high-yield SDF2 expression in Sf9 cells, and how can they be addressed?

Researchers often encounter several challenges when expressing SDF2 in Sf9 cells:

  • Low expression yields:

    • Optimize infection parameters (MOI, timing) through systematic testing

    • Consider codon optimization for insect cell preference

    • Evaluate different promoters (polyhedrin vs. p10)

    • Implement fed-batch culture strategies to extend cell viability

  • Protein degradation:

    • Add protease inhibitors to culture media and during purification

    • Optimize harvest timing to capture peak expression before degradation

    • Consider secretion into protein-free media to simplify purification

    • Test stabilizing additives in the culture medium

  • Incorrect folding or aggregation:

    • Adjust culture temperature (lower temperatures often improve folding)

    • Co-express chaperones or foldases that may assist SDF2 folding

    • Modify cell density at infection to reduce cellular stress

    • Evaluate different signal sequences for secretion efficiency

  • Inconsistent glycosylation:

    • Implement quality control measures to verify batch-to-batch consistency

    • Consider site-directed mutagenesis to eliminate problematic glycosylation sites

    • Explore enzymatic treatments for glycan homogenization if required

Systematic optimization using design of experiments (DOE) approaches has proven effective for similar proteins expressed in Sf9 cells, with feed percentage, cell count, and multiplicity of infection identified as key parameters affecting expression levels and protein quality .

How can researchers address discrepancies between experimental results using SDF2 Human, sf9 and native human SDF2?

When experimental results with recombinant SDF2 Human, sf9 differ from those observed with native human SDF2, consider the following analytical and experimental approaches:

  • Structural comparison:

    • Perform comparative circular dichroism (CD) spectroscopy to assess secondary structure differences

    • Use differential scanning calorimetry to compare thermal stability profiles

    • Conduct detailed glycoanalysis to characterize differences in post-translational modifications

    • Consider native mass spectrometry to evaluate oligomeric states

  • Functional assessment:

    • Develop quantitative binding assays to compare interaction kinetics with known partners

    • Implement cell-based functional assays with appropriate positive and negative controls

    • Perform side-by-side activity comparisons under varying conditions (pH, temperature, salt)

  • Experimental design modifications:

    • Introduce additional controls specific to the expression system

    • Consider using alternative expression systems (mammalian cells) for comparison

    • Implement domain-based studies to isolate regions critical for specific functions

    • Develop correction factors or normalization methods for quantitative comparisons

  • Data interpretation:

    • Acknowledge system limitations in publications and reports

    • Clearly state which form of SDF2 was used in each experiment

    • Consider whether observed differences reveal new insights about SDF2 biology

    • Validate key findings with native protein when possible

By systematically investigating discrepancies, researchers can determine whether differences are methodological artifacts or biologically meaningful variations that provide insights into SDF2 function.

What strategies can improve the solubility and stability of SDF2 Human, sf9 during experimental manipulation?

Maintaining SDF2 solubility and stability is crucial for reliable experimental results. Consider these methodological approaches:

  • Buffer optimization:

    • Screen different buffer compositions (phosphate, Tris, HEPES) at various pH values

    • Test stabilizing additives such as glycerol, sucrose, or specific amino acids

    • Evaluate the effect of salt concentration on solubility and activity

    • Consider the addition of mild detergents below their critical micelle concentration

  • Storage and handling:

    • Determine optimal protein concentration to prevent concentration-dependent aggregation

    • Establish appropriate freeze-thaw protocols or avoid multiple freeze-thaw cycles

    • Investigate lyophilization conditions if long-term storage is required

    • Implement quality control testing before each experimental use

  • Modification strategies:

    • Consider protein engineering to remove aggregation-prone regions

    • Explore fusion partners that enhance solubility (SUMO, MBP, thioredoxin)

    • Evaluate chemical modification approaches (PEGylation) for improved stability

    • Investigate stabilizing formulations with pharmaceutical excipients

  • Analytical monitoring:

    • Implement size exclusion chromatography to monitor aggregation states

    • Use dynamic light scattering for rapid assessment of solution homogeneity

    • Develop activity assays to confirm functional integrity after manipulation

    • Consider thermal shift assays to identify stabilizing conditions

By systematically optimizing these parameters, researchers can develop robust protocols for handling SDF2 Human, sf9 in various experimental contexts.

What are the most promising future research directions for SDF2 Human studies utilizing Sf9-expressed protein?

Several promising research avenues exist for SDF2 Human studies:

  • Structure-function relationships:

    • High-resolution structural determination through X-ray crystallography or cryo-EM

    • Mapping of functional domains through systematic mutagenesis

    • Investigation of SDF2's role in protein-protein interaction networks

    • Correlation of structural elements with cellular functions

  • Therapeutic applications:

    • Exploration of SDF2's potential role in managing ER stress-related diseases

    • Development of SDF2-derived peptides with therapeutic properties

    • Investigation of SDF2 as a biomarker for specific pathological conditions

    • Creation of modified SDF2 variants with enhanced stability or function

  • Comparative biology:

    • Evolutionary analysis of SDF2 across species to identify conserved functional regions

    • Investigation of tissue-specific functions in different physiological contexts

    • Understanding SDF2's role in various stress response pathways

  • Advanced methodologies:

    • Development of SDF2-specific biosensors for real-time monitoring in living cells

    • Application of integrative structural biology approaches combining multiple techniques

    • Utilization of systems biology to position SDF2 within broader cellular networks

These research directions can leverage the advantages of Sf9-expressed SDF2 while acknowledging and addressing the limitations of this expression system.

How can researchers effectively compare and integrate findings from different expression systems for SDF2 Human studies?

To effectively compare and integrate findings across different expression systems:

  • Systematic comparative analysis:

    • Establish standardized assays to evaluate SDF2 from different sources

    • Directly compare structural features (secondary structure, glycosylation, thermal stability)

    • Conduct side-by-side functional assays under identical conditions

    • Maintain detailed records of expression system-specific modifications

  • Data integration strategies:

    • Develop normalization methods to account for system-specific variations

    • Create comprehensive databases documenting system-dependent characteristics

    • Implement meta-analysis approaches to identify consistent findings across systems

    • Establish minimum reporting standards for SDF2 research publications

  • Collaborative approaches:

    • Establish research consortia using complementary expression systems

    • Develop shared resources and standardized materials

    • Implement round-robin testing to evaluate inter-laboratory variability

    • Create open access repositories for protocols and raw data

  • Advanced modeling:

    • Develop in silico approaches to predict system-specific variations

    • Create computational models that integrate data from multiple sources

    • Implement machine learning to identify patterns across diverse datasets

    • Establish predictive frameworks to translate between expression systems

By systematically addressing these aspects, researchers can build a more comprehensive understanding of SDF2 biology that transcends the limitations of any single expression system.

Product Science Overview

Gene and Protein Structure

The SDF2 gene is a protein-coding gene that undergoes alternative splicing, resulting in both coding and non-coding variants . The protein produced by this gene is believed to be a secretory protein, which means it is synthesized in the cell and then secreted out to perform its functions .

Recombinant SDF2 Production

Recombinant SDF2 is produced in Sf9 Baculovirus cells. The recombinant protein is a single, glycosylated polypeptide chain containing 202 amino acids (19-211a.a.) and has a molecular mass of approximately 22.3 kDa . The molecular size on SDS-PAGE appears at approximately 18-28 kDa . This recombinant protein is expressed with a 9 amino acid His tag at the C-terminus and is purified using proprietary chromatographic techniques .

Physical and Chemical Properties

The recombinant SDF2 protein is provided as a sterile filtered colorless solution. The formulation typically contains 50mM Tris-HCl (pH 8.0), 10% glycerol, 0.1M NaCl, 0.1mM PMSF, and 0.5mM EDTA . It is recommended to store the protein at 4°C if it will be used within 2-4 weeks, or at -20°C for longer periods. For long-term storage, adding a carrier protein (0.1% HSA or BSA) is advised to avoid multiple freeze-thaw cycles .

Applications and Usage

SDF2 has various applications in laboratory research. It is used in studies related to cell signaling, protein interactions, and other cellular processes. The protein’s high purity (greater than 95.0% as determined by SDS-PAGE) makes it suitable for various biochemical assays and experiments .

Safety and Handling

ProSpec’s recombinant SDF2 is intended for laboratory research use only. It is not to be used as drugs, agricultural or pesticidal products, food additives, or household chemicals .

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.