FAS Human Recombinant produced in Baculovirus is a single glycosylated polypeptide chain containing 156 amino acids (26-173 aa) and having a molecular mass of 17.7KDa.
FAS is fused to a 8 amino acid His-Tag at C-terminus and purified by proprietary chromatographic techniques.
FAS, also known as tumor necrosis factor receptor superfamily member 6 (TNFRSF6), belongs to the death receptor family within the TNF receptor protein family. This protein plays a critical role in regulating viral infections. FAS is expressed in virtually all cell types, while its corresponding ligand, FasL, is found in activated T cells, natural killer (NK) cells, and dendritic cells. The regulation of FasL and TRAIL on human cytomegalovirus (HCMV)-infected dendritic cells enhances the direct elimination of activated T lymphocytes. Moreover, the activation of FasL in HCMV-infected retinal pigment epithelial cells can contribute to the impairment of neutrophil function in HCMV retinitis.
Recombinant human FAS protein, expressed in Baculovirus, is a single glycosylated polypeptide chain. It consists of 156 amino acids (spanning residues 26-173) and has a molecular weight of 17.7 kDa. The protein includes an 8-amino acid His-tag fused at the C-terminus. Purification is achieved through proprietary chromatographic techniques.
The FAS protein solution has a concentration of 0.5 mg/ml. It is formulated in a solution containing 10% glycerol and Phosphate-Buffered Saline (PBS) at a pH of 7.4.
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To ensure optimal stability during long-term storage, adding a carrier protein (such as 0.1% HSA or BSA) is advisable. It's important to avoid repeated cycles of freezing and thawing.
The purity of the FAS protein is determined to be greater than 95.0% using SDS-PAGE analysis.
Tumor necrosis factor receptor superfamily member 6, Apo-1 antigen, Apoptosis-mediating surface antigen FAS, FASLG receptor, CD95, FAS, APT1, FAS1, APO-1, FASTM, ALPS1A, TNFRSF6.
Sf9, Baculovirus cells.
QVTDINSKGL ELRKTVTTVE TQNLEGLHHD GQFCHKPCPP GERKARDCTV NGDEPDCVPC QEGKEYTDKA HFSSKCRRCR LCDEGHGLEV EINCTRTQNT KCRCKPNFFC NSTVCEHCDP CTKCEHGIIK ECTLTSNTKC KEEGSRSNLE HHHHHH
Sf9 insect cells offer several significant advantages for recombinant human FasL production. The baculovirus-Sf9 system provides high-level expression of functionally active proteins with proper folding and appropriate post-translational modifications. Researchers have successfully produced human FasL under the control of a polyhedrin promoter in this system, yielding sufficient quantities to induce apoptosis in target cells such as T98G human glioblastoma cell lines .
Additional advantages include:
Scalable production capabilities for larger experimental needs
Growth in suspension culture at room temperature without CO2 requirements
Cost-effectiveness compared to mammalian expression systems
Ability to accommodate large protein constructs
Simplified purification through available affinity tag systems
The system's effectiveness is evidenced by the successful expression of functional FasL that retains apoptosis-inducing capabilities when tested against target cells .
Baculovirus vectors in Sf9 cells offer distinct advantages over bacterial, yeast, and some mammalian expression systems for human FasL production:
Expression System | Protein Folding | Post-translational Modifications | Yield | Scale-up Potential |
---|---|---|---|---|
Baculovirus/Sf9 | Typically soluble and properly folded | Many eukaryotic PTMs, different glycosylation | Medium-high | Excellent |
Bacterial (E. coli) | Often forms inclusion bodies | Minimal to none | High | Excellent |
Yeast | Variable, can be problematic | Some PTMs, hyperglycosylation | Medium | Good |
Mammalian | Excellent | Human-like, complete | Low-medium | Challenging |
The evidence indicates that FasL expressed in Sf9 cells retains functional activity, as demonstrated by its ability to specifically induce apoptosis in T98G cells . The selection between these systems should be based on specific requirements for protein authenticity, yield, and intended applications.
For optimal FasL expression in Sf9 cells, several parameters require careful optimization. While specific conditions for FasL are not detailed in the search results, insights can be drawn from related protein expression studies. For instance, in the CXCR4 expression study, researchers infected Sf9 cells at a density of 3.0 × 10^6 cells/ml with a 1:100 dilution of high-titer baculovirus stocks and cultured them for 48 hours before membrane preparation .
Recommended infection parameters include:
Cell density at infection: 1.5-2.0 × 10^6 cells/ml (balances cell density with nutrient availability)
Multiplicity of infection (MOI): 2-10 for primary infections
Harvest time: 48-72 hours post-infection (48 hours was used in study )
Culture temperature: 27-28°C as mentioned in the CXCR4 expression protocol
Culture media: SF 900 II medium supplemented with 5% fetal calf serum
Cell culture typically occurs in Erlenmeyer flasks under rotation at 125 rpm, with cells maintained at densities between 0.5-6.0 × 10^6 cells/ml . These parameters should be systematically optimized for each specific protein construct.
Post-translational modifications (PTMs) of human FasL expressed in Sf9 cells significantly impact its functional activity. As a type II membrane protein that can be proteolytically cleaved to release a soluble form, several PTM considerations are critical:
Glycosylation differences: Sf9 cells perform simpler high-mannose type N-glycosylation compared to the complex glycosylation in mammalian cells. This difference may affect FasL binding affinity, stability, and immunogenicity.
Proteolytic processing: Proper proteolytic processing is essential for generating soluble FasL. The search results demonstrate that FasL is released into the supernatant of cultured Sf9 cells and verified by Western blotting , confirming that Sf9 cells can process the protein appropriately.
Oligomerization: Active FasL exists as a trimer, and proper assembly is essential for activity. Differences in the cellular environment may affect this oligomerization process.
Disulfide bond formation: Correct disulfide bond formation is crucial for FasL structure. While Sf9 cells generally support disulfide bond formation, the efficiency may differ from mammalian systems.
Comprehensive characterization of FasL expressed in Sf9 cells requires multiple complementary analytical methods:
Protein identification and verification:
Structural characterization:
SDS-PAGE under reducing and non-reducing conditions to assess disulfide bonding
Size exclusion chromatography to determine oligomeric state
Circular dichroism spectroscopy for secondary structure analysis
Functional verification:
PTM analysis:
Glycosylation profiling using mass spectrometry or lectin binding assays
Deglycosylation studies to assess impact on activity
Site-specific PTM mapping
These comprehensive approaches ensure that the expressed FasL maintains the correct structural and functional properties necessary for experimental applications. The methods should be selected based on the specific research questions being addressed.
Overcoming poor expression of human FasL in the baculovirus-Sf9 system requires systematic optimization of multiple parameters:
Vector and construct optimization:
Codon optimization for Sf9 cells' preferred codon usage
Testing different signal sequences for secretion
Using strong promoters like polyhedrin (mentioned in study )
Adding purification tags that may enhance expression (histidine tags were successfully used for other proteins in studies and )
Removing problematic sequences (cryptic splice sites, premature termination codons)
Infection optimization:
Optimizing multiplicity of infection (MOI) and cell density at infection
Testing different harvest times (48-72 hours post-infection)
Adjusting temperature, pH, and media composition
Using protease inhibitors to prevent degradation
Cell and virus considerations:
Using different Sf9 cell subclones or alternative insect cell lines
Fresh amplification of baculovirus stocks to ensure viability
Testing different baculovirus backbone vectors
Co-expression strategies:
If these approaches fail to improve expression, alternative systems such as mammalian cell expression might be considered, though the search results indicate successful expression of functional human FasL in Sf9 cells has been achieved .
Detecting residual Sf9 host cell and baculovirus DNA in purified FasL preparations requires sensitive analytical methods. According to search result , this represents a significant analytical challenge in viral vector manufacturing. The most effective methods include:
Quantitative PCR (qPCR):
Targets specific sequences in Sf9 genomic DNA and baculovirus DNA
Can detect very low levels of contaminating DNA (down to a few copies)
Commercial kits have been developed specifically for this purpose and have been purchased by more than 100 different customers with no known issues regarding acceptance by regulatory agencies
Droplet Digital PCR (ddPCR):
Residual DNA-specific assay kits:
For regulatory compliance, it's important to validate these methods for specific production processes and demonstrate consistent clearance of host cell and viral DNA below acceptable limits.
Reliable quantification of apoptotic activity of Sf9-produced human FasL requires robust and sensitive assays:
Flow cytometry-based methods:
Biochemical detection methods:
Caspase activity assays using specific substrates
DNA fragmentation assays
PARP cleavage detection by Western blotting
Cytochrome c release from mitochondria
Cell viability and cytotoxicity assays:
Study specifically confirmed FasL-induced apoptosis by annexin V-fluorescein isothiocyanate staining, which is considered a gold standard for apoptosis detection. For quantitative analysis, dose-response curves should be established to determine EC50 values for batch-to-batch comparisons.
Addressing batch-to-batch variability in FasL activity from Sf9 expression systems requires comprehensive standardization and quality control:
Process standardization:
Establishing master and working cell banks of Sf9 cells
Creating and validating master virus stocks
Standardizing culture media lots and supplements
Implementing consistent protocols for cell maintenance, infection, and harvest
Controlling cell density, viability, and passage number
Critical parameter monitoring:
Analytical characterization:
Functional standardization:
By implementing these strategies, researchers can minimize batch-to-batch variability and ensure consistent FasL activity across production runs.
Calcium signaling assays provide valuable insights into the functional activity of human FasL through several mechanisms:
Mechanistic relationship:
FasL-induced apoptosis often involves calcium flux as a secondary messenger
Elevated intracellular calcium ([Ca2+]i) can activate calcium-dependent endonucleases that contribute to DNA fragmentation during apoptosis
Sustained calcium elevation can trigger mitochondrial permeability transition
Methodological approaches (as described in study ):
Fluorescent Ca2+ indicators like Fluo-3/AM can be used to monitor [Ca2+]i changes
Laser scanning confocal microscopy to record fluorescence at intervals (every 6 seconds for >400 seconds)
Analysis of dynamic [Ca2+]i changes, including transient elevation followed by recovery or sustained elevation
Correlation with apoptotic events:
Sensitivity and early detection:
Calcium flux often occurs as an early event in apoptosis signaling
Can detect cellular responses before morphological changes become apparent
The methodology described in study for measuring [Ca2+]i in Sf9 cells can be adapted to study FasL-induced calcium responses in target cells, providing additional mechanistic information about the apoptotic pathway activation.
When testing human FasL-induced apoptosis in target cells, including appropriate controls is crucial for experimental validity:
Negative controls:
Vehicle control (buffer/medium used for FasL preparation)
Uninfected Sf9 cell supernatant processed identically to FasL-containing supernatant
Heat-inactivated FasL (to control for non-specific protein effects)
FasL preparation incubated with neutralizing anti-FasL antibodies
Positive controls:
Commercial FasL or FasL from a different expression system with known activity
Alternative apoptosis inducers (e.g., staurosporine)
Graded doses of FasL to establish dose-response relationships
Specificity controls:
Fas-negative cell lines that should be resistant to FasL-induced apoptosis
Fas-blocking antibodies to confirm receptor specificity
Pan-caspase inhibitors (e.g., z-VAD-fmk) to confirm caspase dependency
Procedural controls:
Time-course analysis to capture appropriate apoptotic stages
Multiple apoptosis detection methods (e.g., both annexin V staining and caspase activity)
Study used T98G human glioblastoma cell line as target cells and confirmed apoptosis with annexin V-FITC staining, demonstrating the importance of both appropriate target cell selection and specific apoptosis detection methods.
Human FasL expressed in Sf9 cells provides a valuable tool for studying apoptosis resistance mechanisms in cancer cells, as demonstrated in study with glioblastoma cells:
Mechanistic investigations:
Comparative dose-response studies across cancer cell lines with different apoptotic sensitivities
Time-course analysis to identify early versus late blocks in the apoptotic pathway
Combination treatments with sensitizing agents to overcome resistance
Investigation of downstream signaling events using phospho-specific antibodies
Genetic manipulation approaches:
Gene knockdown/knockout studies to identify resistance factors
Overexpression of candidate resistance genes to confer protection
CRISPR/Cas9 screens to discover novel resistance mechanisms
Clinical relevance investigations:
Correlation of in vitro FasL resistance with clinical outcomes
Testing tumor samples from treatment-resistant versus responsive patients
Development of biomarkers for FasL sensitivity/resistance
Study specifically suggests that "induction of apoptosis by the Fas/FasL system could be a new strategy for the treatment of malignant brain tumors"
Calcium signaling studies:
The Sf9-expressed FasL system offers advantages for these studies including scalable production, consistent preparation, and the ability to produce modified variants to probe specific mechanistic questions about the Fas/FasL system in cancer.
Several modifications to human FasL can enhance its therapeutic potential when expressed in Sf9 cells:
Structural modifications:
Creation of stable FasL trimers through leucine zipper or isoleucine zipper fusion
Development of membrane-anchored versus soluble forms to modulate activity
Engineering of point mutations to enhance receptor binding affinity
Domain swapping with related TNF family members for altered functionality
Targeting modifications:
Fusion to antibody fragments (scFv) for cancer cell targeting
Addition of cell-penetrating peptides for intracellular delivery
Incorporation of tumor-homing peptides for improved tumor localization
Creation of bispecific molecules targeting both Fas and tumor-specific antigens
Pharmacokinetic enhancements:
PEGylation or fusion to albumin-binding domains for extended half-life
Incorporation of unnatural amino acids for site-specific conjugation
Engineering of glycosylation sites to optimize serum stability
Safety and control features:
Development of conditional activation systems
Addition of suicide switches or controllable systems
Engineering of variants with reduced off-target toxicity
While study demonstrates the potential of the Fas/FasL system for treating malignant brain tumors, these modifications could further enhance specificity, potency, and safety for therapeutic applications.
Co-expression of other human proteins with FasL in Sf9 cells can significantly impact its functional properties through various mechanisms:
Protein-protein interactions:
Co-expression with natural binding partners to modulate activity
Co-expression with chaperones to improve folding and solubility
Study of FasL in complex with other death-inducing signaling complex (DISC) components
Investigation of regulatory proteins that modify FasL processing or signaling
Technical and methodological impacts:
Signaling pathway reconstitution:
Production and processing effects:
Co-expression with proteases that process FasL
Co-expression with glycosylation-modifying enzymes
Competition for cellular resources potentially affecting yields
Studies and demonstrate successful co-expression of multiple proteins in Sf9 cells, suggesting that similar approaches could be applied to FasL to study its interactions with other proteins in the apoptotic pathway or to improve its production and functional properties.
Scaling up FasL production in Sf9 cells for larger research studies requires attention to several critical factors:
Cell culture scale-up considerations:
Infection strategy optimization:
Process monitoring and control:
Online monitoring of critical parameters (pH, dissolved oxygen, temperature)
Feed strategies for nutrient supplementation
Consistent harvest criteria based on viability and time post-infection
Implementation of scale-appropriate mixing strategies
Downstream processing considerations:
Development of scalable purification methods
Incorporation of filtration steps to remove cell debris and baculovirus
Chromatography process optimization
Implementation of viral inactivation and removal steps
Quality assurance:
With careful attention to these factors, FasL production can be successfully scaled up while maintaining consistent quality and functional activity for larger research studies.
The sFas receptor, also known as the soluble Fas receptor, is a member of the tumor necrosis factor receptor superfamily (TNFRSF6). It is also referred to by several other names, including CD95, Apo-1 antigen, and apoptosis-mediating surface antigen Fas . The sFas receptor plays a crucial role in the regulation of programmed cell death (apoptosis), which is essential for maintaining cellular homeostasis and immune system function.
The human recombinant sFas receptor produced in Sf9 cells is a single glycosylated polypeptide chain containing 156 amino acids (26-173 aa) with a molecular mass of approximately 17.7 kDa . The receptor is fused to an 8 amino acid His-Tag at the C-terminus, which facilitates its purification using proprietary chromatographic techniques . The recombinant protein is produced using the baculovirus expression system in Sf9 insect cells, which allows for high-yield production and proper post-translational modifications.
The sFas receptor is involved in the extrinsic pathway of apoptosis. It binds to its cognate ligand, FasL (Fas ligand), which is expressed on the surface of activated T cells, natural killer (NK) cells, and dendritic cells . Upon binding to FasL, the sFas receptor undergoes a conformational change that triggers the formation of the death-inducing signaling complex (DISC). This complex subsequently activates caspases, which are proteolytic enzymes that execute the apoptotic program by cleaving various cellular substrates .
The recombinant sFas receptor has several applications in research and therapeutic development. It is used to study the mechanisms of apoptosis and to investigate the role of Fas/FasL interactions in various diseases, including cancer, autoimmune disorders, and viral infections . Additionally, the sFas receptor is utilized in drug screening assays to identify potential therapeutic agents that can modulate apoptosis pathways.
The sFas receptor is supplied as a sterile filtered colorless solution containing 10% glycerol and phosphate-buffered saline (pH 7.4) . For short-term storage, it can be kept at 4°C if used within 2-4 weeks. For long-term storage, it is recommended to store the protein at -20°C with the addition of a carrier protein (0.1% HSA or BSA) to prevent degradation . It is important to avoid multiple freeze-thaw cycles to maintain the protein’s stability and activity.