PROCR Human, Sf9 refers to a recombinant human protein C receptor (PROCR) produced in Spodoptera frugiperda Sf9 insect cells via baculovirus expression systems. This glycosylated type I transmembrane protein plays critical roles in blood coagulation, thrombosis regulation, and immune responses. Its expression in Sf9 cells enables high-yield production of functional PROCR for research and therapeutic applications .
PROCR Human, Sf9 is a single glycosylated polypeptide chain containing 435 amino acids (residues 18–210) with a molecular mass of 49.3 kDa. It includes an N-terminal extracellular domain, a transmembrane helix, and a cytoplasmic tail. Key features include:
The Sf9-produced variant is engineered with a 242-amino acid hIgG-His tag at the C-terminus for purification and stability. Its glycosylation pattern mimics native human PROCR, ensuring proper receptor function and ligand binding .
PROCR enhances the activation of protein C, a serine protease critical for anticoagulant pathways. Key roles include:
Thrombosis Regulation:
Immune and Pathogen Interactions:
Reproductive Health:
PROCR Human, Sf9 is employed in diverse studies:
Thrombosis Risk Assessment:
Malaria Therapeutics:
Protein C Receptor, CD201, APC Receptor, EPCR, Centrocyclin, CCD41, CCCA.
Sf9, Baculovirus cells.
ADPSQDASDG LQRLHMLQIS YFRDPYHVWY QGNASLGGHL THVLEGPDTN TTIIQLQPLQ EPESWARTQS GLQSYLLQFH GLVRLVHQER TLAFPLTIRC FLGCELPPEG SRAHVFFEVA VNGSSFVSFR PERALWQADT QVTSGVVTFT LQQLNAYNRT RYELREFLED TCVQYVQKHI SAENTKGSQT SRSYTSLEPK SCDKTHTCPP CPAPELLGGP SVFLFPPKPK DTLMISRTPE VTCVVVDVSH EDPEVKFNWY VDGVEVHNAK TKPREEQYNS TYRVVSVLTV LHQDWLNGKE YKCKVSNKAL PAPIEKTISK AKGQPREPQV YTLPPSRDEL TKNQVSLTCL VKGFYPSDIA VEWESNGQPE NNYKTTPPVL DSDGSFFLYS KLTVDKSRWQ QGNVFSCSVM HEALHNHYTQ KSLSLSPGKH HHHHH.
Sf9 cells derived from Spodoptera frugiperda are widely used in baculovirus expression vector systems due to their robust growth characteristics and efficient protein processing capabilities. These cells have an average diameter of approximately 16 μm and demonstrate high proliferation rates . They can be efficiently cultured in serum-free medium and scaled up in suspension culture, making them ideal for recombinant protein production.
When working with human PROCR expression, Sf9 cells provide several advantages, including eukaryotic post-translational modifications, proper protein folding machinery, and the ability to produce complex membrane proteins like PROCR. The baculovirus system in Sf9 cells typically yields higher expression levels compared to mammalian systems, while maintaining many of the critical post-translational modifications required for functional human PROCR .
For optimal maintenance of Sf9 cells expressing human PROCR:
Culture conditions: Maintain cells at 27-28°C without CO₂ in appropriate insect cell medium. Cells can be grown in adherent culture for maintenance and suspension culture for protein production.
Passage frequency: Subculture cells when they reach 80-90% confluence or at a density of 2-3 × 10⁶ cells/mL in suspension culture. For consistent results, maintain cells in log phase growth.
Cell quality monitoring: Regularly assess cell viability (>95% ideal), doubling time (approximately 18-24 hours), and morphology. The average diameter of healthy Sf9 cells is around 16-18 μm, though transgenic lines may show slight variations in size .
Cryopreservation: Create master and working cell banks at early passages in medium containing 10% DMSO, storing in liquid nitrogen for long-term maintenance of cell line characteristics.
Contamination prevention: Implement strict aseptic technique and regular testing for mycoplasma and other potential contaminants that could affect expression quality.
It's important to note that Sf9 cells constitutively produce reverse transcriptase activity associated with endogenous retroviral-like particles, which should be considered when designing purification strategies for PROCR .
The optimal vector design for human PROCR expression in Sf9 cells should include:
Strong promoters: Very late polyhedrin (polh) or p10 promoters drive high-level expression, though for complex membrane proteins like PROCR, early promoters may sometimes yield better folding. Recent research has shown that combining an hr3 enhancer element with strong viral promoters can significantly increase expression efficiency .
Signal sequences: Include native PROCR signal sequence or replace with an insect-optimized signal sequence (e.g., melittin or gp64) to improve secretion or membrane targeting depending on your experimental goals.
Affinity tags: Incorporate purification tags (His₆, FLAG, etc.) positioned to minimize interference with PROCR function. C-terminal tags are often preferred as they typically cause less disruption to protein trafficking.
Kozak sequence: Optimize the translation initiation context for insect cells (AAAATG rather than mammalian ACCATG).
Codon optimization: While not always necessary, adapting the human PROCR sequence to Sf9 preferred codon usage can sometimes improve expression levels.
Cleavage sites: Include TEV or PreScission protease sites if tag removal is required post-purification.
Polyadenylation signal: Ensure proper mRNA processing with an SV40 or other appropriate polyadenylation signal.
For membrane proteins like PROCR, consider adding a C-terminal GFP tag for easy monitoring of expression and proper folding. Fluorescent tags can significantly streamline troubleshooting by enabling rapid visual assessment of expression efficiency and protein localization .
Optimizing the multiplicity of infection (MOI) is critical for achieving maximum PROCR yield while maintaining protein quality:
Empirical determination: Test a range of MOIs (typically 0.1-10) to determine optimal conditions. For PROCR as a membrane protein, lower MOIs (0.5-2) often yield better results by providing more time for proper folding and post-translational processing.
Infection kinetics monitoring: Track the progression of infection using:
Time-course analysis: For PROCR, harvest timing is critical. Conduct small-scale time-course experiments (typically 48, 72, 96 hours post-infection) to determine optimal harvest time based on both yield and quality assessments.
Virus stock quantification: Accurate MOI calculation requires precise virus titers. Modern transgenic cell lines like Sf9-QE enable more rapid virus quantification (within 6 days) compared to traditional plaque assays that may take 10-12 days .
Scale considerations: Note that optimal MOI may differ between small-scale and large-scale production due to differences in cell-to-cell virus transmission dynamics.
For precise experimental reproducibility, virus quantification should be performed using standardized methods. Fluorescence-based quantification using engineered cell lines like Sf9-QE can provide results approximately 4-6 days faster than traditional plaque assay methods while maintaining comparable accuracy .
Human PROCR is a type I transmembrane protein requiring specialized purification approaches to maintain its native structure:
Membrane preparation: Begin with gentle cell lysis using nitrogen cavitation or moderate sonication to preserve membrane structures. Differential centrifugation (10,000× g followed by 100,000× g ultracentrifugation) effectively separates membrane fractions.
Detergent selection: Screen multiple detergents at their critical micelle concentrations:
Initial extraction: DDM (n-Dodecyl β-D-maltoside, 0.5-1%) or LMNG (Lauryl maltose neopentyl glycol, 0.1%)
Purification buffers: Milder detergents like CHAPS (0.5%) or replacement with amphipols for structural studies
Chromatography sequence:
Initial capture: Immobilized metal affinity chromatography (IMAC) if His-tagged
Intermediate: Ion exchange chromatography to remove contaminating endogenous retroviral-like particles that may co-purify based on their reported density (1.08 g/mL)
Polishing: Size exclusion chromatography to isolate monodisperse protein and remove aggregates
Buffer optimization: Include stabilizers like cholesterol hemisuccinate (CHS, 0.1%) and lipids (phosphatidylcholine, 0.01-0.02%) to maintain PROCR in native-like environment.
Quality control checkpoints:
SDS-PAGE with western blotting at each purification stage
Functional binding assays with Protein C to confirm activity
Thermal stability assays (DSF/nanoDSF) to verify proper folding
It's critical to monitor for co-purification of endogenous retroviral-like particles that are constitutively expressed in Sf9 cells. These particles contain reverse transcriptase activity and can be isolated by density gradient ultracentrifugation, peaking at around 1.08 g/mL . Additional purification steps may be necessary to remove these contaminants if they compromise downstream applications.
Comprehensive characterization of post-translational modifications (PTMs) is essential for understanding functional equivalence between native and Sf9-expressed PROCR:
When evaluating PTMs, consider that Sf9 cells may be chemically induced to alter their gene expression profiles, which could potentially impact the PTM machinery. Treatment with agents like 5-iodo-2′-deoxyuridine (IUdR) has been shown to induce a 33-fold higher reverse transcriptase activity in Sf9 cells , suggesting significant changes in cellular biochemistry that might influence protein modification patterns.
When conducting functional studies using PROCR in Sf9-derived membrane preparations, consider these advanced methodological approaches:
Membrane scaffold preparations:
Nanodiscs: Incorporate purified PROCR into MSP1D1 or other appropriate scaffold proteins with defined lipid compositions to maintain native-like membrane environment
Proteoliposomes: Reconstitute PROCR in liposomes containing phosphatidylcholine/phosphatidylserine (70:30) to mimic endothelial cell membranes
Styrene maleic acid lipid particles (SMALPs): Extract PROCR with native lipid environment preserved
Binding kinetics optimization:
Surface plasmon resonance (SPR) with captured PROCR orientated via C-terminal tags
Bio-layer interferometry with controlled density of immobilized PROCR
Fluorescence correlation spectroscopy for solution-phase kinetics in nanodiscs
Signaling reconstitution:
Co-expression with relevant G-proteins or adaptor proteins
Fluorescent calcium indicators for functional coupling assays
BRET/FRET-based interaction assays with labeled signaling partners
Cryo-EM considerations:
Detergent screening specifically for structural stability
GraFix crosslinking to stabilize complexes
Controlled deglycosylation to reduce conformational heterogeneity
Quality controls:
Circular dichroism to verify secondary structure integrity
Microscale thermophoresis to validate ligand interactions
Negative staining EM to confirm homogeneity
When utilizing Sf9-derived membrane preparations, it's important to account for potential contamination with endogenous retroviral-like particles and extracellular vesicles that have been observed in Sf9 culture supernatants . Transmission electron microscopy and cryo-EM analysis of your preparations can help identify these contaminants, which appear as heterogeneous particles with varying sizes and structures .
Creating customized Sf9 cell lines through gene editing offers several advantages for human PROCR expression:
Glycoengineering approaches:
Knockout of genes encoding insect-specific glycosyltransferases
Knock-in of human glycosyltransferases (GalT, SialT) for complex glycan synthesis
CRISPR-Cas9 multiplexing to modify multiple glycosylation pathway components simultaneously
Chaperone enhancement:
Overexpression of relevant folding chaperones (calreticulin, calnexin)
Engineering of stress-response pathways to improve folding of complex proteins
Integration of human-specific chaperones involved in disulfide bond formation
Proteolysis mitigation:
Knockout of problematic endogenous proteases
RNAi-based suppression of protease expression during protein production phase
Integration of protease inhibitors into secretory pathway
Reporter systems integration:
Implementation methodology:
The generation of transgenic Sf9 cell lines can be achieved using the piggyBac transposon system, which has been successfully employed to create lines with enhanced properties for virus quantification . When developing customized cell lines, it's essential to confirm the integration of your genetic modifications by PCR assay of genomic DNA and to verify phenotypic stability over multiple passages (at least 10) to ensure consistent PROCR expression characteristics .
Distinguishing intrinsic PROCR expression problems from Sf9-related factors requires systematic investigation:
Expression level analysis:
Quantitative western blotting normalized to cell number
Flow cytometry for surface expression (if applicable)
RT-qPCR with validated reference genes to assess mRNA levels
Control experiments:
Express a well-characterized control protein in parallel
Test expression in alternative cell lines (High Five, Sf21)
Compare expression with different promoters and signal sequences
Endogenous factor assessment:
Reference gene selection for expression analysis:
Quality assessment methods:
Size exclusion chromatography to evaluate aggregation state
Thermostability assays to assess proper folding
Ligand binding assays to confirm functionality
When analyzing PROCR expression data, it's important to recognize that Sf9 cells constitutively produce reverse transcriptase activity (approximately 10⁵ pU/μL) associated with retroviral-like particles . These particles may interfere with certain assays or co-purify with your protein of interest. Additionally, treatment with chemical inducers commonly used in expression systems can significantly alter the cellular environment, with compounds like IUdR shown to increase RT activity 33-fold .
When faced with discrepancies between native and Sf9-expressed PROCR functional data, implement this systematic resolution framework:
Structural comparison methodology:
Epitope mapping using monoclonal antibody panels
Hydrogen-deuterium exchange mass spectrometry to compare conformational dynamics
Cross-linking mass spectrometry to assess tertiary structure equivalence
Limited proteolysis patterns to evaluate domain accessibility differences
Post-translational modification analysis:
Comparative glycoprofiling between native and recombinant PROCR
Site-directed mutagenesis of key modification sites to assess functional impact
Enzymatic modification in vitro to "humanize" insect-specific PTMs
Membrane environment considerations:
Lipid composition analysis of native vs. Sf9 membranes
Reconstitution experiments in defined lipid environments
Cholesterol dependence assessment through methyl-β-cyclodextrin depletion studies
Binding partner interactions:
Surface plasmon resonance with concentration series to detect affinity shifts
Competitive binding assays with natural ligands
Co-immunoprecipitation studies to identify differentially associated proteins
Experimental design controls:
Side-by-side comparison with PROCR from alternative expression systems
Domain-swap chimeras to isolate regions responsible for functional differences
Concentration-matched assays to eliminate dosage effects
When investigating functional discrepancies, consider that the cellular environment of Sf9 cells may contain retroviral-like particles and extracellular vesicles that could potentially affect protein characteristics . These particles have been observed by transmission electron microscopy and cryo-EM to have diverse sizes and structures , which might contribute to unexpected interactions with your recombinant PROCR.
| Expression System | Yield (mg/L) | Glycosylation Pattern | Functional Activity (%) | Time to Harvest (days) | Major Advantages | Key Limitations |
|---|---|---|---|---|---|---|
| Sf9/Baculovirus | 5-20 | High-mannose, non-complex | 70-85 | 3-4 | High yield, scalable | Non-human glycosylation |
| Stable Sf9 Lines | 2-8 | High-mannose, non-complex | 65-80 | Continuous | Consistent quality | Lower yield than baculovirus |
| Mammalian HEK293 | 1-5 | Complex, sialylated | 90-100 | 5-7 | Human-like PTMs | Higher cost, lower yield |
| CHO Cells | 2-15 | Complex, partially sialylated | 85-95 | 7-14 | Scalable, regulatory acceptance | Longer production time |
| E. coli (refolded) | 50-100 | None | 30-50 | 1-2 | Highest yield, lowest cost | Requires refolding, no PTMs |
| Cell-free Systems | 0.5-2 | None (or controlled addition) | 60-75 | 1-2 days | Rapid prototyping | Low yield, high cost |
Note: Functional activity is measured as percentage relative to native human PROCR isolated from endothelial cells. Yield ranges represent typical values and may vary based on specific constructs and optimization efforts.
Establishing robust high-throughput screening (HTS) platforms using Sf9-expressed PROCR requires addressing several methodological challenges:
Stable cell line development:
Create Sf9 lines with constitutive or inducible PROCR expression
Incorporate reporter systems (calcium flux indicators, β-arrestin recruitment)
Engineer bicistronic constructs linking PROCR activation to fluorescent/luminescent outputs
Implement the piggyBac transposon system for stable integration and expression
Assay miniaturization:
Optimize cell density for 384/1536-well formats (typically 5,000-15,000 cells/well)
Determine minimum incubation times for adequate signal-to-noise ratio
Balance PROCR expression levels for physiological relevance while maintaining detection window
Detection modality selection:
BRET/FRET-based systems for direct compound binding
Calcium mobilization for functional coupling assays
Label-free technologies (impedance, dynamic mass redistribution)
GFP complementation assays for protein-protein interaction modulation
Quality control metrics:
Implement Z′-factor calculations (aim for >0.5 for robust assays)
Establish dose-response relationships with known ligands
Develop counter-screens to eliminate false positives targeting the reporter system
Institute periodic cell line verification to detect genetic drift
Automation compatibility:
Develop cryopreservation protocols for batch preparation of screening cells
Optimize media formulations for extended assay windows
Establish automated image analysis pipelines for phenotypic screens
When developing these systems, it's important to consider that Sf9 cells constitutively produce retroviral-like particles that contain reverse transcriptase activity . These particles might potentially interfere with certain assay readouts, particularly in fluorescence-based systems. Additionally, chemical treatments commonly used in screening assays may alter the expression of endogenous Sf9 cell components, with compounds like IUdR shown to induce significant changes in cellular activity .
Designing rigorous comparative studies between soluble and membrane-bound PROCR requires careful methodological planning:
Construct design considerations:
Generate precisely matched soluble constructs (truncation at residue 210)
Include identical tags and linkers in both variants
Create GPI-anchored variants as intermediate membrane association form
Consider chimeric constructs with alternative transmembrane domains
Expression strategy harmonization:
Express all variants in parallel using identical Sf9 cell stocks
Maintain consistent MOI and harvest times
Process samples through matched purification protocols
Quantify and normalize final preparations using multiple methods (BCA, A280, quantitative western blot)
Functional equivalence assessment:
Compare protein C binding kinetics using surface plasmon resonance
Assess thrombin binding and inhibition activities
Evaluate calcium signaling capabilities in reconstituted systems
Measure PAR1 interaction differences
Structural characterization:
Analyze secondary structure content via circular dichroism
Compare thermal stability profiles using differential scanning fluorimetry
Assess oligomerization state by size exclusion chromatography with multi-angle light scattering
Perform comparative epitope mapping with conformation-sensitive antibodies
Data normalization approach:
Establish molar equivalence rather than mass concentration
Determine active fraction through active site titration where applicable
Account for orientation effects in immobilized assays
Consider detergent or nanodisc effects on apparent activity
When comparing membrane-bound and soluble PROCR variants, it's essential to consider the potential influence of Sf9 cell-derived membrane components, including endogenous retroviral-like particles that may co-purify with membrane preparations . These particles appear as heterogeneous structures with varying sizes under electron microscopy and might impact the biophysical properties of membrane protein preparations.
Protein-C receptor, also known as endothelial protein C receptor (EPCR), is a critical component in the protein C anticoagulant pathway. This receptor plays a significant role in regulating blood coagulation, inflammation, and cell death. The recombinant form of this receptor, expressed in Sf9 cells, has been extensively studied for its therapeutic potential and biological functions.
The Sf9 cell line, derived from the fall armyworm Spodoptera frugiperda, is widely used for the expression of recombinant proteins. The baculovirus expression system in Sf9 cells is particularly advantageous for producing high levels of functional proteins, including G-protein-coupled receptors (GPCRs) and other complex proteins . This system allows for the co-expression of multiple proteins, facilitating the study of protein interactions and functions.
The protein-C receptor is a transmembrane glycoprotein that binds activated protein C (APC). APC is a serine protease with potent anticoagulant and anti-inflammatory properties . The binding of APC to EPCR enhances its anticoagulant activity by promoting the inactivation of factors Va and VIIIa, which are essential for blood clot formation. Additionally, the APC-EPCR complex can activate protease-activated receptor 1 (PAR-1), leading to various cellular responses, including anti-apoptotic and anti-inflammatory effects .
Recombinant EPCR expressed in Sf9 cells has been investigated for its potential therapeutic applications. The ability to produce large quantities of functional EPCR allows for detailed studies on its role in various diseases, including sepsis, thrombosis, and inflammatory disorders. The recombinant receptor can be used to develop novel therapeutic agents that target the protein C pathway, offering new treatment options for patients with coagulation and inflammatory disorders.