PCOLCE exhibits dual functionality in ECM regulation:
Procollagen Processing Enhancement: Binds type I procollagen C-terminus, increasing collagen C-proteinase activity by 20-fold
Metalloproteinase Regulation: The C-terminal processed fragment (CT-PCPE) inhibits matrix metalloproteinases
Recent studies using Sf9-produced PCOLCE reveal critical oncogenic roles:
The Sf9 expression system preserves native glycosylation patterns critical for function, particularly the N29 glycosylation site essential for protein stability and secretion .
When working with PCOLCE Human, Sf9:
Activity Assays: Use 1-10 μg/ml concentrations for procollagen processing experiments
Detection: Commercial ELISA kits achieve 93-141% recovery in biological samples
Storage: Maintain at -20°C in aliquots with carrier protein to prevent aggregation
Emerging diagnostic applications leverage PCOLCE's disease associations:
Recombinant PCOLCE, expressed in Sf9 insect cells, is a single, glycosylated polypeptide chain with a molecular weight of 46.6 kDa. The protein consists of 433 amino acids (residues 26-449) and, on SDS-PAGE, exhibits an apparent molecular size of approximately 40-57 kDa.
The recombinant PCOLCE is engineered with a 9 amino acid His tag at the C-terminus to facilitate purification, which is carried out using proprietary chromatographic techniques.
The provided PCOLCE protein solution has a concentration of 0.25 mg/ml and is formulated in a buffer comprising 20mM Tris-HCl (pH 8.0), 10% glycerol, and 0.1M NaCl.
The purity of the PCOLCE protein is determined to be greater than 95.0% using SDS-PAGE analysis.
Procollagen C-endopeptidase enhancer 1, Procollagen COOH-terminal proteinase enhancer 1, PCPE-1, Procollagen C-proteinase enhancer 1, Type 1 procollagen C-proteinase enhancer protein, Type I procollagen COOH-terminal proteinase enhancer, PCOLCE, PCPE1, Procollagen C-Endopeptidase Enhancer, Procollagen C-Proteinase, Enhancer 1, Procollagen, Type 1, COOH-Terminal Proteinase Enhancer, Type I Procollagen COOH-Terminal Proteinase Enhancer, Type 1 Procollagen C-Proteinase Enhancer Protein, Procollagen COOH-Terminal Proteinase Enhancer 1.
Sf9, Insect cells.
ADPQTPNYTR PVFLCGGDVK GESGYVASEG FPNLYPPNKE CIWTITVPEG QTVSLSFRVF DLELHPACRY DALEVFAGSG TSGQRLGRFC GTFRPAPLVA PGNQVTLRMT TDEGTGGRGF LLWYSGRATS GTEHQFCGGR LEKAQGTLTT PNWPESDYPP GISCSWHIIA PPDQVIALTF EKFDLEPDTY CRYDSVSVFN GAVSDDSRRL GKFCGDAVPG SISSEGNELL VQFVSDLSVT ADGFSASYKT LPRGTAKEGQ GPGPKRGTEP KVKLPPKSQP PEKTEESPSA PDAPTCPKQC RRTGTLQSNF CASSLVVTAT VKSMVREPGE GLAVTVSLIG AYKTGGLDLP SPPTGASLKF YVPCKQCPPM KKGVSYLLMG QVEENRGPVL PPESFVVLHR PNQDQILTNL SKRKCPSQPV RAAASQDHHH HHH.
PCOLCE (Procollagen C-Endopeptidase Enhancer) is a glycoprotein that enhances the activity of procollagen C-proteinases, which are essential for cleaving the C-terminal propeptide during collagen maturation. PCOLCE significantly increases the catalytic efficiency of these enzymes, thereby facilitating proper collagen fibril formation and extracellular matrix organization. Research demonstrates that PCOLCE is required for efficient procollagen processing, with studies in PCOLCE-null models showing impaired collagen deposition and altered tissue mechanical properties . The protein contains CUB domains that interact with the C-terminal propeptide of procollagens and a netrin-like domain that may contribute to extracellular matrix organization.
While both PCOLCE1 and PCOLCE2 enhance procollagen C-proteinase activity, they exhibit distinct tissue distribution patterns and potentially different functional specificities. PCOLCE1 shows broader tissue expression, while PCOLCE2 demonstrates more tissue-specific expression patterns. In cardiac tissue, for example, PCOLCE2 plays a crucial role in pressure overload-induced collagen deposition, with PCOLCE2-null mice showing significantly reduced insoluble collagen accumulation compared to wild-type mice under transverse aortic constriction (TAC) . Additionally, while both proteins enhance procollagen processing, PCOLCE2 has been implicated in HDL metabolism and reverse cholesterol transport, suggesting divergent moonlighting functions beyond collagen processing . This functional diversity should be considered when designing experiments focused on specific PCOLCE isoforms.
Optimizing human PCOLCE expression in Sf9 cells requires careful attention to several factors. Since PCOLCE contains disulfide bonds and undergoes glycosylation, the expression system must support proper post-translational modifications. For optimal expression, consider: (1) Codon optimization of the human PCOLCE sequence for insect cell expression; (2) Selection of appropriate signal peptides for secretion; (3) Timing of harvest, as extended expression periods may increase yield but risk protein degradation; (4) Cell density and infection multiplicity (MOI) optimization; and (5) Culture conditions including temperature (27-28°C is typically optimal) and media composition. Based on experimental data comparing PCOLCE expression systems, Sf9 cells can yield functional protein when these parameters are properly optimized, though researchers should verify proper folding through activity assays measuring enhancement of procollagen C-proteinase activity.
Verifying proper post-translational modifications of human PCOLCE expressed in Sf9 cells is essential for ensuring protein functionality. Implement a multi-faceted approach including: (1) SDS-PAGE migration pattern analysis, comparing with mammalian-expressed PCOLCE (proper glycosylation affects apparent molecular weight); (2) Mass spectrometry to identify specific post-translational modifications; (3) Glycosidase digestion experiments to characterize N-linked glycans; (4) Activity assays measuring enhancement of procollagen C-proteinase activity compared to mammalian-expressed standards; and (5) Western blot analysis using conformation-specific antibodies. Several studies have shown that while insect cells perform many mammalian-like post-translational modifications, differences in glycosylation patterns can affect protein function, necessitating careful validation of Sf9-expressed PCOLCE against mammalian standards.
Baculovirus expression vector systems (BEVS) are predominantly used for recombinant protein production in Sf9 cells, with several options offering distinct advantages for human PCOLCE expression. Gateway-compatible vectors have demonstrated particular utility, allowing systematic cloning by site-specific recombination . When expressing human PCOLCE, consider vectors containing: (1) Strong promoters like the polyhedrin or p10 promoter for high expression levels; (2) Secretion signal sequences (either native PCOLCE signal or optimized sequences like honeybee melittin); (3) Purification tags positioned to avoid interfering with PCOLCE functional domains (C-terminal tags are generally preferred); and (4) Protease cleavage sites for tag removal. For comparative studies requiring both N- and C-terminal fusion proteins, designing constructs without native stop codons provides flexibility for different experimental approaches . Each vector system offers trade-offs between expression levels, ease of purification, and maintenance of native protein characteristics.
Designing experiments to investigate PCOLCE's role in collagen processing requires a multi-level approach integrating molecular, cellular, and physiological techniques. Based on published methodologies, effective experimental designs include: (1) Comparing wild-type and PCOLCE-null genotypes in disease models, as exemplified in TAC (transverse aortic constriction) studies of pressure-overloaded myocardium ; (2) Quantifying both soluble and insoluble collagen fractions using hydroxyproline analysis to assess collagen processing efficiency; (3) Measuring tissue mechanical properties, like passive stiffness in myocardial tissues, to correlate molecular changes with functional outcomes; (4) Employing immunoblotting to track procollagen processing intermediates (procollagen α1(I), pC collagen α1(I), and fully processed collagen α1(I)) ; and (5) Supplementing in vivo studies with primary cell cultures (e.g., fibroblasts) from wild-type and PCOLCE-null animals to isolate cellular mechanisms. As demonstrated in published research, this comprehensive approach enables identification of PCOLCE's specific contributions to pathological collagen deposition and tissue remodeling.
Several well-validated assays can effectively measure PCOLCE enhancement of procollagen C-proteinase activity, each with distinct advantages. The most robust approaches include: (1) In vitro reconstituted systems using purified components (procollagen substrate, BMP1/tolloid-like proteinases, and PCOLCE) with SDS-PAGE analysis of processing products; (2) Fluorogenic peptide substrates containing BMP1 cleavage sites, allowing real-time kinetic measurements; (3) Primary fibroblast cultures from wild-type and PCOLCE-null sources with immunoblot monitoring of procollagen processing intermediates ; (4) Pulse-chase experiments tracking the conversion of radiolabeled procollagen to mature collagen; and (5) Mass spectrometry-based approaches for detailed quantification of processing products. When designing these assays, researchers must account for the substrate specificity of PCOLCE (which may differ between PCOLCE1 and PCOLCE2) and the potential influence of other extracellular matrix components. Control experiments should include concentration gradients of PCOLCE to establish dose-dependent enhancement and heat-denatured PCOLCE to confirm specificity.
Investigating PCOLCE's role in reverse cholesterol transport (RCT) requires specialized techniques that bridge collagen biology and lipid metabolism research. Based on recent findings linking PCOLCE2 to HDL metabolism , effective experimental approaches include: (1) Macrophage-to-feces RCT assays using radiolabeled cholesterol in PCOLCE-deficient mouse models compared to controls ; (2) HDL particle analysis by nuclear magnetic resonance (NMR) spectroscopy and gradient gel electrophoresis to assess HDL size and subpopulation distribution; (3) In vitro cholesterol efflux capacity assays using apoB-depleted plasma from PCOLCE-null and wild-type animals; (4) Investigation of PCOLCE2 interactions with key RCT proteins like SR-BI through co-immunoprecipitation and surface plasmon resonance; and (5) Hepatic-specific PCOLCE knockout models to distinguish liver-specific effects from systemic impacts. Research has shown that PCOLCE2-deficient mice exhibit dysfunction in HDL-mediated cholesterol efflux despite higher SR-BI protein levels in the liver , suggesting complex regulatory mechanisms that require carefully designed experiments to elucidate.
Purifying human PCOLCE from Sf9 culture supernatants requires strategies that maintain protein activity while achieving high purity. The most effective purification protocol involves: (1) Initial clarification of culture supernatant by centrifugation (10,000×g, 15 minutes) followed by filtration through 0.45μm membranes; (2) Affinity chromatography as the capture step, typically using nickel-NTA for His-tagged constructs or anti-FLAG resin for FLAG-tagged PCOLCE; (3) Ion exchange chromatography (preferably cation exchange at pH 6.5-7.0) to separate PCOLCE from contaminating proteins; (4) Size exclusion chromatography as a final polishing step to remove aggregates and achieve >95% purity; (5) Buffer optimization during purification and storage to maintain stability (typically 50mM HEPES pH 7.4, 150mM NaCl, 5% glycerol). For applications requiring tag removal, include a proteolytic cleavage step after the initial affinity purification, followed by reverse affinity chromatography. Purified PCOLCE should be assessed for activity using procollagen C-proteinase enhancement assays to confirm that functionality is maintained throughout the purification process.
Accurate quantification of PCOLCE protein levels in biological samples requires carefully validated techniques that account for the protein's characteristics and tissue context. The most reliable approaches include: (1) Quantitative Western blotting using recombinant PCOLCE standards of known concentration for calibration curves; (2) Enzyme-linked immunosorbent assays (ELISAs) with validated antibody pairs specific to human PCOLCE; (3) Selected reaction monitoring (SRM) or parallel reaction monitoring (PRM) mass spectrometry using stable isotope-labeled peptide standards; (4) Immunohistochemistry with digital image analysis for spatial distribution assessment in tissue sections; and (5) Proximity ligation assays for detecting PCOLCE interactions with binding partners in situ. For extracting PCOLCE from tissues, optimized protocols typically employ sequential extraction methods to distinguish between soluble and matrix-bound fractions. When analyzing cardiac tissues, studies have shown that two-fold increases in PCOLCE2 occur in pressure-overloaded myocardium , establishing a benchmark for expected fold changes in pathological conditions.
Silencing PCOLCE expression in cell culture models requires techniques optimized for sustained knockdown without off-target effects. Based on experimental approaches in the literature, the most effective strategies include: (1) CRISPR/Cas9 gene editing for complete knockout in appropriate cell lines, with multiple guide RNAs targeting conserved exons of PCOLCE; (2) Short hairpin RNA (shRNA) delivered via lentiviral vectors for stable long-term knockdown in primary cells like fibroblasts; (3) Small interfering RNA (siRNA) transfection for transient knockdown, with carefully validated sequences to minimize off-target effects; (4) Antisense oligonucleotides (ASOs) designed to target PCOLCE mRNA with high specificity; and (5) Inducible knockdown systems for temporal control of PCOLCE suppression. For all approaches, validation of knockdown efficiency through both mRNA quantification (qRT-PCR) and protein assessment (Western blot) is essential. Functional readouts should include procollagen processing analysis by immunoblotting for procollagen intermediates, similar to studies comparing wild-type and PCOLCE-null cardiac fibroblasts .
Reconciling contradictory findings about PCOLCE across disease models requires systematic analysis of experimental variables and biological contexts. Key approaches include: (1) Distinguishing between PCOLCE1 and PCOLCE2 effects, as these paralogs may have distinct functions in different tissues and disease states; (2) Considering tissue-specific microenvironments that might influence PCOLCE activity through interaction with other extracellular matrix components; (3) Examining temporal aspects of disease progression, as PCOLCE's role may vary between acute and chronic phases; (4) Analyzing differences in experimental models, including species differences, knockout strategies, and physiological stressors; and (5) Evaluating potential moonlighting functions of PCOLCE proteins beyond collagen processing, such as PCOLCE2's role in HDL metabolism . For example, while PCOLCE2 deficiency reduces cardiac fibrosis in pressure overload models , it also dysregulates HDL metabolism , potentially yielding contradictory outcomes in cardiovascular disease models depending on which mechanism predominates in specific contexts.
PCOLCE research presents several technical challenges that can lead to misleading results if not properly addressed. Common pitfalls and their solutions include: (1) Antibody cross-reactivity between PCOLCE1 and PCOLCE2 – resolve by using isoform-specific antibodies validated against knockout controls or recombinant proteins; (2) Variable extraction efficiency from tissues with high collagen content – implement sequential extraction protocols with increasingly stringent buffers; (3) Overlooking PCOLCE interactions with other extracellular matrix components – include relevant matrix proteins in in vitro assays to mimic physiological conditions; (4) Misinterpreting phenotypes in knockout models due to compensatory mechanisms – utilize inducible or tissue-specific knockouts and confirm with acute knockdown approaches; and (5) Confounding effects of tags on recombinant PCOLCE function – validate recombinant protein activity against native protein and consider tag-free purification strategies. Published studies demonstrate the importance of these considerations, as seen in cardiovascular research where comprehensive analysis including hydroxyproline quantification, collagen volume fraction measurement, and mechanical testing was necessary to fully characterize PCOLCE2's role .
Expression System | Yield | Post-translational Modifications | Functional Activity | Best Applications |
---|---|---|---|---|
Sf9 Insect Cells | High | Basic glycosylation, correct disulfide bonds | Good | Structural studies, protein-protein interaction assays |
HEK293 Mammalian | Moderate | Complete mammalian glycosylation pattern | Excellent | Functional assays, in vivo studies |
E. coli | Very High | No glycosylation, requires refolding | Variable | Binding domain studies, antibody production |
Cell-free Systems | Low | None | Limited | Rapid screening, domain mapping |
Parameter | Control WT | PCOLCE2-null | WT with TAC | PCOLCE2-null with TAC |
---|---|---|---|---|
LV mass (mg) | 84 ± 9 | 115 ± 11 | 175 ± 12* | 176 ± 12* |
LV/BW (mg/g) | 2.9 ± 0.1 | 3.1 ± 0.1 | 6.3 ± 0.4* | 5.4 ± 0.4* |
Wall thickness (mm) | 0.8 ± 0.1 | 0.9 ± 0.1 | 1.1 ± 0.1* | 1.1 ± 0.1* |
Collagen Volume Fraction | Baseline | Baseline | ~6-fold increase* | ~2-fold increase*† |
Procollagen C-Endopeptidase Enhancer (PCOLCE) is a glycoprotein that plays a crucial role in the maturation of collagen, a primary structural protein in the extracellular matrix of various tissues. The human recombinant form of PCOLCE, produced in Sf9 insect cells, is utilized in research and therapeutic applications to study and enhance collagen processing.
The PCOLCE gene is located on chromosome 7q22.1 and encodes a protein that binds to the C-terminal propeptide of type I procollagen . This binding enhances the activity of procollagen C-proteinase, an enzyme responsible for cleaving the C-terminal propeptides from procollagen molecules, thereby facilitating the formation of mature collagen fibrils .
PCOLCE enhances the enzymatic cleavage of type I procollagen by procollagen C-proteinase. This process is essential for the proper assembly and stabilization of collagen fibrils, which are critical for the structural integrity of connective tissues . The C-terminal processed part of PCOLCE, known as CT-PCPE, may also exhibit metalloproteinase inhibitory activity, further regulating extracellular matrix remodeling .
The recombinant form of PCOLCE is produced using the baculovirus expression system in Sf9 insect cells. This system allows for high-level expression and proper post-translational modifications of the protein, making it suitable for biochemical and structural studies . The human recombinant PCOLCE produced in Sf9 cells retains its functional properties, enabling researchers to investigate its role in collagen maturation and related pathologies.
PCOLCE has significant implications in both clinical and research settings. Its role in collagen maturation makes it a potential target for therapeutic interventions in diseases characterized by abnormal collagen deposition, such as fibrosis and certain connective tissue disorders . Additionally, recombinant PCOLCE is used in various assays to study collagen processing and to develop inhibitors that could modulate its activity for therapeutic purposes .
Mutations or dysregulation of the PCOLCE gene have been linked to several disorders, including Pierpont Syndrome and nonsyndromic sensorineural deafness . Understanding the function and regulation of PCOLCE can provide insights into the pathogenesis of these conditions and aid in the development of targeted therapies.