GZMH Human, sf9

Granzyme-H Human Recombinant, sf9
Shipped with Ice Packs
In Stock

Description

Production and Purification

The protein is synthesized using baculovirus-infected Sf9 cells, followed by purification via immobilized metal affinity chromatography (IMAC) leveraging its C-terminal hexahistidine tag . This system achieves higher yields (0.25–0.5 mg/mL) compared to mammalian or E. coli systems, while retaining native folding and enzymatic activity .

Biological Functions

GZMH plays dual roles in immunity:

  • Apoptosis induction: Synergizes with perforin to cleave caspases and viral proteins (e.g., HBV HBx), triggering target cell death .

  • Viral defense: Degrades hepatitis B virus (HBV) proteins via Met79_{79} cleavage, a mechanism inhibited by Ac-PTSY-chloromethylketone .

  • Immune regulation: Copy number loss in GZMH correlates with resistance to anti-PD-1 therapy in cancers like nasopharyngeal carcinoma (NPC) .

Therapeutic Applications

  • Targeted inhibitors: Tetrapeptide chloromethylketones (e.g., Ac-PTSY-CMK) selectively block GZMH activity, aiding functional studies .

  • Biomarker potential: GZMH expression levels predict response to immune checkpoint inhibitors in melanoma and NSCLC .

Comparison with Other Recombinant Forms

GZMH Human, sf9 differs from E. coli-derived variants (e.g., ProSpec’s ENZ-855) in:

  • Glycosylation: Sf9-derived GZMH includes mammalian-like glycosylation, enhancing stability and receptor binding .

  • Activity: Baculovirus-expressed GZMH shows higher specific activity (>7,000 pmol/min/µg) compared to non-glycosylated forms .

Product Specs

Introduction
Granzyme H, also referred to as GZMH, is a member of the peptidase S1 family. This enzyme plays a crucial role in the eradication of the Hepatitis B Virus (HBV). GZMH targets and cleaves the HBx protein, a vital component for HBV replication, at the Met(79) position. This action effectively disrupts the virus's ability to multiply. The significance of GZMH in HBV clearance is further emphasized by the fact that inhibiting GZMH prevents both GZNH- and lymphokine-activated killer cell-mediated HBx degradation, ultimately hindering HBV elimination. HBV variants lacking the HBx protein exhibit resistance to GzmH-mediated, as well as lymphokine-activated killer cell-mediated, viral clearance.
Description
Produced in Sf9 Baculovirus cells, GZMH is a single, glycosylated polypeptide chain with a molecular weight of 26.2kDa. It consists of 234 amino acids (19-246 a.a.). On SDS-PAGE, the molecular size will appear between 28-40kDa. The GZMH protein is expressed with a 6 amino acid His tag at the C-Terminus and undergoes purification using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The GZMH protein solution is provided at a concentration of 0.25mg/ml. It is formulated in Phosphate Buffered Saline (pH 7.4) containing 10% glycerol.
Stability
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. Adding a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Repeated freezing and thawing of the product should be avoided.
Purity
The purity of GZMH is determined to be greater than 90% based on SDS-PAGE analysis.
Synonyms
Granzyme H, Granzyme H (Cathepsin G-Like 2, Protein H-CCPX), Cytotoxic T-Lymphocyte Proteinase, Cathepsin G-Like 2, CTSGL2, CCP-X, CSP-C, Cytotoxic T-Lymphocyte-Associated Serine Esterase 1, Cathepsin G-Like 2, Protein H-CCPX, Cytotoxin Serine Protease-C, Cytotoxic Serine Protease C, Protein H-CCPX, EC 3.4.21.79, EC 3.4.21, EC 3.4.21, CTLA1, CGL-2 , CGL2.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence
EEIIGGHEAK PHSRPYMAFV QFLQEKSRKR CGGILVRKDF VLTAAHCQGS SINVTLGAHN IKEQERTQQF IPVKRPIPHP AYNPKNFSND IMLLQLERKA KWTTAVRPLR LPSSKAQVKP GQLCSVAGWG YVSMSTLATT LQEVLLTVQK DCQCERLFHG NYSRATEICV GDPKKTQTGF KGDSGGPLVC KDVAQGILSY GNKKGTPPGV YIKVSHFLPW IKRTMKRLHH HHHH.

Q&A

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

GZMH Human, sf9 is a recombinant Granzyme H protein expressed in Spodoptera frugiperda (Sf9) insect cells using a baculovirus expression system. Structurally, it is a single, glycosylated polypeptide chain containing 234 amino acids (specifically residues 19-246 of the full sequence) with a molecular mass of 26.2 kDa . When analyzed via SDS-PAGE, the protein typically appears at approximately 28-40 kDa due to its glycosylation pattern . The recombinant protein is commonly expressed with a 6-amino acid histidine tag at the C-terminus to facilitate purification, and it is typically purified using proprietary chromatographic techniques to achieve greater than 90% purity .

What is the physiological function of GZMH and why is it important for research?

GZMH plays an essential role in viral clearance mechanisms, particularly for hepatitis B virus (HBV). The protein functions by specifically cleaving the HBx protein at the Met(79) position . This is significant because HBx is required for HBV replication, and its degradation by GZMH contributes to viral clearance. Research has shown that GZMH inhibitors can abolish both GZMH-mediated and lymphokine-activated killer cell-mediated HBx degradation and HBV clearance . Additionally, HBx-deficient HBV demonstrates resistance to GZMH-mediated viral clearance, highlighting the specificity of this interaction . These properties make GZMH an important target for immunological and virological research, particularly in studies focused on understanding host defense mechanisms against viral infections.

What are the optimal storage conditions for GZMH Human, sf9?

For optimal preservation of GZMH Human, sf9 activity, specific storage guidelines should be followed. For short-term use (within 2-4 weeks), the protein can be stored at 4°C . For longer periods, storage at -20°C is recommended . To minimize protein degradation during long-term storage, it is advisable to add a carrier protein such as human serum albumin (HSA) or bovine serum albumin (BSA) at a concentration of 0.1% . This helps to stabilize the protein and prevent adsorption to storage container surfaces. Multiple freeze-thaw cycles should be strictly avoided as they can lead to protein denaturation and loss of activity . The standard formulation of commercially available GZMH protein solution (0.25 mg/ml) contains Phosphate Buffered Saline (pH 7.4) and 10% glycerol , which helps maintain protein stability.

How does the glycosylation pattern of Sf9-expressed GZMH differ from mammalian cell-expressed variants, and what are the implications for research applications?

Sf9 insect cells produce proteins with distinct glycosylation patterns compared to mammalian expression systems, which has significant implications for research applications of GZMH. Proteins expressed in Sf9 cells predominantly contain truncated (paucimannose) and high-mannose glycans, with limited capability to produce complex or hybrid glycans . Unlike mammalian cells, Sf9 cells cannot produce sialylated structures due to low levels of sialic acid . These differences arise because insect cells possess different glycan processing enzymes than mammalian cells.

The glycosylation profile of Sf9-expressed GZMH typically includes:

Glycan TypeTypical StructurePrevalence in Sf9-expressed proteins
Truncated (Paucimannose)Man₃GlcNAc₂Fuc₀₋₁Dominant at most sites
High-mannoseMan₅₋₉GlcNAc₂Present at specific sites
HybridMan₃₋₄GlcNAc₃₋₄Limited (<30%)
ComplexVarious elaborate structuresMinimal (<1%)

What are the molecular mechanisms underlying GZMH-mediated HBx degradation, and how can this be experimentally validated?

GZMH mediates HBx degradation through a specific proteolytic mechanism targeting the Met(79) residue of the HBx protein . This cleavage is critical for disrupting HBV replication, as HBx is essential for the viral life cycle. To experimentally validate this mechanism, researchers can employ several complementary approaches:

  • In vitro cleavage assays: Purified GZMH can be incubated with recombinant HBx protein under physiological conditions (pH 7.4, 37°C). The reaction products can be analyzed by SDS-PAGE and Western blotting to detect specific cleavage fragments. Mass spectrometry can confirm cleavage at the Met(79) position.

  • Site-directed mutagenesis: Creating an HBx variant with mutation at the Met(79) position (e.g., M79A) should render it resistant to GZMH cleavage. Comparing the degradation of wild-type versus mutant HBx in the presence of GZMH would validate the specificity of the cleavage site.

  • Cellular assays: HBV-expressing cell lines can be treated with purified GZMH (potentially delivered via cell-penetrating peptides) or co-cultured with GZMH-expressing immune cells. Monitoring HBx levels via immunoblotting and HBV replication via quantitative PCR would demonstrate the functional significance of GZMH-mediated degradation.

  • Inhibitor studies: GZMH-specific inhibitors can be used to block the enzyme's activity in cellular systems. If HBx degradation and HBV clearance are prevented by these inhibitors, this would further confirm GZMH's mechanistic role .

  • HBx-deficient HBV models: Using HBx-deficient HBV strains in experimental systems should demonstrate resistance to GZMH-mediated viral clearance, as observed in previous studies .

The validation of these mechanisms has significant implications for understanding immune responses against HBV and potentially developing novel therapeutic approaches targeting this pathway.

How does the enzymatic activity of GZMH Human, sf9 compare to endogenous human GZMH, and what controls should be implemented in functional assays?

When comparing Sf9-expressed recombinant GZMH to endogenous human GZMH, several important differences in enzymatic activity must be considered. While the recombinant protein preserves the core catalytic function, differences in post-translational modifications, particularly glycosylation patterns, can impact substrate specificity, kinetics, and regulatory interactions.

For rigorous functional assays comparing these variants, the following controls should be implemented:

Control TypePurposeImplementation
Activity normalizationEnsure comparison of equivalent enzymatic unitsDetermine specific activity using standard substrates; normalize all experiments to activity units rather than protein mass
Substrate spectrum analysisIdentify potential differences in substrate preferenceTest multiple peptide substrates with varying sequences around the cleavage site; calculate kcat/Km ratios for each
pH and ionic strength profilingDetect differences in optimal reaction conditionsPerform activity assays across pH range (5.0-9.0) and varying salt concentrations (50-500 mM NaCl)
Inhibitor sensitivityAssess structural differences in the active siteCompare IC50 values for a panel of serine protease inhibitors
Thermal stability comparisonEvaluate conformational stability differencesMonitor activity retention after pre-incubation at elevated temperatures (37-60°C)
Native cellular extractProvide physiologically relevant contextInclude cytoplasmic extracts from relevant immune cells with appropriate co-factors

To specifically evaluate the HBx cleavage activity, recombinant HBx protein should be used as a substrate in parallel with synthetic peptides spanning the Met(79) region. Time-course experiments monitoring substrate depletion and product formation would provide valuable kinetic parameters for comparison between endogenous and recombinant GZMH variants. Additionally, mass spectrometry analysis of cleavage products can confirm that both enzymes target the same peptide bond, reinforcing functional equivalence despite structural differences.

What are the optimal conditions for expressing GZMH in Sf9 cells, and how can expression yields be maximized?

Optimizing GZMH expression in Sf9 cells requires careful attention to several key parameters that influence protein yield and quality. Based on studies of protein expression in Sf9 cells, the following conditions can be recommended:

  • Cell culture conditions: Maintaining Sf9 cells in logarithmic growth phase (1.5-2.5 × 10^6 cells/ml) prior to infection ensures optimal protein production. The cells should have viability >95% at the time of infection .

  • Multiplicity of infection (MOI): For GZMH expression, an MOI between 2-5 is typically optimal. Lower MOI may result in incomplete infection, while higher MOI can cause premature cell death .

  • Time of harvest: Peak expression for secreted proteins like GZMH typically occurs 72-96 hours post-infection, but this should be determined empirically through time-course experiments.

  • Culture medium: Serum-free formulations specifically designed for Sf9 cells (such as Sf-900™ III SFM) generally yield better results for recombinant protein production.

  • Temperature: Lowering the incubation temperature from the standard 27°C to 24-25°C after infection can sometimes increase yields of properly folded proteins by slowing down expression and allowing more time for folding and post-translational modifications.

From studies on the expression of other proteins in Sf9 cells, key parameters affecting expression can be organized as follows:

ParameterOptimal RangeImpact on Expression
Cell density at infection1.5-2.5 × 10^6 cells/mlHigher densities may deplete nutrients too quickly
MOI2-5Affects infection efficiency and cell viability
Harvest time72-96 hours post-infectionProtein-specific, requires optimization
pH6.2-6.4Affects cell health and protein stability
Dissolved oxygen30-60%Impacts cell metabolism and protein folding
Feed supplementation5-10%Prevents nutrient depletion during expression

For scale-up production, strategic feeding protocols and careful monitoring of culture parameters can significantly improve yields. Adding fresh medium (5-10% of culture volume) 24 hours post-infection has been shown to enhance protein expression in Sf9 cultures .

What purification strategies yield the highest purity and activity for GZMH Human, sf9?

Purification of GZMH Human, sf9 requires a strategic approach to maintain both high purity and enzymatic activity. The typical purification workflow leverages the C-terminal 6×His tag and incorporates multiple chromatographic steps:

  • Initial clarification: Following expression, Sf9 cell culture is typically centrifuged at 10,000-12,000 × g for 30 minutes at 4°C to remove cells and debris. The supernatant containing secreted GZMH is filtered through a 0.45 μm membrane to remove remaining particulates.

  • Immobilized Metal Affinity Chromatography (IMAC): The filtered supernatant is applied to a Ni-NTA or similar resin equilibrated with binding buffer (typically 50 mM sodium phosphate, 300 mM NaCl, 10 mM imidazole, pH 7.4). After washing with increasingly stringent buffers (20-50 mM imidazole), GZMH is eluted with 250-300 mM imidazole .

  • Size Exclusion Chromatography (SEC): To remove aggregates and further increase purity, IMAC-purified GZMH is subjected to gel filtration using a suitable resin (e.g., Superdex 75) in a physiological buffer (PBS, pH 7.4).

  • Ion Exchange Chromatography (optional): For exceptionally high purity requirements, an ion exchange step can be included. Given GZMH's theoretical pI of approximately 9.5, cation exchange chromatography would be appropriate.

For optimal recovery of active enzyme, the following considerations are critical:

Purification ConsiderationRecommendationRationale
TemperatureMaintain 4°C throughoutMinimizes proteolytic degradation
Protease inhibitorsInclude in initial lysis bufferPrevents degradation of target protein
Buffer compositionInclude 10% glycerol in all buffersEnhances protein stability
Elution conditionsUse step gradient for IMAC elutionConcentrates protein and reduces contamination
Final formulationPBS (pH 7.4) with 10% glycerolPreserves enzymatic activity during storage
Concentration methodCentrifugal concentrators with 10 kDa cutoffPrevents protein loss while removing small contaminants

Using this optimized purification strategy, GZMH Human, sf9 with >90% purity can be reliably obtained , with yields typically in the range of 5-10 mg per liter of Sf9 culture. Activity assays using chromogenic or fluorogenic peptide substrates should be performed at each purification step to track recovery of enzymatic activity.

How can researchers assess the enzymatic activity and specificity of purified GZMH Human, sf9?

Assessing the enzymatic activity and specificity of purified GZMH Human, sf9 requires a multi-faceted approach that evaluates both general proteolytic function and specific substrate cleavage. The following methodological approaches are recommended:

  • Chromogenic/Fluorogenic peptide assays: Synthetic peptide substrates containing GZMH's preferred cleavage motif coupled to a chromogenic (p-nitroaniline) or fluorogenic (AMC, AFC) leaving group can be used to measure basic enzymatic activity. A typical reaction would include:

    • 1-10 μg purified GZMH

    • 50-200 μM peptide substrate

    • Reaction buffer: 50 mM Tris-HCl, pH 7.5, 100 mM NaCl, 0.01% Tween-20

    • Incubation at 37°C with continuous monitoring of absorbance/fluorescence

  • HBx cleavage assay: To assess the physiologically relevant activity:

    • Incubate purified GZMH (1-5 μg) with recombinant HBx protein (1-2 μg) in physiological buffer

    • Sample aliquots at various time points (0, 15, 30, 60, 120 minutes)

    • Analyze by SDS-PAGE and Western blotting using anti-HBx antibodies

    • Confirm cleavage products by mass spectrometry to verify the Met(79) cleavage site

  • Kinetic parameter determination: For quantitative comparison between different GZMH preparations:

    • Perform substrate concentration series (10-500 μM)

    • Plot initial velocity vs. substrate concentration

    • Determine Km, Vmax, and kcat using appropriate enzyme kinetics software

    • Compare parameters to reference standards or literature values

  • Inhibitor profiling: To confirm the serine protease nature and specificity:

Inhibitor TypeExampleConcentration RangeExpected Effect
Serine proteasePMSF0.1-1 mMStrong inhibition
Chymotrypsin-likeChymostatin10-100 μMModerate inhibition
Trypsin-likeAprotinin0.1-10 μMLimited inhibition
MetalloproteaseEDTA1-10 mMNo inhibition
GZMH-specificCustom peptide inhibitors1-100 μMStrong, specific inhibition
  • pH and temperature profiling: Determine optimal conditions by measuring activity across pH range (5.0-9.0) and temperatures (25-45°C), which helps confirm proper folding and provides useful information for experimental design.

  • Cellular assays: For functional validation of purified GZMH:

    • Deliver GZMH to HBV-expressing cells (using protein transfection reagents)

    • Monitor HBx degradation by immunoblotting

    • Quantify HBV replication by qPCR

    • Compare results to positive controls (such as lymphokine-activated killer cells)

A comprehensive enzymatic characterization using these methods would provide robust validation of both the general proteolytic activity and the specific HBx-targeting function of the purified GZMH Human, sf9 preparation.

What are common challenges in working with GZMH Human, sf9, and how can researchers overcome them?

Researchers working with GZMH Human, sf9 may encounter several challenges that can affect protein quality, yield, and experimental results. The following table outlines common issues and their solutions:

ChallengePossible CausesSolutions
Low expression yieldSuboptimal infection conditions; poor cell viability; inefficient vectorOptimize MOI (2-5); ensure >95% cell viability before infection; verify baculovirus titer; extend harvest time to 72-96 hours
Protein degradationProteolytic activity in culture or during purification; improper storageAdd protease inhibitors during purification; maintain 4°C throughout processing; avoid freeze-thaw cycles; add carrier protein for storage
Loss of enzymatic activityImproper folding; denaturation during purification; aggregationInclude 10% glycerol in all buffers; avoid harsh elution conditions; verify protein folding using circular dichroism; optimize buffer pH and ionic strength
Heterogeneous glycosylationIntrinsic property of Sf9 expression systemAccept as an inherent characteristic; consider enzymatic deglycosylation if homogeneity is critical; alternatively, express in glycosylation-deficient Sf9 cells
Protein aggregationConcentration too high; improper buffer conditions; freeze-thaw damageKeep protein below 1 mg/ml; include stabilizers (glycerol, low concentrations of detergent); optimize buffer formulation based on thermal shift assays
Inconsistent activity in assaysVariable glycosylation; improper storage; assay interferenceNormalize by activity rather than concentration; include appropriate controls; prepare fresh aliquots for critical experiments
Difficulty detecting HBx cleavageSuboptimal reaction conditions; insufficient enzyme or substrate; detection limitsOptimize enzyme:substrate ratio; extend incubation time; use more sensitive detection methods (fluorescent tags, mass spectrometry)

Additionally, researchers may encounter challenges specific to the unique properties of GZMH. For instance, its tendency to cleave HBx at Met(79) might be affected by the structural context of the substrate. To overcome this, using full-length HBx protein rather than just peptide fragments can provide more physiologically relevant results.

For activity assays, background proteolytic activity from contaminating proteases can be minimized by including a panel of protease inhibitors that don't affect serine proteases (e.g., E-64, pepstatin A, EDTA) in the reaction buffer. This allows for more accurate measurement of GZMH-specific activity.

How can researchers validate that Sf9-expressed GZMH maintains appropriate structural and functional properties compared to the native protein?

Validating the structural and functional equivalence of Sf9-expressed GZMH to native human GZMH is crucial for ensuring experimental relevance. A comprehensive validation strategy should include both structural and functional analyses:

Structural Validation:

  • Mass Spectrometry Analysis: Peptide mapping using LC-MS/MS can confirm the primary sequence and identify post-translational modifications. While glycosylation patterns will differ from the native protein, the peptide backbone should be identical .

  • Circular Dichroism (CD) Spectroscopy: CD spectra in the far-UV range (190-260 nm) can confirm that the secondary structure content (α-helices, β-sheets) matches theoretical predictions for properly folded GZMH.

  • Thermal Shift Assays: Differential scanning fluorimetry using SYPRO Orange or similar dyes can assess protein stability and compare melting temperatures between recombinant and native GZMH samples.

  • Size Exclusion Chromatography with Multi-Angle Light Scattering (SEC-MALS): This technique can verify that the protein exists in the expected monomeric state and isn't forming unwanted aggregates.

Functional Validation:

  • Enzyme Kinetics Comparison: Determine key kinetic parameters (Km, kcat, kcat/Km) using standard peptide substrates and compare to literature values for native GZMH. While absolute values may differ, the substrate preference profile should be maintained.

  • HBx Cleavage Specificity: Verify that the recombinant GZMH cleaves HBx at the same position (Met79) as the native enzyme. This can be confirmed by:

    • N-terminal sequencing of cleavage products

    • Mass spectrometry analysis of fragments

    • Mutagenesis studies showing resistance of M79A mutants to cleavage

  • Inhibitor Sensitivity Profile: Compare IC50 values for a panel of serine protease inhibitors between recombinant and native GZMH. Similar inhibition patterns would suggest conserved active site architecture.

  • Functional Cell-Based Assays: Demonstrate that the recombinant GZMH can induce HBV clearance in cellular models when delivered appropriately, similar to native GZMH from immune cells .

A side-by-side comparison table can be created to document these validation parameters:

Validation ParameterNative GZMHSf9-expressed GZMHAcceptance Criteria
Molecular mass (deglycosylated)26.2 kDa26.2 kDa ± 0.5 kDaWithin 2% of theoretical mass
Secondary structure (α-helix %)Literature valueExperimental valueWithin 10% of reference
Thermal stability (Tm)Literature valueExperimental valueWithin 5°C of reference
Km for model substrateLiterature valueExperimental valueWithin 3-fold of reference
HBx cleavage siteMet79Met79Identical cleavage position
HBV clearance in cell modelsPositivePositiveStatistically significant activity

By systematically addressing these structural and functional parameters, researchers can confidently validate that their Sf9-expressed GZMH maintains the essential properties required for meaningful experimental applications.

What are potential applications of GZMH Human, sf9 beyond HBV research, and what experimental approaches would be needed?

While GZMH's role in HBV clearance is well-established, emerging research suggests broader applications for this serine protease. Several promising research directions extend beyond HBV:

  • Other viral infections: GZMH may target proteins from other viruses through similar mechanisms. To investigate this:

    • Perform in silico analysis of viral proteomes to identify potential cleavage sites similar to HBx Met(79)

    • Conduct in vitro cleavage assays with recombinant viral proteins

    • Validate findings in infection models using GZMH-expressing immune cells

  • Cancer immunotherapy: As a cytotoxic protease, GZMH may have applications in cancer cell targeting:

    • Evaluate GZMH-mediated cytotoxicity against various cancer cell lines

    • Identify cancer-specific substrates through proteomics approaches

    • Engineer delivery systems (such as immunoconjugates) to target GZMH to cancer cells

    • Assess synergy with existing immunotherapeutic approaches

  • Autoimmune disorders: Understanding GZMH's role in immune-mediated damage:

    • Profile GZMH expression in various autoimmune conditions

    • Identify self-antigens that might be targeted by GZMH

    • Develop specific inhibitors as potential therapeutic agents

    • Create animal models with GZMH deficiency or overexpression

  • Diagnostic biomarker development: GZMH levels or activity as indicators of immune activation:

    • Develop sensitive and specific assays for GZMH in biological fluids

    • Correlate GZMH levels with disease progression in viral infections or cancer

    • Evaluate GZMH as a predictive biomarker for treatment response

  • Structural biology and drug design: Using recombinant GZMH for inhibitor development:

    • Obtain high-resolution crystal structures of GZMH alone and in complex with substrates

    • Conduct structure-based virtual screening for novel inhibitors

    • Design peptidomimetic inhibitors based on optimal substrate sequences

    • Develop activity-based probes for imaging GZMH activity in cells and tissues

For each of these applications, the Sf9-expressed GZMH provides a consistent and scalable source of protein for both preliminary studies and advanced experimental approaches. The purified protein can be used to establish baseline enzymatic parameters, develop specific assays, and screen for interaction partners or inhibitors before moving to more complex cellular and animal models.

How might glycoengineering of Sf9 cells improve the properties of expressed GZMH for research applications?

Glycoengineering of Sf9 cells represents a promising approach to enhance the properties of expressed GZMH for advanced research applications. The native glycosylation pattern of Sf9 cells is limited primarily to paucimannose and high-mannose structures, which differs significantly from human glycosylation patterns . Strategic glycoengineering could address several limitations:

  • Humanization of glycosylation patterns: Engineering Sf9 cells to express mammalian glycosyltransferases would allow for more human-like glycosylation:

    • Introduction of β1,4-galactosyltransferase and α2,6-sialyltransferase genes

    • Expression of N-acetylglucosaminyltransferases to enable complex glycan formation

    • Knockout of insect-specific fucosyltransferases that create immunogenic epitopes

  • Elimination of potentially allergenic structures: Di-fucosylation at the GlcNAc bound to asparagine can elicit allergenic responses in mammals . Engineering approaches could:

    • Knockout or downregulate the fucosyltransferases responsible for core fucosylation

    • Express mammalian fucosidases to remove these modifications post-translationally

  • Site-specific glycan modification: Different glycosylation sites on GZMH may have varying importance for function and stability:

    • Identify critical glycosylation sites through mutagenesis studies

    • Engineer site-selective glycosylation using modified consensus sequences

    • Express glycosidases that target specific glycan structures

  • Enhancement of protein stability and solubility: Optimized glycosylation can improve biophysical properties:

    • Engineer glycans that increase solubility without affecting enzyme activity

    • Design glycosylation patterns that enhance thermal stability

    • Reduce glycan heterogeneity to improve batch-to-batch consistency

The benefits of glycoengineered GZMH would include:

PropertyStandard Sf9-GZMHGlycoengineered GZMHResearch Benefit
ImmunogenicityHigher due to insect-specific glycansLower with humanized glycansBetter compatibility for in vivo studies
StabilityVariableEnhancedLonger shelf-life and experimental windows
ConsistencyHeterogeneous glycoformsMore homogeneousImproved reproducibility between experiments
Structural studiesChallenging due to glycan heterogeneityFacilitated by uniform glycansHigher resolution structural data
Physiological relevanceLimited by non-human glycansImproved by mammalian-like glycansMore accurate modeling of in vivo function

Implementation strategies could include CRISPR/Cas9-mediated genome editing of Sf9 cells, construction of specialized baculovirus vectors carrying glycosylation enzymes, or development of entirely new insect cell lines optimized for human-like glycosylation. While technically challenging, such approaches would significantly enhance the utility of Sf9-expressed GZMH for both basic research and potential therapeutic applications.

What are the key considerations for researchers planning experiments with GZMH Human, sf9?

Researchers planning experiments with GZMH Human, sf9 should consider several critical factors to ensure successful outcomes. These considerations encompass technical aspects of handling the protein, experimental design parameters, and interpretative frameworks for results.

First, understanding the inherent properties of Sf9-expressed GZMH is essential. The recombinant protein contains insect-specific glycosylation patterns that differ from the native human enzyme . While the core enzymatic function is preserved, these structural differences may influence certain protein-protein interactions or immunological recognition. Researchers should validate whether these differences are relevant to their specific experimental questions.

Second, proper handling and storage protocols are crucial for maintaining GZMH activity. The protein should be stored at -20°C for long-term preservation, with addition of carrier proteins (0.1% HSA or BSA) for stability . Multiple freeze-thaw cycles must be avoided, and working aliquots should be prepared to minimize degradation .

Third, experimental design should include appropriate controls and validation steps. These include:

  • Enzyme activity controls using standard substrates

  • Specificity controls using HBx variants with mutations at the cleavage site

  • Negative controls using heat-inactivated GZMH

  • Positive controls such as native GZMH where available

Fourth, researchers should consider the biological context of their experiments. While in vitro studies with purified components provide valuable mechanistic insights, translating these findings to cellular and in vivo systems requires careful consideration of delivery methods, physiological concentrations, and potential interactions with endogenous inhibitors.

Finally, interpretation of results should acknowledge both the strengths and limitations of using recombinant GZMH. While it provides a consistent and readily available source of the enzyme for mechanistic studies, findings should ultimately be validated using primary immune cells expressing native GZMH when translational relevance is important.

Product Science Overview

Introduction to Granzymes

Granzymes are a family of serine proteases that play a crucial role in the immune system. They are primarily found in the granules of cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells. These enzymes are released during the immune response to target and induce apoptosis in infected or malignant cells .

Granzyme-H: An Overview

Granzyme-H (GzmH) is one of the five human granzymes, alongside Granzyme A, B, K, and M. It is a chymotrypsin-like serine protease predominantly expressed in NK cells . GzmH has unique substrate specificity and plays a significant role in the innate immune response against tumors and viral infections .

Recombinant Granzyme-H Production

The recombinant form of Granzyme-H, produced in sf9 insect cells, is utilized for research and therapeutic purposes. The sf9 cell line, derived from the fall armyworm (Spodoptera frugiperda), is commonly used in biotechnology for the production of recombinant proteins. This system allows for the expression of human proteins with post-translational modifications similar to those in mammalian cells .

Structural Insights

The structure of Granzyme-H has been studied to understand its substrate specificity and mechanism of action. The enzyme contains a unique RKR motif that helps define its binding regions, indicating a preference for acidic residues at specific sites . This structural information is crucial for designing inhibitors that can selectively block the enzymatic and cytotoxic activity of GzmH .

Biological Functions

Granzyme-H is involved in the immune defense against transformed and virus-infected cells. It induces apoptosis through various pathways, contributing to the elimination of harmful cells. Recent studies have also highlighted its role in extracellular matrix alteration and tissue homeostasis .

Therapeutic Potential

Given its role in immune responses, Granzyme-H has potential therapeutic applications. Understanding its structure and function can aid in the development of targeted therapies for cancer and viral infections. Additionally, recombinant Granzyme-H can be used in research to further explore its biological activities and potential clinical 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 2024 Thebiotek. All Rights Reserved.