ANG Human, Sf9

ANG Human Recombinant, Sf9
Shipped with Ice Packs
In Stock

Description

ANG produced in Sf9 Baculovirus cells is a single, glycosylated polypeptide chain containing 132 amino acids (25-147a.a.) and having a molecular mass of 15.2kDa.
ANG is fused to a 6 amino acid His-Tag at C-terminus and purified by proprietary chromatographic techniques.

Product Specs

Introduction

Angiogenin (ANG), a member of the RNase A family, plays a crucial role in the formation of new blood vessels (angiogenesis). It achieves this by cleaving tRNA at specific sites (anticodon loops), generating tRNA-derived stress-induced fragments (tiRNAs). These tiRNAs have been shown to suppress protein synthesis and initiate the formation of stress granules. Additionally, ANG contributes to the synthesis of ribosomal RNA. Its interaction with RNH1, observed in living organisms (in vivo), is vital for regulating its angiogenic activity.

Description

Produced using Sf9 insect cells (specifically, Baculovirus expression system), our ANG protein is a single, glycosylated polypeptide chain comprising 132 amino acids (spanning positions 25 to 147a.a.). It has a molecular weight of 15.2 kDa. For ease of purification and detection, a 6-amino acid Histidine tag (His-Tag) is fused to the C-terminus of the protein. Purification is carried out using proprietary chromatographic techniques.

Physical Appearance
A clear, colorless solution that has undergone sterile filtration.
Formulation

The ANG protein is supplied in a solution containing 0.25mg/ml of ANG protein, 10% glycerol, and Phosphate Buffered Saline (pH 7.4).

Stability
For short-term storage (2-4 weeks), the ANG protein can be stored at 4°C. For extended storage, it is recommended to freeze the protein at -20°C. The addition of a carrier protein such as 0.1% HSA or BSA is advised for long-term storage. It is crucial to avoid repeated cycles of freezing and thawing to maintain protein integrity and activity.
Purity

The purity of our ANG protein is greater than 90% as determined by SDS-PAGE analysis.

Synonyms

Angiogenin, ANG, Ribonuclease 5, RNase 5, RNASE5, ribonuclease, RNase A family, 5, ALS9, HEL168, MGC22466, MGC71966, RNASE4, RAA1.

Source

Sf9, Baculovirus cells.

Amino Acid Sequence

ADPQDNSRYT HFLTQHYDAK PQGRDDRYCE SIMRRRGLTS PCKDINTFIH GNKRSIKAIC ENKNGNPHRE NLRISKSSFQ VTTCKLHGGS PWPPCQYRAT AGFRNVVVAC ENGLPVHLDQ SIFRRPHHHH HH.

Q&A

What makes Sf9 cells suitable for human ANG expression?

Sf9 cells have become a preferred platform for heterologous protein expression due to their ability to perform complex post-translational modifications while maintaining high expression yields. These insect cells have been the expression system of choice for numerous membrane proteins that have yielded over 100 high-resolution structures, including eukaryotic transporters, ATP-gated channels, receptors, and regulatory proteins . For human ANG expression specifically, Sf9 cells offer significant advantages in proper protein folding and biological activity preservation compared to bacterial expression systems.

When optimizing Sf9 expression systems, researchers typically determine optimal conditions through pilot studies varying the multiplicity of infection (MOI) and cell density. For instance, studies with other human proteins have shown optimal expression at an MOI of 2 and a density of 2 × 10^6 cells/ml, with protein expression peaking approximately 48 hours post-infection when cell viability drops to around 40% . Similar optimization protocols can be applied for human ANG expression.

How do I confirm the functionality of expressed human ANG in Sf9 cells?

Confirming functionality of human ANG expressed in Sf9 cells requires multiple approaches:

  • Western blot analysis: Detect the expressed protein using antibodies against ANG or any tags incorporated into the construct. Expression can be monitored at different time points post-infection to determine optimal harvest time, similar to other protein expression studies in Sf9 cells .

  • Ribonucleolytic activity assay: Since human ANG possesses ribonucleolytic activity, functionality can be assessed using standard RNase assays.

  • Angiogenesis assays: Functional ANG promotes angiogenesis, which can be confirmed through in vitro endothelial cell proliferation, migration, or tube formation assays.

  • Glycosylation analysis: Human ANG may undergo N-glycosylation in Sf9 cells. Treatment with PNGase F under non-denaturing conditions can help identify if expressed ANG is glycosylated, as demonstrated in other protein studies where glycosylated proteins show a band shift after enzyme treatment .

What are the key considerations for designing a baculovirus vector for human ANG expression?

When designing a baculovirus vector for human ANG expression, consider the following critical factors:

  • Signal sequence selection: The native human ANG signal sequence may be replaced with an insect-specific signal sequence (like gp67) to enhance secretion in Sf9 cells.

  • Affinity tag placement: N-terminal or C-terminal tags (His6, GST, or MBP) facilitate purification but may affect ANG activity. C-terminal tags are often preferred as they less frequently interfere with signal sequence processing.

  • Promoter selection: The polyhedrin or p10 promoter is typically used for high-level expression in the late phase of infection.

  • Codon optimization: While not always necessary, adapting the human ANG coding sequence to Sf9 codon usage preferences can potentially improve expression levels.

  • Cell line selection: Consider using the novel Sf-RVN cell line, which is free from rhabdovirus contamination while maintaining similar growth and morphological characteristics to standard Sf9 cells . This is particularly important for research that may lead to therapeutic applications.

How can I optimize human ANG yield and solubility in Sf9 cells?

Optimizing human ANG yield and solubility in Sf9 cells requires a multifactorial approach:

  • Infection parameters optimization:

    • Conduct a matrix experiment varying MOI (0.5-10) and cell density (1-3 × 10^6 cells/ml)

    • Monitor protein expression at different time points (24-96 hours post-infection)

    • Typical optimal conditions for many proteins are MOI of 2, cell density of 2 × 10^6 cells/ml, and harvest at 48 hours post-infection

  • Medium formulation:

    • Use chemically defined media specifically optimized for Sf9 cells

    • Consider EX-CELL CD Insect Cell Medium, which has been shown to support robust growth and high productivity of various proteins in Sf9-RVN cells

  • Temperature modulation:

    • Lower culture temperature (24-26°C instead of 27-28°C) during expression phase may improve solubility

    • Implement temperature shifts: grow cells at 27°C, then reduce to 24°C post-infection

  • Addition of solubility enhancers:

    AdditiveWorking ConcentrationEffect on Protein Solubility
    Glycerol2-10%Stabilizes protein structure
    Arginine50-200 mMReduces aggregation
    Sorbitol0.5-1 MEnhances folding
    DMSO0.5-2%Aids in protein solubilization
  • Co-expression strategies:

    • Co-express chaperones (such as Hsp70, BiP) to assist proper folding

    • Use dual promoter vectors for simultaneous expression of ANG and folding partners

What are the major challenges in purifying human ANG from Sf9 cells and how can they be addressed?

Purifying human ANG from Sf9 cells presents several challenges that require specific strategies:

  • Cellular localization determination:

    • Determine if ANG is secreted into the medium or retained intracellularly

    • For secreted ANG, concentrate the medium using tangential flow filtration

    • For intracellular ANG, optimize cell lysis conditions (sonication, French press, or detergents)

  • Purification strategy development:

    • Initial capture: Utilize affinity chromatography (if tagged) or ion exchange chromatography

    • Intermediate purification: Apply size exclusion chromatography to separate monomeric ANG from aggregates

    • Polishing step: Employ hydrophobic interaction chromatography or a second ion exchange step

  • Addressing proteolytic degradation:

    • Add protease inhibitor cocktails during extraction and purification

    • Process samples quickly and maintain low temperatures (4°C)

    • Consider adding EDTA (1-5 mM) to inhibit metalloproteases

  • Overcoming contamination with endogenous particles:

    • Sf9 cells produce endogenous retroviral-like particles with RT activity

    • These particles have a buoyant density of about 1.08 g/mL and can be induced by treatment with 5-iodo-2′-deoxyuridine (IUdR)

    • Use additional purification steps (density gradient centrifugation) to separate protein from these particles

    • Consider using the rhabdovirus-negative Sf-RVN cell line to reduce contamination risks

  • Maintaining biological activity:

    • Include stabilizers (glycerol, trehalose) in purification buffers

    • Avoid freeze-thaw cycles by aliquoting purified protein

    • Test various buffer compositions to identify optimal stability conditions

How can I address inconsistent glycosylation patterns in human ANG expressed in Sf9 cells?

Inconsistent glycosylation of human ANG in Sf9 cells is a common challenge that can be addressed through several approaches:

  • Characterization of glycosylation heterogeneity:

    • Use mass spectrometry to identify glycoforms

    • Apply lectin blotting to characterize specific glycan structures

    • Conduct PNGase F treatment under non-denaturing conditions to confirm N-linked glycosylation, as demonstrated with other proteins in Sf9 cells

  • Strategies to improve glycosylation consistency:

    • Cell culture optimization:

      • Maintain consistent cell density and viability

      • Standardize medium composition and supplement with glycosylation precursors (mannose, GlcNAc)

      • Control infection parameters (MOI, time of harvest)

    • Genetic modifications:

      • Consider using engineered Sf9 cell lines with humanized glycosylation pathways

      • Introduce genes encoding human glycosyltransferases

    • Process modifications:

      ModificationImplementation ApproachExpected Outcome
      Temperature shiftLower to 24°C post-infectionMore complete glycosylation
      Nutrient supplementationAdd UDP-GlcNAc and GDP-mannoseEnhanced glycosylation substrate availability
      pH controlMaintain at 6.2-6.4Optimized glycosyltransferase activity
      Osmolality adjustment300-320 mOsm/kgImproved glycosylation consistency
  • Post-purification approaches:

    • Use lectin affinity chromatography to isolate specific glycoforms

    • Consider enzymatic homogenization of glycans using endoglycosidases

    • Implement in vitro glycan remodeling with glycosyltransferases

How do endogenous retroviral-like particles in Sf9 cells potentially impact human ANG studies?

The presence of endogenous retroviral-like particles in Sf9 cells poses several considerations for human ANG studies:

  • Characterization of particle interference:
    Recent studies have identified that Sf9 cells constitutively produce retroviral-like particles containing reverse transcriptase (RT) activity . These particles have diverse sizes and morphologies, including viral-like particles and extracellular vesicles, with a low buoyant density of approximately 1.08 g/mL . When purifying ANG, these particles may co-purify depending on the methods used.

  • Inducible nature of retroviral-like particles:
    Chemical treatments like 5-iodo-2′-deoxyuridine (IUdR) can induce a 33-fold higher RT activity in Sf9 cells . Researchers should be aware that certain experimental treatments or stress conditions could potentially increase particle production, affecting downstream analyses.

  • Biosafety considerations:
    Infectivity studies using various human and non-human primate cell lines have shown no evidence of replicating retrovirus or virus entry . This suggests minimal risk for human infection, but regulatory agencies may still require testing for adventitious agents in ANG preparations intended for certain applications.

  • Mitigation strategies:

    • Use density gradient separation techniques to isolate ANG from retroviral-like particles

    • Consider using the Sf-RVN cell line, which has been developed as a rhabdovirus-negative alternative while maintaining similar growth characteristics to standard Sf9 cells

    • Implement additional filtration steps (nanofiltration) to remove viral-sized particles

    • Apply nuclease treatments to reduce nucleic acid contamination

What are the comparative advantages of Sf9 versus HEK293 cells for structural studies of human ANG?

When conducting structural studies of human ANG, researchers must carefully consider the expression system. Both Sf9 and HEK293 cells offer distinct advantages:

How can I establish a reliable quantitative assay for comparing the biological activity of Sf9-expressed versus native human ANG?

Establishing a reliable quantitative assay to compare the biological activity of Sf9-expressed versus native human ANG requires multiple complementary approaches:

  • Ribonucleolytic activity assays:

    • Substrate specificity analysis: Use synthetic RNA substrates containing ANG-specific cleavage sites

    • Kinetic parameters determination: Calculate Kcat/Km values to compare catalytic efficiency

    • Inhibition profile: Test sensitivity to ribonuclease inhibitor (RI)

    Standardize assay conditions (pH, temperature, buffer composition) and include reference standards of known activity for normalization between experiments.

  • Cell-based angiogenesis assays:

    • Endothelial cell proliferation: Measure BrdU incorporation or MTT reduction in HUVECs

    • Migration assays: Quantify scratch wound healing or transwell migration

    • Tube formation: Analyze pattern formation on Matrigel

    For all cell-based assays, develop dose-response curves (EC50) and calculate relative potencies compared to native ANG.

  • Receptor binding and internalization studies:

    • Surface plasmon resonance: Determine binding kinetics (kon, koff, KD) to ANG receptors

    • Fluorescence microscopy: Track internalization of fluorescently labeled ANG

    • Receptor activation: Measure downstream signaling events (e.g., phosphorylation)

  • Comparative potency determination:

    Assay TypeMeasurement ParameterAcceptance Criteria
    RibonucleolyticRelative activity (%)80-120% of reference
    Endothelial proliferationEC50 ratio0.8-1.2
    Receptor bindingKD ratio0.8-1.2
    Nuclear translocationNuclear/cytoplasmic ratio≥90% of reference
  • Statistical validation:

    • Determine assay precision through replicate testing (target CV ≤15%)

    • Establish assay accuracy using reference standards (recovery 80-120%)

    • Calculate the minimum significant ratio (MSR) for comparative testing

    • Use appropriate positive and negative controls for system suitability

To ensure reliable comparisons, implement a reference standard calibration approach where each test includes a dose-response curve of native human ANG. Express results as relative potency to minimize inter-assay variability. For comprehensive characterization, employ orthogonal methods and correlate findings across different biological activity assays.

What emerging technologies could improve human ANG expression in insect cell systems?

Several cutting-edge technologies hold promise for enhancing human ANG expression in insect cell systems:

  • CRISPR/Cas9 genome editing of Sf9 cells:

    • Generate knockout lines lacking proteases that might degrade ANG

    • Engineer enhanced secretion pathway components

    • Create humanized glycosylation pathways for more authentic post-translational modifications

    • Develop Sf9 cells with integrated biosafety features, building upon the rhabdovirus-negative Sf-RVN cell line approach

  • Synthetic biology approaches:

    • Design entirely synthetic promoters optimized for ANG expression

    • Create synthetic untranslated regions (UTRs) to enhance mRNA stability and translation efficiency

    • Develop synthetic signal sequences specifically optimized for ANG secretion

  • Advanced baculovirus expression systems:

    • MultiBac system for co-expression of ANG with chaperones or binding partners

    • flashBAC ultraTM system for improved protein expression and reduced proteolysis

    • Disabled viral chitinase and cathepsin genes to enhance protein stability

  • Novel bioprocess technologies:

    TechnologyApplicationPotential Benefit
    Perfusion bioreactorsContinuous processing3-5× higher volumetric productivity
    Acoustic cell retentionHigh-density cultureUp to 10× higher cell densities
    Microfluidic systemsProcess intensificationReduced process development time
    Single-use technologiesFlexible manufacturingDecreased contamination risk
  • Artificial intelligence and machine learning:

    • Predict optimal codon usage for ANG expression in Sf9 cells

    • Design experimental matrices for multiparameter optimization

    • Develop predictive models for protein yield based on vector design and culture conditions

How might the structural differences between Sf9-expressed and native human ANG impact therapeutic applications?

Structural differences between Sf9-expressed and native human ANG may have significant implications for therapeutic applications:

  • Glycosylation-related considerations:

    • Sf9 cells produce high-mannose type N-glycans rather than complex mammalian glycans

    • These differences may affect:

      • Serum half-life (typically shorter for high-mannose glycans)

      • Immunogenicity (high-mannose glycans may be more immunogenic)

      • Tissue distribution and receptor binding kinetics

    • Potential mitigation through glycoengineered Sf9 cell lines or chemoenzymatic remodeling of glycans post-purification

  • Disulfide bond formation and tertiary structure:

    • Human ANG contains three disulfide bonds critical for its structure and function

    • While Sf9 cells are generally efficient at disulfide bond formation, incorrect pairing can occur

    • Structural characterization using circular dichroism, thermal shift assays, and limited proteolysis can identify potential differences

    • X-ray crystallography or cryo-EM can provide definitive structural comparisons

  • Potential contaminants and their impact:

    • Endogenous retroviral-like particles from Sf9 cells could co-purify with ANG

    • While these particles have not shown infectivity in human cells , they may:

      • Trigger innate immune responses

      • Interfere with ANG activity assays

      • Raise regulatory concerns for therapeutic applications

    • The use of rhabdovirus-negative Sf-RVN cells may help address these concerns

  • Stability and aggregation propensity:

    • Differences in post-translational modifications may affect protein stability

    • Comparative studies should assess:

      • Thermal stability (Tm values)

      • Resistance to proteolysis

      • Aggregation kinetics under physiological conditions

      • Long-term storage stability

  • Functional comparison framework:

    ParameterAssessment MethodAcceptance Criteria for Therapeutic Use
    Structural similarityCD spectroscopy, FTIR≥90% spectral overlap
    Thermal stabilityDSC, DSFΔTm ≤ 3°C
    Ribonucleolytic activityEnzymatic assays80-120% of native ANG
    Receptor activationCell signaling assaysEC50 within 0.8-1.2× range
    ImmunogenicityT-cell activation, ADANo significant increase

For therapeutic development pathways, regulatory authorities may require additional characterization and comparability studies between Sf9-expressed and mammalian cell-expressed or native human ANG. Early consultation with regulatory agencies is recommended to establish acceptable criteria for structural and functional similarity.

What are the best methods for monitoring and optimizing baculovirus infection efficiency for human ANG expression?

Optimizing baculovirus infection efficiency for human ANG expression requires robust monitoring techniques and systematic parameter adjustment:

  • Quantitative analysis of viral titer:

    • Plaque assay: Traditional gold standard but labor-intensive

    • End-point dilution (TCID50): More sensitive but time-consuming

    • qPCR-based methods: Rapid and precise quantification of viral DNA

    • Flow cytometry: Using fluorescent reporter genes in recombinant baculovirus

  • Infection parameter optimization matrix:

    • MOI optimization: Test range from 0.1-10 MOI

    • Cell density at infection: Optimal typically at 1-2 × 10^6 cells/ml

    • Time of harvest: Monitor expression at 24-96 hours post-infection

    • Culture medium: Consider specialized formulations like EX-CELL CD Insect Cell Medium

  • Real-time monitoring approaches:

    • Reporter gene co-expression: Include fluorescent proteins under the control of the same or independent promoter

    • Inline dielectric spectroscopy: Monitor changes in cell morphology during infection

    • Metabolic flux analysis: Track changes in glucose consumption and lactate production

    • Oxygen uptake rate (OUR): Changes correlate with infection progression

  • Visualization techniques:

    TechniqueInformation ProvidedTiming Post-Infection
    Light microscopyCell enlargement, density12-96 hours
    Viability stainsMembrane integrity24-96 hours
    Fluorescence microscopyProtein localization (if tagged)24-72 hours
    Electron microscopyVirus particle formation24-72 hours
  • Expression monitoring protocol:

    • Collect samples at 24, 48, 72, and 96 hours post-infection

    • Track cell viability and diameter (optimal harvest typically when viability drops to 40-60%)

    • Analyze ANG expression by Western blot using antibodies against ANG or affinity tags

    • Consider developing an ELISA for rapid quantification of ANG in culture supernatant

How can I develop a robust purification protocol for human ANG from Sf9 cells that maximizes yield and maintains activity?

Developing a robust purification protocol for human ANG from Sf9 cells requires a systematic approach balancing yield, purity, and activity preservation:

  • Harvest and initial processing optimization:

    • Determine optimal harvest time (typically 48-72 hours post-infection)

    • Evaluate centrifugation parameters (speed, time, temperature) for cell separation

    • For secreted ANG, concentrate culture supernatant using tangential flow filtration

    • For intracellular ANG, optimize lysis conditions (buffer composition, mechanical methods)

  • Multistep purification strategy:

    • Capture step: Immobilized metal affinity chromatography (IMAC) for His-tagged ANG or ion exchange chromatography for untagged protein

    • Intermediate purification: Size exclusion chromatography to remove aggregates and separate monomeric ANG

    • Polishing step: Hydrophobic interaction chromatography or a second ion exchange step

  • Critical process parameter identification:

    ParameterOptimization RangeImpact on Results
    pH5.0-8.0Affects binding efficiency and stability
    Salt concentration0-500 mMInfluences ionic interactions and solubility
    Reducing agents0-5 mM DTT/BMEImpact on disulfide bonds (use with caution)
    Stabilizing additives5-20% glycerol, 0.1-1 M sucroseEnhance stability during purification
    Flow rate0.5-5 ml/minAffects resolution and binding efficiency
  • Activity preservation strategies:

    • Maintain low temperature (4°C) throughout purification

    • Include protease inhibitors in all buffers

    • Minimize exposure to air/oxidation

    • Add stabilizers like glycerol or sucrose to final formulation

    • Consider arginine as an aggregation suppressor

  • Separation from retroviral-like particles:

    • Implement density gradient ultracentrifugation steps to separate ANG from Sf9 endogenous retroviral-like particles that have a buoyant density of approximately 1.08 g/mL

    • Apply size-based separation techniques (filtration through appropriate molecular weight cutoffs)

    • Consider using the rhabdovirus-negative Sf-RVN cell line to reduce initial contamination

  • Quality control checkpoints:

    • Purity: SDS-PAGE, SEC-HPLC (target >95% purity)

    • Identity: Mass spectrometry, N-terminal sequencing

    • Activity: Ribonucleolytic assay, cell-based functional assays

    • Endotoxin and host cell protein: LAL test, ELISA (critical for therapeutic applications)

    • Viral clearance validation: Demonstrate removal of retroviral-like particles if using standard Sf9 cells

Product Science Overview

Structure and Production

ANG produced in Sf9 Baculovirus cells is a single, glycosylated polypeptide chain containing 132 amino acids (25-147a.a.) and has a molecular mass of approximately 15.2 kDa . The molecular size on SDS-PAGE appears at approximately 13.5-18 kDa . This recombinant form of ANG is fused to a 9 amino acid His-Tag at the C-terminus, which facilitates its purification through chromatographic techniques .

Mechanism of Action

ANG cleaves transfer RNA (tRNA) within anticodon loops to produce tRNA-derived stress-induced fragments (tiRNAs). These tiRNAs inhibit protein synthesis and trigger the assembly of stress granules, which are involved in cellular stress responses . Additionally, ANG stimulates ribosomal RNA synthesis, contributing to its role in promoting cell growth and survival .

Biological Functions
  1. Angiogenesis: ANG is a highly effective mediator of new blood vessel formation. This function is critical in processes such as wound healing and tumor growth, where the formation of new blood vessels is essential for supplying nutrients and oxygen to tissues .

  2. Neuroprotection: ANG has been shown to have neuroprotective effects, particularly in the context of neurodegenerative diseases. It helps in the survival of motor neurons and has been implicated in conditions like amyotrophic lateral sclerosis (ALS) .

  3. Stress Response: By producing tiRNAs, ANG plays a role in the cellular response to stress. These fragments help in the formation of stress granules, which are involved in protecting cells under adverse conditions .

Applications

The recombinant form of ANG produced in Sf9 cells is primarily used for laboratory research purposes. It is utilized in studies related to angiogenesis, cancer research, and neuroprotection. The protein is supplied as a sterile, filtered, colorless solution and is formulated with 10% glycerol and phosphate-buffered saline (pH 7.4) .

Storage and Stability

For optimal stability, ANG should be stored at 4°C if it will be used within 2-4 weeks. For longer storage periods, it is recommended to keep the protein frozen at -20°C. To prevent degradation, it is advisable to add a carrier protein (0.1% HSA or BSA) and avoid multiple freeze-thaw cycles .

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.