IFN a 2a Human

IFN-Alpha 2a Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biochemical Characteristics

IFNα2a is a 166-amino acid glycoprotein with a molecular weight of 18–22 kDa . Key properties include:

PropertyValueSource
Molecular Weight19–22 kDa (reduced); 14–19 kDa (non-reduced)
Purity≥95% (SDS-PAGE, HPLC)
Specific Activity≥1.0 × 10⁷ IU/mg
Endotoxin Levels<1 EU/μg
Expression SystemE. coli, HEK293

The protein shares 59% amino acid sequence identity with murine IFNα2 and activates the IFNAR1/IFNAR2 receptor complex .

Mechanism of Action

IFNα2a binds to the type I interferon receptor (IFNAR), triggering a signaling cascade:

  1. JAK-STAT Activation: Phosphorylation of TYK2 and JAK1 kinases leads to STAT1/STAT2 activation .

  2. ISGF3 Formation: STAT1/STAT2 dimerize with IRF9, forming the ISGF3 complex that translocates to the nucleus .

  3. Gene Transcription: ISGF3 induces >300 interferon-stimulated genes (ISGs), inhibiting viral replication and enhancing immune surveillance .

Antiviral Use

  • Hepatitis B/C: Reduces viral load by activating innate antiviral pathways .

  • HIV: Enhances immune response in combination therapies .

Anticancer Use

Cancer TypeResponse RateStudy
Multiple Myeloma22.4%
Renal Cell Carcinoma13.8%
Cutaneous Lymphoma100%
Metastatic Colorectal CancerNo added benefit vs. 5-FU alone

A phase III trial in colorectal cancer (n=245) showed no survival benefit for IFNα2a + 5-FU vs. 5-FU alone (median survival: 13.9 vs. 13.2 months) . Toxicity (fatigue, leukopenia) was higher in the IFNα2a group .

Manufacturing and Standardization

  • WHO Standard: The 2nd WHO International Standard (NIBSC 95/650) ensures bioassay consistency .

  • Production Systems:

    • E. coli-derived IFNα2a: Non-glycosylated, lyophilized .

    • HEK293-derived IFNα2a: Glycosylated, higher stability .

ManufacturerCatalogActivityPrice
BPS Bioscience90158-A≥98% purity$130
ProteintechHZ-1066≤0.06 ng/mL EC₅₀N/A
PBL Assay Science11100-11.0 × 10⁷ IU/mg$335

Research Findings

  • Antiviral Activity: ED₅₀ of 3–30 pg/mL against encephalomyocarditis virus in HeLa cells .

  • Immune Modulation: Enhances NK cell activity and upregulates MHC class I expression .

  • SARS-CoV-2: IFNα2a induces ACE2 isoform expression in respiratory epithelia, potentially influencing viral entry .

Product Specs

Introduction
IFN-alpha, produced by macrophages, exhibits antiviral properties by stimulating the production of protein kinase and oligoadenylate synthetase.
Description
Recombinant Human IFN Alpha 2a, produced in E. coli, is a non-glycosylated polypeptide chain consisting of 165 amino acids, with a molecular weight of 19241 Dalton. The distinction between IFNA2A and IFNA2B lies in the amino acid at position 23, with IFNA2A having lysine and IFNA2B having arginine. The gene encoding Interferon-alpha-2a was derived from human leukocytes. Purification of IFNA2A is achieved through proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White Lyophilized Powder
Formulation
Lyophilized without any additional ingredients.
Solubility
Reconstitute the lyophilized Interferon-alpha-2a in sterile 18MΩ-cm H2O at a minimum concentration of 100µg/ml. This solution can be further diluted with other aqueous solutions.
Stability
While Lyophilized IFNA-2A remains stable at room temperature for 3 weeks, it is recommended to store it desiccated below -18°C. After reconstitution, store IFN-alpha 2a at 4°C for 2-7 days. For long-term storage, freeze at -18°C after adding a carrier protein (0.1% HSA or BSA). Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 97.0% as determined by both RP-HPLC and SDS-PAGE analysis.
Biological Activity
The specific activity, determined through a viral resistance assay using bovine kidney MDBK cells, is 270,000,000 IU/mg.
Protein Content
Protein quantification was performed using two independent methods: UV spectroscopy at 280 nm (extinction coefficient of 0.924 for a 0.1% solution, calculated using PC GENE software) and RP-HPLC analysis with a calibrated IFN-a 2a Reference Standard.
Synonyms

Leukocyte IFN, B cell IFN, Type I IFN, IFNA2, IFN-a 2a, Interferon-alpha-2a.

Source
Escherichia Coli.
Amino Acid Sequence

CDLPQTHSLGSRRTLMLLAQMRKISLFSCLKDRHDFGFPQEEFGNQFQKAETIPVLHEM IQQIFNLFSTKDSSAAWDETLLDKFYTELYQQLNDLEACVIQGVGVTETPLMKEDSILAV RKYFQRITLYLKEKKYSPCAWEVVRAEIMRSFSLSTNLQESLRSKE.

N-terminal methionine has been completely removed enzymatically.

Q&A

How does IFN-α 2a differ from other type I interferons?

IFN-α 2a belongs to a family of 13 functional subtypes of IFN-α that share 75-99% sequence homology . Specifically, IFN-α 2a differs from IFN-α 2b by only one amino acid, yet this subtle difference influences receptor binding kinetics and downstream biological responses .

All Type I interferons (including IFN-α subtypes) signal through a common cell surface receptor composed of two subunits: a 100 kDa ligand-binding subunit (IFNAR2) and a 125 kDa ligand-binding and signal transduction subunit (IFNAR1) . Despite using the same receptor complex, different subtypes can elicit varying degrees of antiviral, anti-proliferative, and immunomodulatory effects, suggesting subtle differences in receptor engagement dynamics and downstream signaling pathway activation.

What analytical methods are recommended for verifying IFN-α 2a identity and purity?

For comprehensive characterization of IFN-α 2a, researchers should employ multiple complementary methods:

Protein identity and purity:

  • SDS-PAGE (≥95% purity under both reducing and non-reducing conditions)

  • Mass spectrometry (for precise molecular weight determination and detection of modifications)

  • Peptide mapping/mass fingerprinting (for sequence verification)

Structural integrity:

  • Two-dimensional 15N-1H heteronuclear single quantum coherence (HSQC) NMR spectroscopy (for tertiary structure confirmation)

  • Circular dichroism (for secondary structure assessment)

  • Size exclusion chromatography (to detect aggregation and fragmentation)

Biological activity:

  • Antiviral cytopathic effect (CPE) inhibition assays:

    • A549/EMCV system (EC50 ≤ 0.005 ng/mL; ≥ 2.0 × 108 units/mg)

    • MDBK/VSV system

Contaminant analysis:

  • Endotoxin testing (acceptance criterion: ≤1 EU/μg)

  • Host cell protein ELISA (for recombinant preparations)

What expression systems provide optimal yield of biologically active IFN-α 2a?

Multiple expression systems can produce recombinant human IFN-α 2a, each with distinct advantages:

E. coli expression systems:

  • Most widely used for research-grade IFN-α 2a as the protein lacks glycosylation

  • Cold induction (reduced temperature) protocols significantly improve soluble protein yield

  • Codon-optimized strains enhance expression of this human protein in bacterial systems

  • SUMO-fusion strategies allow production of IFN-α 2a with the native N-terminus after tag removal

Mammalian expression systems:

  • HEK293 cells can produce IFN-α 2a with mammalian post-translational modifications

  • Typically yield lower protein amounts but may provide advantages for certain applications

For maximizing both yield and activity in E. coli systems, the following approach is recommended:

  • Use expression vectors with SUMO or similar precision-cleavable tags

  • Employ cold induction protocols (15-18°C for 16+ hours)

  • Include solubility enhancers in lysis buffers (e.g., mild detergents or stabilizing agents)

What purification strategy yields the highest specific activity for recombinant IFN-α 2a?

An optimized multi-step purification strategy is essential for obtaining IFN-α 2a with maximum specific activity:

Step 1: Initial capture

  • For tagged constructs: Immobilized metal affinity chromatography (IMAC)

  • For untagged proteins: Cation exchange chromatography (pH 4-5) exploiting IFN-α 2a's basic isoelectric point

Step 2: Tag removal (if applicable)

  • SUMO protease digestion followed by reverse IMAC to remove both tag and protease

  • Precision proteases that leave no additional amino acids at the N-terminus are preferred

Step 3: Polishing

  • Size exclusion chromatography to remove aggregates and ensure monomeric state

  • Separation from contaminating proteins with similar binding properties

Critical considerations throughout purification:

  • Buffer systems at pH 4.5-5.5 often provide optimal stability

  • Include carrier proteins (0.1-0.3% BSA) in final formulations to prevent activity loss through adsorption

  • Minimal exposure to room temperature; maintain samples at 2-8°C during purification

  • Filter sterilization using low protein-binding filters for final preparations

This strategy typically yields >95% pure protein with specific activity of ≥2.0 × 108 units/mg in antiviral assays .

How can researchers troubleshoot expression problems with IFN-α 2a?

When facing challenges with IFN-α 2a expression, researchers should systematically address these common issues:

For poor expression levels:

  • Verify codon optimization for expression host

  • Test multiple promoter systems (T7, tac, etc.)

  • Optimize induction conditions (IPTG concentration, timing, temperature)

  • Screen multiple expression strains with different genetic backgrounds

For inclusion body formation:

  • Reduce induction temperature (15-18°C) and extend induction time (16-24 hours)

  • Lower inducer concentration (0.1-0.5 mM IPTG)

  • Test solubility-enhancing fusion partners (SUMO, thioredoxin, MBP)

  • Co-express with molecular chaperones (GroEL/GroES, DnaK/DnaJ)

For poor solubility during purification:

  • Adjust lysis buffer composition (add stabilizers like glycerol, mild detergents)

  • Evaluate different pH conditions (typically pH 4-6 range)

  • Consider step-wise refolding protocols if necessary

For low biological activity:

  • Confirm correct disulfide bond formation

  • Ensure carrier protein is present in dilute solutions to prevent adsorption losses

  • Verify protease cleavage resulted in correct N-terminal sequence

  • Test activity immediately after purification before storage/freeze-thaw cycles

Systematic documentation of conditions tested and results observed will facilitate identification of the specific bottleneck limiting successful expression.

What are the standard bioassays for measuring IFN-α 2a activity?

Several validated bioassays are available for quantifying IFN-α 2a activity, each with distinct advantages for different research applications:

Antiviral cytopathic effect (CPE) inhibition assays:

Cell Line/VirusEC50 ValueAdvantagesApplications
A549/EMCV~1 U/mLHuman cell line, high sensitivityPotency determination, human-specific activity
MDBK/VSV~5 U/mLHistorical standard, robust systemStandardized potency testing, cross-species studies

Key methodological considerations:

  • Activity should be expressed in International Units (IU) calibrated against the WHO International Standard for IFN-α 2a (NIBSC code: 95/650)

  • Full dose-response curves with 8-12 dilutions are recommended for accurate potency determination

  • Include appropriate controls (positive reference standard, negative control, neutralizing antibody control)

  • Report specific activity (units/mg protein) to normalize for protein concentration differences

For the highest reliability in comparative studies, researchers should:

  • Use parallel line bioassay statistical methods for potency calculations

  • Perform at least three independent assays

  • Report confidence intervals for potency estimates

  • Document complete assay conditions including cell passage number and virus stock

How should IFN-α 2a be handled to preserve biological activity?

Proper handling procedures are critical for maintaining IFN-α 2a biological activity throughout research applications:

For lyophilized preparations:

  • Store at -20°C to -70°C in original sealed containers

  • Allow sealed vial to reach room temperature before opening to prevent moisture condensation

  • Reconstitute by gently directing buffer against the vial wall, not directly onto the protein cake

  • Allow 5-10 minutes for complete dissolution with gentle swirling (avoid vortexing)

For reconstituted/liquid preparations:

  • For initial reconstitution:

    • Use sterile distilled water or PBS

    • Prepare to a concentration of 63,000 International Units per mL when following standard protocols

  • For dilutions:

    • Use carrier protein when dilution is required

    • Initial dilutions (1:10, 1:100) should be made in:

      • Cell culture medium containing 5-10% calf serum, OR

      • PBS (pH 7.0-7.4) containing 0.3% bovine casein

  • Storage recommendations:

    • Prepare single-use aliquots to avoid freeze-thaw cycles

    • Short-term (1-7 days): 2-8°C

    • Long-term: -70°C or colder

    • Avoid repeated freeze-thaw cycles which significantly reduce activity

These handling practices typically ensure >90% recovery of the expected biological activity.

What receptor binding assays complement functional assays for IFN-α 2a characterization?

Receptor binding assays provide critical mechanistic insights that complement functional bioassays:

Surface plasmon resonance (SPR) binding assays:

  • Measures binding kinetics to isolated IFNAR1 and IFNAR2 receptor components

  • Provides association rate (kon), dissociation rate (koff), and equilibrium dissociation constant (KD)

  • Can detect subtle differences between IFN-α subtypes or variant preparations

  • Advantages: Direct quantification of receptor interaction without cellular context variables

Cell-based receptor binding assays:

  • Flow cytometry with fluorescently labeled IFN-α 2a

  • Competitive binding assays using labeled reference IFN-α

  • Quantifies binding to natural receptor complexes in cellular context

  • Advantages: Accounts for receptor organization in membrane and accessory proteins

JAK-STAT pathway activation assays:

  • Western blot or flow cytometry for STAT1/STAT2 phosphorylation

  • Quantification of ISG induction by RT-qPCR

  • Measures immediate downstream signaling events

  • Advantages: Confirms functional receptor engagement and signal transduction

When performing these assays, researchers should consider:

  • Using carrier-free IFN-α 2a preparations to avoid interference from carrier proteins

  • Including positive controls (reference standard) and negative controls (heat-inactivated samples)

  • Testing multiple cell types when possible, as receptor expression and organization vary by cell type

What are the optimal storage conditions for different IFN-α 2a preparations?

Storage requirements vary based on IFN-α 2a formulation state and intended duration:

For lyophilized IFN-α 2a:

  • Long-term storage: -20°C to -70°C in sealed, original containers

  • Protect from moisture, which can trigger hydrolysis reactions

  • Shield from light to prevent photo-oxidation of sensitive amino acids

For reconstituted/liquid IFN-α 2a:

  • Short-term (up to 7 days): 2-8°C with carrier protein (0.1-0.3% BSA)

  • Medium-term (1-3 months): -20°C in appropriate buffer

  • Long-term (>3 months): -70°C or lower in small single-use aliquots

  • Critical: Avoid repeated freeze-thaw cycles which significantly reduce activity

Buffer considerations for optimal stability:

  • pH 4.5-5.5 generally provides maximum stability

  • Include carrier protein (0.1-0.3% BSA) to prevent adsorption to container surfaces

  • For carrier-free preparations, use siliconized or low-protein-binding containers

Activity retention should be verified periodically using appropriate bioassays, as protein may remain physically stable while experiencing subtle conformational changes that reduce biological activity.

What methods can detect early signs of IFN-α 2a degradation?

Early detection of IFN-α 2a degradation requires a multi-method analytical approach:

Physical stability indicators:

  • Size exclusion chromatography (SEC): Detects aggregation and fragmentation

  • Dynamic light scattering (DLS): Sensitive to early-stage aggregation

  • UV-visible spectroscopy (260/280 nm ratio): Changes may indicate tertiary structure alterations

  • Intrinsic fluorescence spectroscopy: Shifts in emission maximum indicate conformational changes

Chemical stability analysis:

  • Reversed-phase HPLC: Detects oxidation and deamidation products

  • Isoelectric focusing (IEF): Identifies charge variants resulting from chemical modifications

  • Mass spectrometry: Provides detailed mapping of modifications (oxidation, deamidation)

Functional indicators:

  • Comparative dose-response in antiviral assays: Shifts in EC50 or maximum effect

  • Receptor binding kinetics: Changes in association or dissociation rates

  • JAK-STAT pathway activation: Reduced STAT phosphorylation efficiency

Accelerated stability protocols:

  • Exposure to elevated temperature (25-40°C) for defined periods

  • Mechanical stress testing (agitation, freeze-thaw cycles)

  • pH-stress studies (exposure to pH range 3-8)

Implementing a subset of these methods in routine quality monitoring provides early warning of stability issues before complete activity loss occurs.

How can researchers formulate IFN-α 2a for maximum stability in different experimental applications?

Optimal formulation strategies for IFN-α 2a depend on the specific experimental application:

For cell culture applications:

  • Base buffer: PBS or cell culture medium

  • Stabilizers: 0.1-0.3% BSA or 0.3% bovine casein

  • pH range: 7.0-7.4 (physiological)

  • Concentration: Prepare at 2-5× working concentration to allow for dilution in culture

  • Storage: Aliquot and store at -70°C; thaw immediately before use

For biochemical and binding assays:

  • Base buffer: 20-50 mM sodium phosphate or sodium acetate

  • pH range: 4.5-5.5 (stability optimum)

  • Ionic strength: 100-150 mM NaCl

  • Stabilizers: For carrier-free applications, use polysorbate 20 (0.005-0.01%) instead of protein carriers

  • Storage: Prepare fresh or use within 24 hours at 2-8°C

For long-term storage of research samples:

  • Base buffer: 20 mM sodium phosphate or sodium acetate

  • pH: 4.5-5.0

  • Stabilizers: 0.1% BSA + 5% trehalose or sucrose

  • Storage: -70°C in silicon-coated or low-binding tubes

  • Aliquot size: Single-use volumes to avoid freeze-thaw cycles

For in vivo experimental applications:

  • Base buffer: Sterile PBS

  • pH: 7.2-7.4

  • Purity requirement: Endotoxin ≤1 EU/μg

  • Filter sterilization: Use low protein-binding filters

  • Quality control: Test bioactivity before administration

These formulation strategies maximize stability while maintaining compatibility with downstream experimental applications.

How does the JAK-STAT pathway mediate IFN-α 2a cellular responses?

The JAK-STAT pathway transduces IFN-α 2a signals through a coordinated sequence of molecular events:

Receptor engagement and initiation:

  • IFN-α 2a binds to IFNAR2 with high affinity

  • The IFN-α 2a/IFNAR2 complex recruits IFNAR1, forming a ternary signaling complex

  • Receptor dimerization brings receptor-associated Janus kinases (JAK1 and TYK2) into proximity

  • JAKs undergo cross-phosphorylation and activate the cytoplasmic domains of the receptors

STAT protein recruitment and activation:

  • Phosphorylated receptor domains serve as docking sites for STAT proteins (primarily STAT1 and STAT2)

  • JAKs phosphorylate recruited STATs at specific tyrosine residues

  • Phosphorylated STATs form homo- and heterodimers

  • STAT1-STAT2 heterodimers recruit IRF9 to form the ISGF3 complex

Nuclear translocation and gene regulation:

  • Activated STAT complexes translocate to the nucleus

  • ISGF3 binds to Interferon-Stimulated Response Elements (ISREs)

  • STAT1 homodimers bind to Gamma-Activated Sequences (GAS)

  • Transcriptional activation of hundreds of Interferon-Stimulated Genes (ISGs)

Negative regulation mechanisms:

  • Induced SOCS (Suppressor of Cytokine Signaling) proteins provide negative feedback

  • Protein tyrosine phosphatases dephosphorylate activated pathway components

  • Protein inhibitors of activated STATs (PIAS) block STAT-DNA binding

This signaling cascade mediates the diverse biological effects of IFN-α 2a, including antiviral, anti-proliferative, and immunomodulatory activities through the coordinated expression of ISGs with varied cellular functions .

What experimental models best represent IFN-α 2a antiviral mechanisms?

Several experimental systems effectively model IFN-α 2a's antiviral effects for research applications:

Cell-based virus inhibition systems:

SystemKey FeaturesBest Applications
A549/EMCVHuman lung cells, RNA virus, EC50 ~1 U/mL Standard potency testing, respiratory virus research
MDBK/VSVBovine kidney cells, high sensitivity, EC50 ~5 U/mL Cross-species studies, historical standardization
Huh7/HCV repliconHuman hepatoma cells, no infectious virus neededHepatitis-specific mechanisms, clinically relevant

Molecular pathway analysis methods:

  • Quantitative RT-PCR arrays for ISG expression profiles

  • Western blot analysis of antiviral effector proteins (MX1, OAS1, PKR)

  • CRISPR/Cas9 knockout approaches to identify essential pathway components

Mechanistic research considerations:

  • Include time-course analyses to distinguish between early and late response genes

  • Compare responses across multiple cell types to identify tissue-specific variations

  • Use pathway inhibitors to dissect specific signaling branches

  • Include IFN-α neutralizing antibodies as specificity controls

Clinically relevant research models:

  • Hepatitis virus models (particularly relevant as IFN-α 2a is used in hepatitis treatment)

  • Cancer cell lines for anti-proliferative effects

  • Primary human leukocytes for immunomodulatory studies

These systems collectively provide comprehensive insights into the multifaceted antiviral mechanisms of IFN-α 2a, from receptor binding through effector functions.

How do different cell types vary in their response to IFN-α 2a?

Cell type-specific responses to IFN-α 2a result from the interplay of multiple regulatory factors:

Receptor expression variations:

  • Baseline IFNAR1/IFNAR2 expression differs significantly between cell types

  • Immune cells typically express higher receptor levels than non-immune cells

  • IFNAR1:IFNAR2 ratio varies by cell type, influencing signaling pathway preference

  • Receptor internalization and recycling kinetics differ between cell lineages

Signaling pathway component differences:

  • Varied expression levels of JAK-STAT pathway proteins across cell types

  • Cell-specific expression of negative regulators (SOCS, PIAS families)

  • Differential accessibility of ISG promoters based on epigenetic landscape

  • Pre-existing activation states of signaling components

Functional response patterns:

  • Leukocytes: Enhanced antigen presentation, increased cytotoxicity, cytokine production

  • Epithelial cells: Strong antiviral response, moderate anti-proliferative effects

  • Hepatocytes: Robust induction of both antiviral and metabolic gene programs

  • Pancreatic β-cells: Enhanced HLA-B-restricted antigen presentation

Research implications:

  • Cell type selection critically impacts experimental outcomes in IFN-α 2a research

  • Multiple cell types should be tested when developing IFN-α 2a-based therapeutics

  • Tissue-specific responses may explain differential efficacy and side effects in clinical applications

  • Comprehensive analysis requires examining both canonical (JAK-STAT) and non-canonical pathways

Understanding these cell-specific response patterns is essential for predicting therapeutic outcomes and developing targeted applications of IFN-α 2a in different disease contexts.

How can structure-function relationships in IFN-α 2a be systematically investigated?

Systematic investigation of IFN-α 2a structure-function relationships requires multi-disciplinary approaches:

Structural analysis techniques:

  • NMR spectroscopy: Provides high-resolution solution structure information

  • X-ray crystallography: Reveals atomic-level details of protein-receptor complexes

  • Hydrogen-deuterium exchange mass spectrometry: Maps dynamic regions and binding interfaces

Mutational analysis strategies:

  • Alanine scanning mutagenesis of surface residues

  • Conservative versus non-conservative substitutions at key positions

  • Creation of chimeric proteins between different IFN-α subtypes

  • Targeted modification of flexible loop regions (residues Gly44-Ala50, Ile100-Lys112)

Functional characterization assays:

  • Surface plasmon resonance (SPR) to measure binding kinetics to IFNAR1 and IFNAR2

  • Cell-based receptor assembly assays using FRET or BiFC

  • Pathway-specific reporter assays for different signaling branches

  • Antiviral versus anti-proliferative activity comparisons

Experimental design considerations:

  • Test multiple functional readouts for each structural modification

  • Include comparative analysis with other IFN-α subtypes

  • Examine species-specificity of structure-function relationships

  • Correlate biophysical measurements with biological activities

This integrated approach enables identification of structural determinants for specific functional properties of IFN-α 2a, potentially guiding development of variants with optimized biological profiles.

What methodologies can distinguish between the various biological activities of IFN-α 2a?

IFN-α 2a exhibits multiple distinct biological activities that can be differentiated through specialized assays:

Antiviral activity measurements:

  • Virus-specific protection assays (using different viruses: VSV, EMCV, HCV)

  • ISG expression profiling focused on antiviral effectors (MX1, OAS, PKR)

  • Viral replication inhibition in relevant target cells

Anti-proliferative activity assessments:

  • Growth inhibition assays in sensitive cell lines (Daudi, melanoma lines)

  • Cell cycle analysis by flow cytometry (G0/G1 arrest)

  • Apoptosis detection (Annexin V staining, caspase activation)

  • Colony formation assays for long-term effects

Immunomodulatory function evaluation:

  • Natural killer (NK) cell activation assays

  • Dendritic cell maturation and function tests

  • MHC class I upregulation measurements

  • Cytokine production profiling in immune cells

Comparative analysis approaches:

  • Activity ratio calculations between different functions

  • Dose-response comparisons across activity types

  • Time-course differences between rapid and delayed responses

  • Competition assays with other Type I IFNs

These methodologies help researchers distinguish the primary mechanisms underlying IFN-α 2a effects in different experimental and therapeutic contexts, allowing targeted applications that maximize beneficial activities while minimizing unwanted effects.

How can researchers optimize IFN-α 2a for specific research applications?

Researchers can optimize IFN-α 2a preparations for specific applications through targeted modifications and formulation strategies:

For enhanced stability:

  • Site-directed mutagenesis of oxidation-prone residues

  • Addition of stabilizing excipients (trehalose, human serum albumin)

  • Modified buffer formulations with antioxidants

  • Lyophilization with optimal cryoprotectants

For altered receptor specificity:

  • Targeted mutations at receptor binding interfaces

  • Creation of chimeric interferons with mixed subtype properties

  • Yeast surface display for directed evolution of binding properties

  • Computational design of modified binding surfaces

For controlled bioavailability:

  • PEGylation at specific residues

  • Fusion to half-life extending proteins (Fc, albumin)

  • Incorporation into controlled-release formulations

  • Targeted delivery systems for tissue-specific action

For specialized research applications:

  • Fluorescent or affinity tagging for tracking studies

  • Isotopic labeling for NMR or mass spectrometry studies

  • Creation of antagonist variants for pathway dissection

  • Development of reporter-linked constructs for live-cell imaging

Implementation considerations:

  • Verify retention of critical biological activities after modification

  • Compare stability profiles of modified versus native protein

  • Validate specificity using receptor competition assays

  • Assess immunogenicity of modified constructs for in vivo applications

These optimization approaches enable researchers to tailor IFN-α 2a properties for specific experimental requirements, enhancing research capabilities across diverse applications from basic mechanistic studies to translational research.

Product Science Overview

Structure and Production

IFN-Alpha 2a is a member of the type I interferon family, which includes several related proteins that share over 95% amino acid sequence homology . The recombinant form of IFN-Alpha 2a is typically produced in human embryonic kidney cells (HEK293) or E. coli . The protein has a predicted molecular mass of approximately 19 kDa and is often purified to a high degree of purity, exceeding 95% as determined by SDS-PAGE .

Mechanism of Action

The primary function of IFN-Alpha 2a is to bind to specific cell surface receptors, known as IFN-alpha receptors, which consist of two subunits: IFN-alpha R1 and IFN-alpha R2 . Upon binding to these receptors, IFN-Alpha 2a triggers a cascade of intracellular signaling pathways that lead to the expression of various antiviral and immunomodulatory genes. This results in the inhibition of viral replication and modulation of the immune response .

Therapeutic Applications

Recombinant IFN-Alpha 2a has been extensively used in the treatment of various viral infections and cancers. It has demonstrated efficacy in conditions such as chronic hepatitis B and C, hairy cell leukemia, and Kaposi’s sarcoma . The antiviral activity of IFN-Alpha 2a is measured using assays that evaluate its ability to inhibit the cytopathic effects of viruses on cultured cells .

Formulation and Stability

The recombinant IFN-Alpha 2a protein is typically formulated in phosphate-buffered saline (PBS) and may contain carrier proteins such as bovine serum albumin (BSA) to enhance stability . The protein is shipped at ambient temperature and should be stored at -20 to -70 °C to maintain its stability. It is important to avoid repeated freeze-thaw cycles to preserve its bioactivity .

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.