Recombinant Human Interferon alpha-2 protein (IFNA2) (Active)

Shipped with Ice Packs
In Stock

Description

IFNA2 is produced via recombinant DNA technology in diverse expression systems, each conferring distinct properties:

Expression SystemFormPuritySpecific ActivityEndotoxin Level
E. coliNon-glycosylated>97%5.5 × 10⁵ IU/mg≤0.1 EU/μg
YeastNon-glycosylated>98%Not specified<1 EU/μg
CHO/HEK293 cellsGlycosylated>95%1.00 × 10⁷ IU/mg<1 EU/μg

Data compiled from .

Glycosylated forms (e.g., mammalian cell-expressed IFNA2) exhibit enhanced stability and receptor affinity due to post-translational modifications like O-glycosylation at Thr106 . Non-glycosylated variants (e.g., E. coli-derived) retain bioactivity but require higher doses for equivalent efficacy .

Mechanism of Action

IFNA2 binds to the type I interferon receptor (IFNAR), a heterodimer of IFNAR1 and IFNAR2 subunits, initiating a JAK/STAT signaling cascade:

  1. Receptor Binding: IFNA2 binds IFNAR2 first (Kd ~nM), followed by IFNAR1 (Kd ~μM), forming a ternary complex .

  2. Signal Transduction:

    • Activation of JAK1 (IFNAR2-bound) and TYK2 (IFNAR1-bound).

    • Phosphorylation of STAT1 and STAT2.

    • Formation of ISGF3 (STAT1/STAT2/IRF9 complex), which translocates to the nucleus to induce interferon-stimulated genes (ISGs) .

  3. Biological Effects:

    • Antiviral: ISGs (e.g., MX1, OAS) inhibit viral replication.

    • Immunomodulatory: Upregulates MHC class I/II, promoting immune surveillance .

Clinical Applications

IFNA2 has been approved for treating multiple conditions, with evolving roles in combination therapies:

IndicationKey Findings
Chronic Hepatitis CCombined with ribavirin, IFNA2 achieves sustained virological response (SVR) rates of ~40–50%. PEGylated forms (e.g., PEG-IFNα2) improve SVR to ~60% .
Chronic Myeloid Leukemia (CML)Enhances major molecular response rates when combined with tyrosine kinase inhibitors (TKIs) .
Melanoma/RCCProlongs survival in metastatic stages, though efficacy is limited by toxicity .
COVID-19Anecdotal reports suggest antiviral potential, though not yet standardized .

Adverse Effects: Flu-like symptoms, myelosuppression, and neuropsychiatric issues (e.g., depression) necessitate dose adjustments .

Research and Development

Recent studies highlight innovative approaches to optimize IFNA2:

  • Plant-Based Production: Aloe vera engineered to express IFNA2 achieved antiviral activity (2,108 IU/mg in pulp extracts), demonstrating transgenic plant viability .

  • High-Throughput Screening: HEK-Blue™ IFN-α/β cells enable rapid detection of IFNA2 activity and inhibitor testing (e.g., anifrolumab targeting IFNAR1) .

  • PEGylation: Enhances half-life, enabling weekly dosing in combination therapies .

Biological Activity Parameters

Critical metrics for IFNA2 efficacy include:

ParameterValueAssay Method
Specific Activity1.00 × 10⁷ IU/mg (glycosylated)ISG induction in HEK-Blue™ cells
Endotoxin Purity<1 EU/μgLAL assay
Receptor AffinityIFNAR2: nM; IFNAR1: μM rangeSurface plasmon resonance

Data from .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered PBS, pH 7.4.
Form
Lyophilized powder
Lead Time
5-10 business days
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend that this vial be briefly centrifuged prior to opening to bring the contents to the bottom. Please reconstitute protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% of glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final concentration of glycerol is 50%. Customers can use this as a reference.
Shelf Life
The shelf life is influenced by multiple factors, including storage conditions, buffer ingredients, temperature, and the inherent stability of the protein. Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. The shelf life of the lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
IFN-alpha-2, LeIF A
Datasheet & Coa
Please contact us to get it.
Expression Region
24-188aa
Mol. Weight
19.2 kDa
Protein Length
Full Length of Mature Protein
Purity
>97% as determined by SDS-PAGE.
Research Area
Immunology
Source
Yeast
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function
Produced by macrophages, IFN-alpha exhibits antiviral activities.
Gene References Into Functions
  1. Genetic mutation IFNA2p.Ala120Thr diminishes the binding affinity of interferon alpha 2 to the interferon receptor. This weakened binding reduces phosphorylation of transcription factor STAT91, subsequently leading to lower expression of interferon-stimulated genes against hepatitis B virus. PMID: 28958921
  2. Elevated protein aggregation and diminished functionality of IFNA2a upon oxidation correlate with the site of modification identified through proteolysis-coupled mass spectrometry and localized structural alterations within the protein detected by 2D NMR. PMID: 30324266
  3. These experimental findings establish the retromer complex as a pivotal spatiotemporal regulator of IFNAR endosomal sorting and a novel factor influencing type-I IFN-induced JAK/STAT signaling and gene transcription. PMID: 27917878
  4. PVT1 interacts with STAT1 to inhibit IFN-alpha signaling and tumor cell proliferation. PMID: 29715456
  5. This investigation explored the molecular signals induced by ARI (azacytidine (A) and romidepsin (R), with IFNalpha2) treatment and found that this drug combination enhanced the accessibility to regulatory sequences of IFN-stimulated genes and IFN transcription regulatory factors that were epigenetically silenced in both colorectal cancer cells and Dendritic Cells. PMID: 28615266
  6. All the tested TLRAs elicited greater infant IFN-alpha2 production compared to newborn and adult blood. Conversely, CpG induced greater IFN-gamma, IL-1beta, IL-4, IL-12p40, IL-10 and CXCL8 in newborn blood compared to infant and adult blood. PMID: 27081760
  7. Interferon-alpha-induced CD100 expression on naïve CD8(+) T cells enhances antiviral responses to hepatitis C infection through CD72 signal transduction. PMID: 28222623
  8. Tumor-associated M2 macrophages in mycosis fungoides acquire immunomodulatory function upon exposure to IFN-alpha2a and IFN-gamma. PMID: 27342040
  9. Variant models of hIFNalpha-2b displayed structural and conformational changes suggesting that alterations to hIFNalpha-2b may be a risk factor, in addition to other known factors associated with the onset/progression of female breast carcinoma. PMID: 27403569
  10. Elevated serum IFNA2 is associated with adenoma. PMID: 25793917
  11. Data suggests IFNA2 binding to the extracellular domain of IFN receptors IFNAR1 or IFNAR2 promotes proximity between intracellular domains. Signaling is contingent upon the duration of activation and affinity of binding rather than specific conformational changes. PMID: 26679999
  12. This report details biased expression of human interferon alpha-2b and Escherichia coli methionine amino peptidase genes under the control of a single promoter in E. coli. PMID: 27087087
  13. Interferon alpha-2b gene mutations were identified amongst brain tumor patients. The highest percentage of frameshift mutations was observed. Patients were found to be under environmental stress from contaminated drinking water and local gamma radiations. PMID: 25837663
  14. PPARalpha activation by an agonist WY-14643 could potentiate IFN-alpha responses, reverse IFN-alpha refractoriness, and enhance viral eradication in hepatocytes. PMID: 25734487
  15. Modulates transitional B cell signaling and function in Systemic Lupus Erythematosus patients. PMID: 25678471
  16. Chronic exposure to low doses of radiations by occupational workers exhibits a significant correlation with mutational effects on the interferon alpha 2b gene, further evidenced by depressed interferon alpha levels in serum. PMID: 25768396
  17. IFN-beta proved significantly more effective than IFNA2 in protecting human head and neck squamous cell carcinoma lines from oncolysis by vesicular stomatitis. PMID: 25995245
  18. This review explores the structure, mechanism of action, and biological activities of IFNalpha2. PMID: 25982860
  19. This study established, for the first time in a large panel of pancreatic cancer cell lines, the effects of IFN-alpha/-beta on the expression of type-I IFN receptors. PMID: 24460759
  20. IFN-1ant may be a therapeutic candidate for the treatment of specific viral infections without inducing the immunomodulatory and antiproliferative functions of wild-type IFN. PMID: 24866020
  21. IFNalpha serum marker could be utilized to identify a group of rheumatoid Arthritis patients exhibiting increased disease activity, endothelial progenitor cell imbalance, an enhanced proinflammatory profile, and a higher cardiovascular risk. PMID: 24465874
  22. These findings establish crucial and essential roles for SKAR in regulating the mRNA translation of IFN-sensitive genes and the induction of IFN-alpha biological responses. PMID: 25049393
  23. Hepatitis B virus could induce both SOCS-1 and 3 expression irrespective of endogenous interferon levels. PMID: 24636575
  24. IL6 is an inducer of IRF9 expression in prostate cancer and a sensitizer for the antiproliferative effects of IFNalpha2. PMID: 23913484
  25. IFNA2 inhibits viral protein expression through PKR activation, leading to a decrease in viral protein synthesis. PMID: 24089560
  26. These studies identify IFNalpha originating from lymph nodes, rather than blood leukocytes, as a potential source of the IFN-I signature contributing to immune activation in HIV-1 infection. PMID: 23437155
  27. Pre-treatment waking hypothalamic-pituitary-adrenal (HPA) axis response is more pronounced in subjects transitioning to major depressive disorder during INFalpha2a treatment and may constitute a vulnerability factor in patients with hepatitis C virus infection. PMID: 22571879
  28. The association with clinical disease and the activation of multiple inflammatory cytokines supports a role for IFN-alpha2 in disease perpetuation in a substantial subset of systemic lupus erythematosis patients. PMID: 23213068
  29. Obese subjects exhibited a diminished capacity to produce IFN-alpha and IFN-beta in response to TLR ligands. This response was associated with increased basal levels of SOCS3 but not SOCS1. PMID: 22951153
  30. This analysis delves into how an N-acetylgalactosamine residue at threonine 106 modifies the dynamics and structure of interferon alpha2a around the glycosylation site. PMID: 23184955
  31. The authors concluded that IFN-alpha-induced inhibition of miR-122 may negatively affect the anti-hepatitis B virus function of IFN-alpha. PMID: 23055569
  32. Results indicated that missense mutations in transmembrane protein 2 p.Ser1254Asn, interferon alpha 2 p.Ala120Thr, its regulator NLR family member X1 p.Arg707Cys, and complement component 2 p.Glu318Asp were associated with chronic hepatitis B. PMID: 22610944
  33. Successful immunostimulation by IFNalpha2b may contribute to clinical improvement in tongue squamous cell carcinoma patients. PMID: 22885264
  34. Data indicate that the combination of AAV2.IL-4 and AAV2.IFN-alpha was not significantly different compared to AAV2.IL-4 alone. PMID: 22685550
  35. The crystal structures of two human type I IFN ternary signaling complexes containing IFNalpha2 and IFNomega reveal recognition modes and heterotrimeric architectures that are unique among the cytokine receptor superfamily but conserved between type I IFNs. PMID: 21854986
  36. Structural amino acid changes in the C-helix interacting with IFNAlphaR1 may alter signaling dynamics leading to elevated APOBEC3 and lower IDO by an engineered mutant derived from the amino-terminal region of IFNalpha21b and the COOH-terminus from IFNalpha2c. PMID: 21757613
  37. Interferon-stimulated genes exert their negative feedback action by capitalizing on the weakness of IFNalpha2 binding to the receptor. PMID: 22731491
  38. Protein kinase, DNA-activated catalytic polypeptide (PRKDC), was confirmed to play a role in MyD88-induced IFNA2 activation and IL-8 secretion. PMID: 22332739
  39. Variation at IFNA2 -173 and IFNA8 -884 conditions reduced IFN-alpha production and increased susceptibility to SMA and mortality. PMID: 22570109
  40. Data indicate that the production of IFN-alpha in the supernatant of transfected cells was approximately 3.15 ng/mL. PMID: 22482409
  41. IFNalpha rapidly down-regulates BCL-6 mRNA in purified mouse normal germinal center B cells. PMID: 22204827
  42. This study demonstrated that IFNalpha2, a type I interferon, increases the expression of TLR3 on human dermal fibroblasts. PMID: 21223583
  43. Interferon alpha 2 regulates MAPK and STAT1 pathways in human hepatoma cells. PMID: 21466707
  44. Degos disease is a distinct vascular injury syndrome where a dysregulated interferon-alpha response, in conjunction with membranolytic attack complex deposition, may contribute to the unique vascular changes. PMID: 21411783
  45. These findings indicate that the expression of MxA, 2',5'-OAS, and PKR is upregulated by the PI3K-AKT signal pathway, and the Raf-MEK-ERK signal pathway exerts a negative regulatory effect on the expression of MxA without a significant impact on 2',5'-OAS and PKR. PMID: 20309637
  46. Studies indicate that it remains unclear whether the interruption of IFNalphaA and IL-10 signaling in the absence of CD73 activity stems from a deficiency of its product adenosine or an accumulation of its substrate nucleotides. PMID: 21057730
  47. Data demonstrates that pDCs and type I IFNs promote inflammatory responses and wound healing in injured skin. PMID: 21115688
  48. The results suggest that hyper-induction of TNF-alpha in human macrophages is not consistently associated with a highly pathogenic phenotype of avian and human influenza viruses. PMID: 20532927
  49. Strategies are outlined to maximize the expression of correctly processed human INF alpha2b protein in Pichia. PMID: 20159042
  50. Data shows that serum concentrations of pegylated interferon alpha-2b increased in a dose-dependent manner. PMID: 19621225

Show More

Hide All

Database Links

HGNC: 5423

OMIM: 147562

KEGG: hsa:3440

STRING: 9606.ENSP00000369554

UniGene: Hs.211575

Protein Families
Alpha/beta interferon family
Subcellular Location
Secreted.

Q&A

How does Recombinant Human Interferon alpha-2 compare to other type I interferons?

IFNA2 was the first highly active IFN subtype to be cloned in the early 1980s and subsequently became the prototypic type I IFN used in fundamental research and clinical applications . Unlike IFNβ, which can be specifically produced in certain contexts, IFNA2 is typically produced alongside other IFNα subtypes rather than in isolation .

The specific activity of IFNA2, like other interferons, is determined by the stability of the IFN-receptor ternary complex, which depends on individual affinity to IFNAR1 and IFNAR2. Different type I IFN subtypes exhibit differential activities because the relationship between binding affinity and biological response varies among subtypes . For instance, the slope of the antiproliferative activity versus affinity is higher than the slope for anti-VSV (vesicular stomatitis virus) activity relative to receptor binding .

What is the molecular mechanism of IFNA2 signaling?

IFNA2 exerts its biological activity by assembling a ternary complex with the IFNAR1 and IFNAR2 receptor chains. The formation of this complex follows sequential binding where:

  • IFNA2 first binds to IFNAR2 with high affinity (nM range)

  • The IFNA2-IFNAR2 complex then recruits IFNAR1 with lower affinity (μM range)

  • The complete ternary complex initiates signal transduction

This binding activates associated JAK kinases, which phosphorylate STAT1 and STAT2 transcription factors. These form the ISGF3 transcription factor complex that translocates to the nucleus and induces transcription of interferon-stimulated genes (ISGs) .

The pathway can be represented as:
IFNA2 → IFNAR1/IFNAR2 complex → JAK kinase activation → STAT1/STAT2 phosphorylation → ISGF3 formation → ISG transcription

While the JAK/STAT pathway is the primary signaling mechanism in all cell types, IFNA2 can also activate other signaling factors in a cell type-dependent manner, resulting in diverse biological responses .

How does cell cycle phase affect cellular response to IFNA2 treatment?

Research indicates that the IFN-α/IFNAR2 axis specifically sensitizes cells to apoptosis during the S/G2/M phases of the cell cycle . This phase-specific sensitivity suggests that experimental timing of IFNA2 administration can significantly impact results, particularly in apoptosis studies or anti-cancer applications.

When designing experiments involving IFNA2-induced apoptosis, researchers should consider:

  • Cell synchronization protocols to enrich for S/G2/M phase cells

  • Time-lapse imaging to correlate cell cycle phase with apoptotic response

  • Cell cycle analysis in conjunction with apoptosis assays to confirm phase-specific effects

This phase-specific activity may explain some of the variable responses observed in different experimental systems and highlights the importance of considering cell cycle dynamics in IFNA2 research.

What are the optimal expression systems for producing bioactive Recombinant Human IFNA2?

The choice of expression system significantly impacts the structural characteristics and biological activity of recombinant IFNA2. Commonly used systems include:

Expression SystemAdvantagesLimitationsTypical Applications
E. coliHigh yield, cost-effective, simpler purificationLacks post-translational modifications, potential endotoxin contaminationBasic research, studies not requiring glycosylation
CHO cellsProper glycosylation, authentic 3D structure, low endotoxinLower yield, higher cost, complex purificationClinical research, studies requiring full biological activity
HEK293 cellsHuman-like post-translational modificationsSimilar limitations to CHOApplications requiring human-specific modifications

When selecting a recombinant IFNA2 preparation, researchers should consider the specific requirements of their experimental system and whether post-translational modifications are critical for the biological activity being studied.

What quality control parameters should researchers verify when using commercial Recombinant IFNA2?

Critical quality parameters to evaluate include:

  • Purity: Should be ≥95-98% as determined by SDS-PAGE and HPLC analysis

  • Endotoxin levels: Preferably ≤1 EU/μg to prevent non-specific immune activation that could confound experimental results

  • Biological activity: Verification using reporter cell lines such as HEK-Blue IFN-α/β cells that specifically respond to type I interferons

  • Protein concentration: Accurate determination using validated methods such as BCA assay or spectrophotometry

  • Storage conditions and stability: Typically stored at -80°C with minimal freeze-thaw cycles to preserve activity

Prior to experimental use, researchers should perform validation tests specific to their biological system, as activity can vary between different cell types and assay conditions.

How can researchers effectively design dose-response experiments using IFNA2?

Designing robust dose-response experiments with IFNA2 requires consideration of several factors:

  • Dose range selection: Based on the literature, typical effective concentrations range from 10 pM to 100 nM, with most cellular responses occurring in the 1-10 nM range. Include at least 6-8 concentrations spanning 3-4 log scales for accurate EC50 determination.

  • Time-course considerations: IFNA2 responses can be biphasic, with some genes induced rapidly (0.5-2 hours) and others showing delayed induction (12-24 hours). Design sampling timepoints accordingly.

  • Cell type variability: Sensitivity to IFNA2 varies widely among cell types due to differences in receptor expression levels and downstream signaling components. Include relevant positive and negative control cell lines.

  • Readout selection: Different biological activities (antiviral, antiproliferative, immunomodulatory) may have different dose-response relationships . Choose appropriate readouts for your specific research question.

  • Curve fitting: Use appropriate mathematical models (typically four-parameter logistic) for analysis, as IFNA2 dose-response curves can exhibit variable Hill slopes depending on the biological response measured.

When comparing different IFNA2 preparations or experimental conditions, it is essential to determine complete dose-response curves rather than using single concentrations, as relative potencies may vary depending on the concentration range examined.

What are the recommended methods for studying IFNA2 interaction with its receptors?

Several complementary approaches can be employed to study IFNA2-receptor interactions:

  • Surface Plasmon Resonance (SPR): Allows determination of binding kinetics (kon and koff) and equilibrium dissociation constants (KD). IFNA2 typically shows nanomolar affinity for IFNAR2 and micromolar affinity for IFNAR1 .

  • Bioluminescence Resonance Energy Transfer (BRET): Enables real-time monitoring of receptor interactions in living cells, revealing dynamics of ternary complex formation.

  • Fluorescence microscopy with labeled IFNA2: Allows visualization of receptor binding, internalization, and trafficking. Labeling should be performed at sites that don't interfere with receptor binding.

  • Mutagenesis studies: Systematic mutation of key residues in IFNA2 can identify critical interaction sites. Known interaction residues include those forming the 18 nm² binding interface with receptors .

  • Competitive binding assays: Using labeled IFNA2 and unlabeled competitors to compare binding affinities of different IFN subtypes or mutants.

When studying receptor interactions, it's important to account for the sequential binding mechanism where IFNA2 typically binds first to IFNAR2 and then to IFNAR1 in a bi-dimensional reaction to form the signaling-competent ternary complex .

How should researchers design experiments to study IFNA2's antiviral effects?

When investigating IFNA2's antiviral properties, consider the following methodological approaches:

  • Pre-treatment vs. post-infection protocols:

    • Pre-treatment (6-24 hours before infection) evaluates prophylactic potential

    • Post-infection treatment assesses therapeutic efficacy

    • Concurrent administration examines immediate interference with viral entry

  • Appropriate viral models:

    • Select viruses with known sensitivity to type I interferons

    • Include controls such as viruses with documented mechanisms of interferon evasion

    • Consider both RNA and DNA viruses to examine breadth of activity

  • Readout methods:

    • Viral titer determination (plaque assays, TCID50)

    • Viral protein expression (Western blot, flow cytometry)

    • Viral nucleic acid quantification (qPCR, RT-qPCR)

    • Reporter viruses expressing luciferase or fluorescent proteins

  • Mechanistic analysis:

    • Combine with analysis of interferon-stimulated gene (ISG) expression

    • Use receptor blocking antibodies to confirm specificity

    • Consider JAK inhibitors to verify signaling pathway dependence

A comprehensive experimental design should include dose-response relationships at different time points relative to infection, as the antiviral efficacy of IFNA2 is highly dependent on timing and concentration .

What is the role of IFNA2 in COVID-19 research and what experimental considerations are important?

Research suggests that deficiency of type I interferons (including IFNA2) in the blood may be a hallmark of severe COVID-19, providing rationale for therapeutic approaches . When designing COVID-19-related IFNA2 experiments, consider:

  • Timing of interferon response: SARS-CoV-2 can delay and antagonize early interferon responses, so time-course experiments are critical

  • Cell type selection:

    • Primary human airway epithelial cells (grown at air-liquid interface)

    • Alveolar epithelial cells

    • Immune cells (particularly plasmacytoid dendritic cells)

  • Relevant readouts:

    • Viral replication kinetics

    • Cell-intrinsic versus extrinsic effects

    • Inflammatory cytokine profiles

    • ACE2 expression modulation

  • Safety considerations: When working with SARS-CoV-2, appropriate biosafety level (BSL-3) facilities and protocols must be followed

  • Translational models: Ex vivo human lung tissue models or humanized mouse models may provide more relevant data than standard cell lines

When investigating IFNA2 in COVID-19 research, it's important to distinguish between prophylactic and therapeutic applications, as timing appears critical for clinical efficacy based on both basic research and clinical trials .

How can researchers differentiate between IFNA2-specific effects and general type I interferon responses?

Distinguishing IFNA2-specific effects from general type I interferon responses requires sophisticated experimental approaches:

  • Receptor subtype targeting:

    • Use blocking antibodies specific for different epitopes on IFNAR1 and IFNAR2

    • Apply CRISPR-Cas9 to generate receptor subunit variants with altered binding domains

    • Employ receptor mutants with selective defects in specific downstream signaling pathways

  • Comparative studies with multiple IFN subtypes:

    • Include other IFNα subtypes, IFNβ, and IFNω in parallel experiments

    • Use equipotent concentrations based on standardized bioassays rather than equal mass concentrations

    • Compare dose-response relationships for different biological activities

  • Signaling dynamics analysis:

    • Monitor temporal patterns of STAT phosphorylation

    • Examine nuclear translocation kinetics

    • Assess duration of activated signaling complexes

  • Transcriptomic analyses:

    • Compare gene induction profiles across different IFN subtypes

    • Analyze time-dependent changes in gene expression

    • Use bioinformatic approaches to identify IFNA2-specific gene signatures

Current evidence suggests that while most basic signaling mechanisms are shared among type I interferons, IFNA2 may exhibit unique kinetics, potency relationships across different bioactivities, and potentially tissue-specific effects .

What approaches are most effective for studying the immunomodulatory properties of IFNA2?

IFNA2's immunomodulatory functions can be studied using these methodological approaches:

  • Immune cell subset analysis:

    • Flow cytometry panels to assess effects on multiple immune populations

    • Consider both direct effects on target immune cells and indirect effects mediated by other responding cells

    • Include functional markers (activation, exhaustion, memory phenotypes)

  • Cytokine network analysis:

    • Multiplex cytokine assays to capture downstream mediators

    • Systems biology approaches to model cytokine networks

    • Single-cell secretion analysis to identify cellular sources

  • Functional immune assays:

    • Antigen presentation capacity

    • T cell proliferation and cytotoxicity

    • NK cell activation and cytotoxicity

    • Antibody production by B cells

  • In vivo models:

    • Humanized mouse models for studying human-specific responses

    • Conditional knockout systems to assess cell type-specific contributions

    • Reporter systems to track responding cell populations

  • Temporal considerations:

    • Acute versus chronic exposure models

    • Analysis of refractory states and tolerance development

    • Memory-like effects in innate immune cells

When studying immunomodulatory properties, researchers should recognize that IFNA2 effects are highly context-dependent, influenced by the microenvironment, concurrent stimuli, and the activation state of target cells .

What are common pitfalls in IFNA2 experimental design and how can they be overcome?

Researchers frequently encounter these challenges when working with IFNA2:

  • Loss of activity during storage/handling:

    • Store concentrated stock at -80°C in small aliquots to avoid freeze-thaw cycles

    • Add carrier protein (0.1% BSA or HSA) to dilute solutions to prevent adsorption to tubes

    • Validate activity after reconstitution using reporter cell lines

  • Variable cell responsiveness:

    • Screen cell lines for IFNAR1/IFNAR2 expression levels prior to experiments

    • Consider the impact of cell density and passage number on receptor expression

    • Include positive control cell lines with well-characterized responses

  • Specificity confirmation issues:

    • Include neutralizing antibodies against IFNA2 or receptor blocking antibodies as controls

    • Use JAK inhibitors to confirm signaling pathway dependence

    • Consider IFNAR knockout controls using CRISPR-Cas9

  • Reconciling contradictory results:

    • Document complete experimental conditions including cell source, medium composition, and serum concentration

    • Consider the impact of endogenous interferon production in your system

    • Account for potential species-specificity when comparing to literature data

  • Endotoxin contamination:

    • Verify endotoxin levels (<1 EU/μg) in commercial preparations

    • Include polymyxin B controls in sensitive experiments

    • Consider the use of endotoxin-resistant cell lines for comparative studies

Careful attention to these methodological considerations can significantly improve experimental reproducibility when working with IFNA2.

How should researchers address reproducibility challenges when comparing results across different IFNA2 preparations?

To ensure comparable results across different IFNA2 preparations:

  • Standardize activity measurements:

    • Establish internal reference standards with defined biological units

    • Perform parallel testing of new lots against reference standards

    • Use multiple bioassays to create activity profiles rather than single measurements

  • Document preparation characteristics:

    • Expression system (E. coli, CHO cells)

    • Purification method and purity assessment

    • Specific activity in standardized assays

    • Endotoxin levels and testing method

  • Consider formulation differences:

    • Presence of carrier proteins (HSA, BSA)

    • Buffer composition and pH

    • Preservatives or stabilizers

    • Glycosylation patterns if derived from different expression systems

  • Implement calibration protocols:

    • Design experiments with overlapping dose ranges when comparing preparations

    • Calculate relative potencies rather than absolute values

    • Use internal controls treated with a reference standard

  • Data normalization approaches:

    • Normalize to maximal response for each preparation

    • Use area under the curve (AUC) calculations for dose-response comparisons

    • Consider EC50 shifts as indicators of relative potency

By systematically addressing these variables, researchers can minimize reproducibility issues that arise from comparing results obtained with different IFNA2 preparations.

What are emerging techniques for studying IFNA2 biology at the single-cell level?

Single-cell technologies are revolutionizing our understanding of IFNA2 biology:

  • Single-cell RNA sequencing (scRNA-seq):

    • Reveals heterogeneity in cellular responses to IFNA2

    • Identifies previously unrecognized responding cell populations

    • Maps temporal dynamics of gene expression at single-cell resolution

  • Mass cytometry (CyTOF):

    • Simultaneously measures multiple signaling events and surface markers

    • Characterizes diverse phenotypic responses across cell types

    • Reveals signaling trajectories in complex cell populations

  • Live-cell imaging with fluorescent reporters:

    • Tracks real-time dynamics of IFNAR complex formation

    • Monitors single-cell signaling kinetics using STAT translocation reporters

    • Correlates signaling dynamics with cellular phenotypes

  • Spatial transcriptomics:

    • Maps interferon-stimulated gene expression in tissue context

    • Reveals local microenvironmental influences on IFNA2 responses

    • Identifies cellular niches with distinctive response patterns

  • Single-cell secretome analysis:

    • Characterizes secretory profiles of individual cells after IFNA2 stimulation

    • Links cellular phenotype to functional output

    • Identifies paracrine signaling networks

These emerging technologies will help resolve longstanding questions about cell-to-cell variability in IFNA2 responses and may identify specialized cell populations with unique functional roles in interferon biology.

How can computational approaches enhance our understanding of IFNA2 signaling networks?

Advanced computational methods are increasingly important for deciphering complex IFNA2 signaling networks:

  • Systems biology modeling:

    • Ordinary differential equation (ODE) models of JAK-STAT pathway dynamics

    • Boolean network models of downstream gene regulatory networks

    • Agent-based models of cell population responses

  • Network inference approaches:

    • Bayesian network analysis to infer causal relationships

    • Weighted gene co-expression network analysis (WGCNA)

    • Time-dependent network perturbation analysis

  • Machine learning applications:

    • Prediction of interferon-responsive elements in promoter regions

    • Classification of cell type-specific response patterns

    • Identification of biomarkers predictive of IFNA2 therapeutic response

  • Multi-omics integration:

    • Combined analysis of transcriptomics, proteomics, and phosphoproteomics data

    • Integration of epigenetic profiles with transcriptional responses

    • Correlation of metabolomic changes with functional outcomes

  • Molecular dynamics simulations:

    • Modeling IFNA2-receptor interactions at atomic resolution

    • Predicting effects of mutations on binding interface stability

    • Virtual screening for molecules modulating IFNA2-receptor interactions

These computational approaches can generate testable hypotheses about emergent properties of IFNA2 signaling networks that may not be apparent from reductionist experimental approaches alone.

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.