Recombinant Human Interferon alpha-1/13  (IFNA1)  (Active)

Shipped with Ice Packs
In Stock

Description

Mechanism of Action

IFNA1 activates through a dual-phase signaling mechanism:

  1. Receptor Binding: Binds IFNAR1 with 0.191 μM affinity (BLI assay)

  2. Downstream Effects:

    • Induces STAT1 phosphorylation

    • Upregulates antiviral effectors (GBP2, MDA5)

    • Modulates IRF3/IRF7 transcription factors

Key Regulatory Features:

  • Unique promoter architecture enables early response via IRF3 activation

  • Exhibits dose-dependent glutaminase induction (10-50 IU/mL)

  • Shows distinct signaling kinetics compared to IFNβ

Antiviral Therapeutics

  • Effective against herpes simplex virus (HSV) through TLR3-MyD88 pathway activation

  • Demonstrates HIV neuroprotection at 10 IU/mL

Neurodegenerative Disease Research

Model SystemKey FindingsSource
APP/PS1 mice (AD)2× increased prefrontal cortex IFNα vs WT
HIV-associated dementiaCSF levels correlate with cognitive decline
SOD1(G93A) ALSIFNAR deficiency slows disease progression

Autoimmunity Studies

  • Peg-IFNα-2a attenuates EAE severity (SJL/J mice)

  • Reduces myelin-specific CD4+ T cell responses

Comparative Bioactivity

Assay TypeResponse ThresholdMax Effect Concentration
GBP2 Expression0.1 ng/mL1 ng/mL
MDA5 Upregulation10 ng/μLN/A
STAT1 Phosphorylation10 IU/mL50 IU/mL

Production Considerations

  • Expression Systems:

    • E. coli: Higher yield (mg/L scale)

    • Mammalian Cells: Proper glycosylation

  • Formulation:

    • Lyophilized with 3% mannitol/5% trehalose

    • Requires PBS (pH 7.4) reconstitution

Clinical Relevance

  • Shows paradoxical effects: Neuroprotective in MS models vs neurotoxic in HIV dementia

  • Phase-specific activity in Alzheimer's progression

  • Potential therapeutic window: 0.1-50 IU/mL range

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered PBS, pH 7.4, containing 3% Mannitol, 5% Trehalose, 0.05% Tween-80.
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 the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% 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 various factors such as storage conditions, buffer ingredients, storage temperature, and the protein's intrinsic stability.
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 use. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
IFN-alpha-1/13, LeIF D
Datasheet & Coa
Please contact us to get it.
Expression Region
24-189aa
Mol. Weight
19.5 kDa
Protein Length
Full Length of Mature Protein
Purity
>97% as determined by SDS-PAGE.
Research Area
Immunology
Source
E.coli
Species
Homo sapiens (Human)
Target Names
IFNA1;
Uniprot No.

Target Background

Function
Interferon-alpha (IFN-alpha), produced by macrophages, exhibits antiviral activities. It stimulates the production of two key enzymes: a protein kinase and an oligoadenylate synthetase.
Gene References Into Functions
  1. SOCS2 impairs IFN/JAK/STAT signaling by reducing the stability of tyrosine kinase 2 (TYK2), downregulating the expression of type I and III IFN receptors, attenuating the phosphorylation and nuclear translocation of STAT1. PMID: 28496097
  2. Significant associations were observed for 4 variants in IFNAR2, IFNLR1 with hepatitis B virus infection, and IFNLR1-rs4649203 was associated with hepatitis B recovery. Moreover, the authors demonstrated the clear relevance of 5 polymorphisms in IFNA1, IFNA2, IFNL4 with hepatocellular carcinoma. PMID: 29080269
  3. HIV-1 IIIB infection of myeloid ThP-1 cells also reduced the IFN-alpha-mediated induction of the anti-viral gene, ISG15, but not MxA, revealing a functional consequence of this HIV-1-mediated immune evasion strategy. PMID: 29580840
  4. Therefore, these results demonstrate the importance of MxB in alpha interferon-mediated inhibition of HIV-1 infection. PMID: 29925663
  5. Our results illustrate a novel regulatory role of TWEAK, in which its activity positively regulates type I IFN pathway in lupus nephritis (LN) based on preclinical models. Our findings suggest TWEAK could act as a critical target in preventing renal damage in patients with LN. PMID: 29333443
  6. Myeloid cells contribute more to the whole blood interferon signature in systemic lupus erythematosus than their lymphocytic counterpart. Very similar leukocyte subsets reveal distinctive IFN signatures. IFN alpha mixes up composition of blood and leads to a preferential neutropenia, yielding relative lymphocytosis. PMID: 28357476
  7. the polymorphic variant of IFNA1 (-2) gene is associated with chronic HBV infection PMID: 27101083
  8. Cerebrospinal fluid interferon alpha levels correlate with neurocognitive impairment in ambulatory HIV-Infected individuals. PMID: 27400930
  9. Data suggest a central role of XBP1 in TLR7-induced IFNalpha production and identify XBP1 as a potential novel therapeutic target in IFNalpha-driven autoimmune and inflammatory diseases. PMID: 28408069
  10. Study shows IFN-alpha rapidly induces a profound shift in whole brain network structure, impairing global functional connectivity and the efficiency of parallel information exchange. PMID: 26697999
  11. The review focuses on the value of the type I and III interferon subtypes (alphas, beta and lambdas) as therapeutics for prevention and treatment of viral infections (influenza, herpes, human immunodeficiency virus and hepatitis viruses). PMID: 27544015
  12. MiR-181a is an important mediator for interferons-induced SAMHD1 expression in astrocytes and microglia, but not for inhibition of HIV-1 infection induced by IFN-alpha. PMID: 27219130
  13. the expression of certain TAM components was reduced as a result of prolonged degradation of MYD88 by Porphyromonas gingivalis infection. PMID: 28076786
  14. Type I interferons (IFNs) signature is seen in a significant proportion of anti-nuclear antibody-positive (ANA(+) individuals and appears to be associated with ANA titre and type of autoantibodies, rather than with the presence or development of clinical systemic autoimmune rheumatic diseases (SARDs) symptoms. PMID: 28245862
  15. study found that endogenous IFNalpha autocrinally promotes the expression of Interferon-Stimulated Gene (ISG) mRNAs in IL-3-, but not in IFNlambda3 plus IL-3-, treated plasmacytoid dendritic cells (pDCs); production of IFNalpha by IFNlambda3 plus IL-3-treated pDCs is mostly dependent on endogenously produced TNFalpha PMID: 27513213
  16. results demonstrate that Sphingosine 1-phosphate lyase (SPL) is a host factor that augments type I IFN responses during influenza A virus infection; study delineates the relationship between IKKepsilon and SPL, which provides a mechanistic understanding of the pro-IFN activity of SPL PMID: 28600291
  17. These data suggest that plasmacytoid dendritic cells producing IFN-alpha and IL-33 play a pivotal role in the chronic fibro-inflammatory responses underlying murine autoimmune pancreatitis and human IgG4-related autoimmune pancreatitis. PMID: 28373582
  18. These findings also identify STAT3 as a therapeutic target against viral infection and highlight it as an essential pathway component for endogenous and therapeutic IFN-alpha responsiveness. PMID: 27988795
  19. Mothers of children affected by autoimmune congenital heart block had a significantly higher expression IFN-alpha. PMID: 28501799
  20. Suppression of the exogenous Type I IFN-induced Jak/STAT signaling by NSs might be one of the mechanisms of Severe fever with thrombocytopenia syndrome (SFTS) to evade host immune surveillance. PMID: 28234991
  21. the paper shows the data on the role of interferons -alpha and -beta in infections - not only commonly known viral infections, but also bacterial, fungal and parasitic. PMID: 28388697
  22. High Type I Interferon expression due to hypomethylation is associated with Systemic Lupus Erythematosus. PMID: 28085900
  23. these studies identify phosphorylation of S734-STAT2 as a new regulatory mechanism that negatively controls the type I IFN-antiviral response. PMID: 27802159
  24. The synthetic IFNP molecule exerted its antitumor activity by upregulating the downstream genes involved in the STAT1 pathway and in apoptosis. Using a cell receptor binding assay, we showed that this Jurkat-binding peptide facilitated the binding affinity of IFNalpha to the cell surface type I IFN receptor PMID: 28578326
  25. KSHV-encoded viral IRF4 interacts with the host IRF7 and inhibits interferon-alpha production. PMID: 28342865
  26. Data indicate ARID3a(+) B cells as a type of effector B cell, and link ARID3a expression in B lymphocytes to interferon alpha (IFNa)-associated inflammatory responses in systemic lupus erythematosus (SLE). PMID: 27522115
  27. On one hand, hepatitis B virus activates MMP-9 in infected patients and leukocytes. On the other hand, MMP-9 facilitates hepatitis B virus replication through repressing IFN/JAK/STAT signaling, IFNAR1 function, and IFN-alpha action. PMID: 28122987
  28. Interferon-alpha Induces the apoptosis of Hela cells by activating both the intrinsic mitochondrial pathway and endoplasmic reticulum stress-induced pathway. PMID: 27827850
  29. Decreased interferon alpha (IFN-alpha) secretion induced by Toll-like receptor 7 and Toll-like receptor 8 (TLR7/TLR8) activation was observed in common variable immunodeficiency (CVID), which was recovered with Toll-like receptor 9 (TLR9) signaling. PMID: 27392462
  30. Plasmacytoid dendritic cells (pDCs) and type 1 interferon (FNalpha/beta)frequently accompanied plasmablast/plasma cells (PB/PCs). PMID: 27102764
  31. These results establish that activation of STAT pathway is essential for anti-hepatitis c virus efficacy of IFN-alpha. PMID: 27855377
  32. IL-2 therapy should be used as a first- or second-line therapy following IFN-a therapy. IL-2 may have a lower response if it is used after molecular-targeted therapy or other treatments PMID: 27630356
  33. results show that IFNA1 rs1332190 and IFNA17 rs9298814 SNPs may play an important role in Crimean-Congo hemorrhagic fever susceptibility PMID: 26694082
  34. Interferon-alpha-induced TRIM22 interrupts hepatitis c virus replication by ubiquitinating NS5A. PMID: 25683609
  35. our data indicate that by targeting PTP1B, miR-744 plays a feed-forward role in regulating type I IFN signaling pathway. PMID: 26259828
  36. the findings demonstrate that treatment with anti-CD20-hIFNalpha reverses resistance of B-NHL PMID: 26398317
  37. NS of severe fever with thrombocytopenia syndrome virus inhibited the activity of IFN-alpha1, IFN-beta, IFN-lambda1 and IFN-lambda2 through inhibition of STAT1 phosphorylation. PMID: 26353965
  38. interferon (IFN)-alpha, weakens activation of the anti-bacterial interleukin (IL)-1/IL-22 axis in human peripheral blood mononuclear cells exposed to viable B. burgdorferi PMID: 26152778
  39. Shp-2 contributes to the control of respiratory syncytial virus replication and progeny production in pulmonary alveolar epithelial cells by interfering with IFN-alpha-induced Jak/Stat1 pathway activation PMID: 26119280
  40. Interferon alfa induces high response rates with low toxicity in patients with polycythemia vera. PMID: 26261238
  41. This study suggests that suppression of miR-122 induced by hepatitis B virus infection, leads to the inactivation of interferon expression, which in turn enhances hepatitis B virus replication. PMID: 25766860
  42. Data suggest that Viral microRNA hcmv-miR-UL112 subverts innate immunity by downregulating type I interferons IFN-alpha and IFN-beta signaling to inhibit natural killer (NK) cell cytotoxicity. PMID: 25530545
  43. Interferon-alpha and interferon-beta are at the crossroads of allergy and viral infections. (Review) PMID: 26026068
  44. The interaction between AIP and IRF7 is enhanced upon virus infection, and AIP potently inhibits IRF7-induced type I IFN (IFN-alpha/beta) production. PMID: 25911105
  45. Interferon-alpha inhibits CD4 T cell responses to interleukin-7 and interleukin-2 and selectively interferes with Akt signaling. PMID: 25784743
  46. Data show that chemokine CXCL13 production by monocytes required toll-like receptor 7 activation and secretion of interferon-alpha. PMID: 25667414
  47. Studies indicate that type I interfereon signaling cascade is initiated by the binding of the cytokines to the high-affinity interferon alpha receotor. PMID: 25747907
  48. Results show that histone-lysine N-methyltransferase Set7 facilitates hepatitis C virus (HCV) replication through the attenuation of interferon-alpha (IFN-alpha) signaling pathways and IFN-related effectors. PMID: 25681344
  49. Domain 2 of the hepatitis C virus core protein critically affected the magnitude of host IFN-alpha responses. PMID: 25552725
  50. Hepatitis C virus NS3-4A similarly diminished both human and mouse MAVS-dependent signaling in human and mouse cells and MAVS induces both type I and type III interferons, which together control the hepatitis C virus replication. PMID: 25609814

Show More

Hide All

Database Links

HGNC: 5417

OMIM: 147578

KEGG: hsa:3439

STRING: 9606.ENSP00000276927

UniGene: Hs.37026

Protein Families
Alpha/beta interferon family
Subcellular Location
Secreted.

Q&A

Basic Research Questions

  • What is Recombinant Human Interferon alpha-1/13 (IFNA1) and how is it produced?

    Recombinant Human Interferon alpha-1/13 (IFNA1) is a type I interferon cytokine that belongs to the alpha/beta interferon family. It is primarily produced by macrophages and possesses significant antiviral activities .

    The recombinant protein is typically expressed in either bacterial systems (E. coli) or mammalian cells (HEK 293). The choice of expression system affects post-translational modifications:

    Expression SystemMolecular WeightPost-translational ModificationsPurity StandardsCommon Applications
    E. coli19.5 kDaMinimal>96% by SDS-PAGEIn vitro studies
    HEK 293Varies (24-189 aa range)Glycosylation present>95% purityIn vivo studies

    Production typically involves gene cloning, expression in the chosen system, followed by purification using chromatography techniques. Quality control includes tests for endotoxin levels (<1 EU/μg), purity verification by SDS-PAGE/HPLC, and biological activity assessment .

  • How does IFNA1's activity compare to other interferon alpha subtypes?

    IFNA1 has distinct characteristics that differentiate it from other interferon alpha subtypes:

    • Receptor binding: IFNA1 has approximately 100-fold lower affinity for IFNAR2 compared to most other IFN-α subtypes, while maintaining relatively high affinity for IFNAR1

    • Genetic conservation: IFNA1 shows lower polymorphism frequency in human populations, suggesting evolutionary importance

    • Transcriptional regulation: Unlike most other IFN-α subtypes, IFNA1 can be induced by IRF3 activation alone, without requiring IRF7

    • Genetic redundancy: Humans have two genes (IFNA1 and IFNA13) that express identical IFNα1 proteins, suggesting important evolutionary conservation

    Key substitutions in the protein sequence affecting receptor binding include:

    • F27S: Decreases affinity for IFNAR2 by 4-fold

    • R22S: Together with S27, decreases affinity by ~14-fold

    • K31M: May contribute to decreased IFNAR2 affinity by disrupting helix structure

  • What are the primary biological functions of IFNA1?

    IFNA1 exhibits several key biological functions critical to immune responses:

    1. Antiviral activity: Stimulates production of protein kinase and oligoadenylate synthetase enzymes that inhibit viral replication

    2. Immunomodulatory effects:

      • Activation of natural killer (NK) cells via STAT1 pathway

      • Regulation of monocyte and neutrophil recruitment during viral pneumonia

      • Modulation of antigen presentation pathways

    3. Cell signaling: Triggers JAK/STAT signaling pathways that induce interferon-stimulated genes (ISGs)

    4. Regulatory functions:

      • Balances immune activation and suppression during viral infections

      • May restrict type 2 immunopathology through regulation of group 2 innate lymphoid cells

    The biological activities of IFNA1 are concentration-dependent, with specific activity in antiviral assays typically measured at no less than 1.0×10^8 IU/mg .

Advanced Research Applications

  • What signaling pathways are activated by IFNA1 and how do they differ from other interferons?

    IFNA1 triggers complex signaling cascades primarily through the JAK/STAT pathway:

    1. Receptor binding: IFNA1 binds to the heterodimeric receptor complex composed of IFNAR1 and IFNAR2, with uniquely higher relative affinity for IFNAR1 than other IFN-α subtypes

    2. JAK activation: Receptor engagement activates Janus kinases (TYK2 and JAK1) associated with the cytoplasmic domains of IFNAR1 and IFNAR2

    3. STAT phosphorylation: Activated JAKs phosphorylate STAT proteins, primarily STAT1 and STAT2, forming:

      • STAT1-STAT2 heterodimers

      • STAT1 homodimers

      • STAT1-STAT3 heterodimers

    4. Transcription factor complex formation: Phosphorylated STAT complexes associate with IRF9 to form ISGF3 complexes

    5. Nuclear translocation: These complexes translocate to the nucleus where they bind to specific DNA sequences:

      • ISRE (Interferon-Stimulated Response Elements)

      • GAS (Gamma-Activated Sequences)

    6. Gene induction: This leads to expression of hundreds of ISGs with antiviral, antiproliferative, and immunomodulatory functions

    Unique to IFNA1 is its distinct activation pattern of STAT proteins compared to other subtypes, leading to differential gene expression profiles in human T cells and dendritic cells .

  • How can researchers accurately measure IFNA1 activity in experimental settings?

    Several complementary approaches can be employed to measure IFNA1 activity:

    1. Antiviral assays:

      • Human replicon assays using Huh-7 cells containing HCV replicons

      • Cytopathic effect reduction assays using susceptible cell lines

      • These typically measure EC50 values and can determine specific activity (IU/mg)

    2. Reporter gene assays:

      • Cells transfected with ISRE-luciferase reporters

      • Measures transcriptional activation of interferon-responsive elements

      • Allows high-throughput screening with quantitative readout

    3. ELISA-based detection:

      • Sandwich ELISA for protein quantification (detection range: 7.8-500pg/mL)

      • Sensitivity typically around 1.95pg/mL

      • Measures concentration but not biological activity

    4. Receptor binding assays:

      • Surface plasmon resonance to measure binding kinetics to IFNAR1/IFNAR2

      • Can determine association/dissociation constants (Ka/Kd)

    5. Phospho-STAT detection:

      • Western blotting or flow cytometry for pSTAT1/pSTAT2

      • Directly measures initial signaling events

      • Can be performed in target cells of interest

    Standard curves using reference material with known potency should be included. Intra- and inter-assay variability should be monitored (typically <4% and <7% respectively for validated assays) .

  • What experimental controls should be included when studying IFNA1 effects?

    Robust experimental design for IFNA1 research should include:

    1. Positive controls:

      • Commercial recombinant IFNA1 with established activity (e.g., European Pharmacopoeia standard)

      • Other well-characterized type I interferons (IFN-β) for comparative analysis

    2. Negative controls:

      • Heat-inactivated IFNA1 (95°C for 5 minutes)

      • Buffer-only treatment

      • Isotype-matched irrelevant protein

    3. Receptor blockade controls:

      • Anti-IFNAR1/IFNAR2 neutralizing antibodies

      • JAK inhibitors (e.g., Ruxolitinib)

      • Demonstrates specificity of observed effects

    4. Dose-response relationships:

      • Minimum 5-point titration covering 2-3 logs

      • Typically ranging from 0.1-100 ng/mL

      • Establishes EC50/IC50 values

    5. Time-course experiments:

      • Early timepoints (15 min-2 hr) for signaling events

      • Intermediate timepoints (2-12 hr) for gene expression

      • Late timepoints (12-72 hr) for phenotypic changes

    6. Cell-type specific validation:

      • Test effects on multiple relevant cell types (T cells, dendritic cells, etc.)

      • Account for differential responses between cell types

    Including these controls ensures that observed effects are specific to IFNA1 activity and not due to experimental artifacts or contamination.

  • How does IFNA1 contribute to antiviral immunity at the molecular level?

    IFNA1 establishes an antiviral state through multiple complementary mechanisms:

    1. Induction of restriction factors:

      • Protein kinase R (PKR): Phosphorylates eIF2α, inhibiting viral protein synthesis

      • 2'-5'-oligoadenylate synthetase (OAS): Activates RNase L, degrading viral RNA

      • Mx proteins: GTPases that trap viral ribonucleoproteins

      • IFITM proteins: Block viral cell entry and membrane fusion

    2. Cell-intrinsic immunity enhancement:

      • Upregulation of pattern recognition receptors (PRRs)

      • Amplification of downstream signaling pathways

      • Creates positive feedback loop for sustained antiviral response

    3. Regulation of cell death pathways:

      • Sensitization to apoptotic signals in infected cells

      • Upregulation of pro-apoptotic factors

      • Elimination of viral reservoirs

    4. Coordination of adaptive immunity:

      • Enhanced antigen presentation via MHC class I upregulation

      • Promotion of memory T cell generation and maintenance

      • B cell activation and antibody production

    5. Modification of cellular metabolism:

      • Shifts from oxidative phosphorylation to glycolysis

      • Alters lipid metabolism to create unfavorable environment for viral replication

    The kinetics of IFNA1 responses follow distinct patterns, with early effects on cell-intrinsic immunity followed by later coordination of adaptive responses through transcriptional and post-transcriptional regulation .

  • What are the key methodological considerations for IFNA1 stability in laboratory experiments?

    Maintaining IFNA1 stability is critical for experimental reproducibility:

    1. Storage conditions:

      • Lyophilized: Store at -20°C to -80°C

      • Reconstituted: Add glycerol (final 30-50%) and store at -20°C or -80°C

      • Working aliquots can be stored at 4°C for up to one week

    2. Reconstitution protocols:

      • Use sterile, deionized water or manufacturer-recommended buffer

      • For lyophilized preparations, centrifuge vial before opening

      • Reconstitute to 0.1-1.0 mg/mL concentration

      • Avoid vigorous shaking or vortexing that may cause denaturation

    3. Buffer considerations:

      • Optimal pH range: 7.0-7.4

      • Common formulation: PBS with 4% mannitol and 1% HSA

      • Carrier proteins (HSA, BSA) prevent adsorption to surfaces

    4. Avoiding freeze-thaw cycles:

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

      • Each freeze-thaw cycle can reduce activity by 5-15%

      • Maximum recommended cycles: 3

    5. Activity monitoring:

      • Periodic testing in bioassays to confirm retained activity

      • Circular dichroism can be used to verify intact secondary structure

      • SDS-PAGE to check for aggregation or degradation

    6. Protein denaturation prevention:

      • Avoid exposure to extreme temperatures

      • Minimize exposure to air/oxidation

      • Use low-binding tubes and pipette tips

      • Keep away from strong light exposure

    Proper handling and storage procedures are essential for maintaining IFNA1 biological activity throughout the experimental timeline.

  • How can pharmacokinetic-pharmacodynamic (PK-PD) modeling be applied to IFNA1 research?

    PK-PD modeling provides quantitative frameworks for understanding IFNA1 behavior in experimental systems:

    1. Key PK parameters to measure:

      • Volume of distribution (Vd)

      • Clearance rate (CL)

      • Half-life (t½)

      • Area under the curve (AUC)

      • Maximum concentration (Cmax)

    2. PD biomarkers for IFNA1 activity:

      • Phosphorylation of STAT proteins

      • Induction of interferon-stimulated genes (MX1, OAS1, etc.)

      • Antiviral activity in cellular or animal models

      • Changes in immune cell activation markers

    3. Modeling approaches:

      • Direct effect models (linking concentration to response)

      • Indirect effect models (incorporating production/degradation rates)

      • Tolerance/rebound models (accounting for adaptation)

      • Signal transduction models (capturing complex signaling networks)

    4. Implementation example from literature:
      In a study with TLR-7 agonists that induce IFNA1, researchers used:

      • A biophase distribution model to account for absorption

      • A target-mediated drug disposition model for interferon induction

      • An effect compartment to link interferon levels to antiviral activity

    5. Software tools for implementation:

      • NONMEM, Phoenix WinNonlin, or similar PK-PD modeling software

      • R packages (nlme, PKPDmodels) for statistical analysis

      • SimBiology (MATLAB) for mechanistic systems models

    This approach allows prediction of dosing regimens, understanding of inter-individual variability, and translation between experimental models and clinical applications.

Clinical Research Applications

  • What is the therapeutic potential of IFNA1 compared to other interferon formulations?

    IFNA1 has distinct properties relevant to its therapeutic applications:

    1. Comparative receptor binding profile:

      • Lower affinity for IFNAR2 but higher relative affinity for IFNAR1 compared to other subtypes

      • May result in different signaling cascade activation patterns

      • Potentially different side effect profiles due to differential ISG induction

    2. Applications in viral infections:

      • Hepatitis B and C virus infections: Established efficacy of type I interferons

      • COVID-19: Potential to normalize dysregulated innate immunity

      • Other viral infections: Human papillomavirus, Kaposi's sarcoma associated with HIV

    3. Administration routes and formulations:

      • Inhalation/nebulization: Commonly used for respiratory infections like COVID-19

      • Subcutaneous: Standard for systemic delivery

      • Pegylated formulations: Extended half-life

    4. Comparative efficacy data:

      Interferon TypeCommon ApplicationsRelative PotencyHalf-lifeMain Advantages
      IFNA1Viral infectionsVariable by subtypeShortSpecific receptor binding profile
      Pegylated IFN-αHepatitis B/CHighExtendedLonger duration of action
      IFN-βMultiple sclerosisDifferent ISG profileIntermediateAuto-immune regulation
    5. Safety considerations:

      • Inhalation of IFNA1 showed no association with delayed-phase thrombocytopenia

      • May have hepatoprotective effects during COVID-19 infection

      • Lower risk of elevated alanine aminotransferase in patients receiving inhaled IFNA1

    The therapeutic application of IFNA1 requires balancing its antiviral efficacy with potential immunomodulatory effects, considering the context-specific benefits and risks.

  • How can researchers investigate IFNA1 resistance mechanisms?

    Investigating IFNA1 resistance requires multi-level analytical approaches:

    1. Receptor expression and signaling analysis:

      • Flow cytometry for IFNAR1/IFNAR2 surface expression

      • Phospho-flow for STAT1/STAT2 activation

      • Western blot for total and phosphorylated signaling proteins

      • RNA-seq for JAK-STAT pathway component expression

    2. Genetic analysis techniques:

      • Targeted sequencing of IFNAR1/IFNAR2 and downstream effectors

      • CRISPR-Cas9 screening to identify resistance mediators

      • Single-cell analysis to identify resistant subpopulations

    3. Assessment of viral evasion strategies:

      • Viral protein expression that antagonizes IFNA1 signaling

      • Measuring viral protein interactions with IFNAR components

      • Evaluation of viral mutations correlating with treatment failure

    4. Auto-antibody screening:

      • ELISA to detect anti-IFNA1 autoantibodies

      • Neutralization assays to assess functional impact

      • Particularly important in diseases like COVID-19 where autoantibodies against type I IFNs correlate with disease severity

    5. Epigenetic regulation analysis:

      • Chromatin immunoprecipitation (ChIP) for histone modifications at ISG promoters

      • Methylation analysis of CpG islands in IFN pathway genes

      • ATAC-seq to assess chromatin accessibility changes

    6. Longitudinal monitoring protocols:

      • Serial sampling before, during, and after IFNA1 treatment

      • Correlation of molecular markers with clinical/experimental outcomes

      • Development of predictive biomarkers for resistance

    This comprehensive approach helps differentiate between intrinsic resistance mechanisms, acquired resistance, and viral evasion strategies.

  • What methodologies are most appropriate for studying IFNA1's role in autoimmune conditions?

    Investigating IFNA1 in autoimmune contexts requires specialized methodologies:

    1. Quantification of IFNA1 in patient samples:

      • Digital ELISA (Simoa) for ultrasensitive detection

      • IFNA1-specific assays to distinguish from other subtypes

      • Standardization against international reference materials

    2. Gene expression profiling:

      • IFN signature scoring using validated gene sets

      • NanoString technology for targeted quantification

      • Single-cell RNA-seq to identify cellular sources and responders

    3. Functional assays:

      • Reporter cells expressing ISRE-luciferase constructs

      • Ex vivo stimulation of patient PBMCs

      • Serum/plasma transfer experiments

    4. Animal models:

      • Conditional knockout of IFNAR in specific cell types

      • Inducible expression systems for temporal control

      • Humanized mice expressing human IFNAR components

    5. Therapeutic intervention studies:

      • IFNAR-blocking antibodies

      • JAK inhibitors as downstream signaling blockers

      • Correlation of IFNA1 levels with treatment responses

    6. Genetic association studies:

      • SNP analysis in IFNA1 and pathway genes

      • Correlation with disease severity and progression

      • Functional validation of identified variants

    These approaches are particularly relevant for studying conditions like systemic lupus erythematosus (SLE), which has a high type I IFN signature, while considering the dual nature of IFNs in promoting or ameliorating autoimmunity in different contexts.

  • How can researchers distinguish between direct and indirect effects of IFNA1 treatment in complex biological systems?

    Differentiating direct from indirect IFNA1 effects requires careful experimental design:

    1. Cell-specific receptor knockout/knockdown:

      • CRISPR-Cas9 deletion of IFNAR1/IFNAR2

      • siRNA knockdown in specific cell populations

      • Conditional knockout mouse models with cell-specific Cre expression

    2. Timing analysis:

      • Early transcriptomic responses (1-4 hours): Likely direct effects

      • Late responses (12-24+ hours): Potential secondary effects

      • Pulse-chase experiments with actinomycin D to block secondary transcription

    3. Direct target identification:

      • ChIP-seq for STAT1/STAT2 binding sites

      • Motif analysis for ISRE/GAS elements

      • Integration with expression data to confirm functional relevance

    4. Secondary mediator blockade:

      • Neutralizing antibodies against induced cytokines

      • Receptor antagonists for secondary signaling pathways

      • Small molecule inhibitors of downstream effectors

    5. In vitro vs. in vivo comparison:

      • Isolated cell systems to establish direct effects

      • Co-culture systems to identify cell-cell interactions

      • In vivo models to capture full complexity

    6. Systems biology approaches:

      • Network analysis to identify signaling hubs

      • Mathematical modeling of signaling cascades

      • Multi-omics integration (transcriptomics, proteomics, metabolomics)

    These strategies help create a comprehensive map of IFNA1's effects, distinguishing between primary receptor-mediated responses and secondary effects mediated by induced factors.

  • What are the sex-specific considerations when studying IFNA1 responses in research models?

    Sex differences significantly impact IFNA1 biology and should be addressed methodologically:

    1. Prevalence of autoantibodies:

      • Studies show markedly higher prevalence of anti-type I IFN autoantibodies in males (12.5%) compared to females (2.6%) with severe COVID-19

      • Odds ratio of 5.22 [95% CI: 2.27-14.80] for males vs. females

      • May influence experimental outcomes if not controlled for

    2. X-chromosome influences:

      • IFNA1 signaling components may be differentially expressed due to X-chromosome inactivation

      • Evidence from patients with X-linked incontinentia pigmenti suggests X-chromosome linkage of autoantibody production

      • Consider X-chromosome inactivation status in female subjects

    3. Experimental design considerations:

      ConsiderationMethodologyRationale
      Sex-matched controlsUse same-sex controlsPrevents confounding by sex-specific responses
      Hormonal influencesControl for estrous/menstrual cycleHormones modulate IFN responses
      Sample size calculationsPower for sex-stratified analysisMay require larger total sample size
      Cell sourcesDocument donor sex for in vitro studiesCell intrinsic sex differences exist
      Data reportingAlways report subject sexEnables meta-analysis and replication
    4. Age interactions with sex:

      • Studies indicate age-dependent increases in anti-IFN autoantibodies

      • Age-sex interactions may confound results if not accounted for

      • Age-matching should accompany sex-matching in study design

    5. Applications in drug development:

      • Different dosing requirements potentially needed by sex

      • Sex-specific adverse effect profiles may emerge

      • Pharmacokinetic differences may affect drug exposure

    Incorporating sex as a biological variable in IFNA1 research is essential for valid, reproducible, and translatable findings.

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.