IFNA1A Human

IFN-Alpha 1a Human Recombinant
Shipped with Ice Packs
In Stock

Description

IFN-alpha 1a Human Recombinant produced in E.Coli is a single, non-glycosylated, polypeptide chain containing 167 amino acids and having a molecular mass of 19.5kDa. The IFNA1A ( V115A ) is purified by proprietary chromatographic techniques. 

Product Specs

Introduction
There are at least 23 known variants of Interferon alpha (IFN-alpha). These proteins vary in molecular mass from 19 to 26 kDa and consist of 156-166 or 172 amino acids. A conserved sequence region is shared by all IFN-alpha subtypes between amino acid positions 115-151, while the amino-terminal ends show variation. Many IFN-alpha subtypes differ in their sequences by only one or two positions. Naturally occurring variants also include proteins with a 10 amino acid truncation at the carboxy-terminal end.
Description

Recombinant Human IFN-alpha 1a, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 167 amino acids. It has a molecular mass of 19.5 kDa. This IFNA1A (V115A) variant is purified using proprietary chromatographic techniques.

Physical Appearance
Sterile Filtered White lyophilized (freeze-dried) powder.
Formulation

The IFN-a 1a protein was lyophilized from a 0.2µm filtered concentrated solution in PBS (pH 7.4) containing 3% Mannitol, 5% Trehalose, and 0.05% Tween-80.

Solubility
To reconstitute the lyophilized IFN alpha 1a, it is recommended to dissolve it in sterile 18MΩ-cm H2O at a concentration of not less than 100µg/ml. This solution can be further diluted in other aqueous solutions.
Stability

Lyophilized IFNA1A, while stable at room temperature for 3 weeks, should be stored desiccated at a temperature below -18°C. After reconstitution, IFN-alpha 1a should be stored at 4°C for 2-7 days. For long-term storage, it is recommended to store below -18°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.

Purity

Purity is greater than 97.0% as determined by:
(a) Reverse Phase High-Performance Liquid Chromatography (RP-HPLC) analysis.
(b) Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis (SDS-PAGE) analysis.

Biological Activity
The specific activity, as measured in a viral resistance assay, was determined to be 100,000,000 IU/mg.
Synonyms

IFN-alpha 1a, IFN-a 1a, IFN alpha 1a.

Source
Escherichia Coli.
Amino Acid Sequence

MCDLPETHSL DNRRTLMLLA QMSRISPSSC LMDRHDFGFP QEEFDGNQFQ KAPAISVLHE LIQQIFNLFT TKDSSAAWDE DLLDKFCTEL YQQLNDLEAC VMQEERVGET PLMNADSILA VKKYFRRITL YLTEKKYSPC AWEVVRAEIM RSLSLSTNLQ ERLRRKE.

Q&A

What is the genomic structure and location of the human IFNA1 gene?

The IFNA1 (interferon alpha 1) is a protein-coding gene located on chromosome 9 in the human genome. It belongs to a cluster of thirteen functional type I interferon genes situated at the 9p21.3 cytoband spanning approximately 400 kb. This cluster includes multiple IFNα subtypes (IFNA1, IFNA2, IFNA4, IFNA5, IFNA6, IFNA7, IFNA8, IFNA10, IFNA13, IFNA14, IFNA16, IFNA17, and IFNA21), as well as genes encoding other type I interferons such as IFNω (IFNW1), IFNɛ (IFNE), IFNк (IFNK), and IFNβ (IFNB1), plus 11 IFN pseudogenes . The human IFNA genes likely arose through gene conversion or recent duplication events, as evidenced by their high sequence homology .

How does IFNA1 relate to other interferon alpha subtypes in humans?

IFNA1 is one of 13 different IFN-alpha genes sharing a high degree of amino acid sequence homology, suggesting a common ancestor gene. Interestingly, mature IFNA1 and IFNA13 proteins are identical, despite being encoded by different genes . While all IFN-α subtypes signal through the same heterodimer receptor (IFNAR) expressed ubiquitously by mammalian cells, evolutionary genetic studies and functional antiviral assays strongly suggest differential functions and activities among the subtypes . The high conservation of multiple distinct IFN-α genes through evolution strongly indicates that these subtypes serve non-redundant roles in immune defense and regulation, rather than merely acting as amplifiers of a single biological activity .

What are the primary biological functions of IFNA1 protein?

The IFNA1 gene encodes interferon alpha-1 (IFN-α1), a cytokine belonging to the type I interferon family with significant antiviral, antiproliferative, and immunomodulatory properties . Type I interferons, including IFN-α1, are critical for host responses to viral infections and function by binding to the IFN-α receptor (IFNAR) consisting of IFNAR1 and IFNAR2 subunits . Upon receptor binding, IFNA1 triggers signal transduction pathways that regulate the transcription of over 2,000 genes collectively known as the "Interferome" . These pathways are essential for inflammation control, immunoregulation, tumor cell recognition, and T-cell responses .

What techniques are most effective for detecting differential expression of IFNA1 versus other IFN-α subtypes?

Detecting differential expression of specific IFN-α subtypes presents significant technical challenges due to their high sequence homology. The most effective approaches combine modified quantitative real-time PCR (qRT-PCR) techniques with specialized probes. Recommended methods include:

  • Molecular beacon (MB) probes: These contain hairpin loops that sequester fluorophores adjacent to quenchers until binding to specific templates occurs, allowing for single-base discrimination .

  • Locked nucleic acid (LNA) probes: These contain high-affinity nucleic acid analogues that stiffen the probe and raise its melting temperature (Tm), enhancing base mismatch discrimination .

  • LNA inhibitors: When necessary, these can be used to block cross-reactivity with highly similar subtypes .

For optimal results, include a four-point standard curve to compensate for differences in primer/probe set efficiency, expressing results as copy numbers per μg RNA rather than relative to housekeeping genes (HKGs), as HKG expression may vary according to stimulation and cell type .

How can researchers accurately genotype IFNAR1 promoter polymorphisms that may affect IFNA1 signaling?

Accurate genotyping of IFNAR1 promoter polymorphisms requires specialized approaches, particularly for variable number tandem repeat (VNTR) regions. A comprehensive methodology includes:

  • For next-generation sequencing data: Implement a computational pipeline that transforms alignment files (.bam) into paired-end .fastq files using Samtools-bam2fq, then map against artificial reference chromosomes using alignment tools such as Bowtie2 in "very sensitive local mode" .

  • For VNTR analysis: Create artificial reference chromosomes by splitting the IFNAR1 promoter sequence at the VNTR site (e.g., -77VNTR), leaving short repeats at each hanging end to prevent non-specific mapping due to low complexity repeats .

  • For assembly and validation: Extract mapped reads to de novo assemble the alleles using assembly software like MIRA, then analyze using bioinformatics tools such as R DNAstrings package to count tandem repeats .

  • For SNP calling: Use Samtools mpileup and bcftools to identify variations in single nucleotide polymorphisms from mapping alignments .

  • For quality control: Perform visual inspection of mapping alignments using genome browsers like IGV on a subset of samples .

This approach overcomes common limitations in standard variant-calling pipelines that often miss heterozygous VNTRs with significant length differences between alleles .

How do cellular sources and stimuli affect differential expression of IFNA1 compared to other IFN-α subtypes?

The expression of IFNA1 and other IFN-α subtypes varies significantly based on both cell type and stimulatory ligands. Research has shown that:

Methodologically, analyzing these patterns requires cell-type isolation techniques coupled with stimulus-specific time-course experiments and subtype-specific detection methods as described in FAQ 2.1 .

What factors influence the transcriptional regulation of IFNA1 gene expression?

The transcriptional regulation of IFNA1 is influenced by multiple factors:

  • Promoter polymorphisms: Several polymorphisms in the IFNAR1 promoter region, including three SNPs and one variable number tandem repeat (VNTR) at positions -568, -408, -3, and -77 respectively, can affect receptor expression levels .

  • Epigenetic modifications: Chromatin accessibility and histone modifications at the IFNA1 locus likely affect its transcriptional activity during different immune responses.

  • Transcription factors: The interferon regulatory factor (IRF) family, particularly IRF3 and IRF7, plays crucial roles in regulating type I interferon gene expression in response to pathogen recognition.

  • Feed-forward amplification: Type I interferons can induce expression of transcription factors that further enhance interferon gene expression, creating positive feedback loops during antiviral responses.

  • Cell-type specific factors: Tissue-specific transcription factors contribute to differential expression of IFNA1 across cell types, with plasmacytoid dendritic cells having specialized regulatory mechanisms allowing for rapid and robust IFN production .

Research approaches should include chromatin immunoprecipitation (ChIP) experiments to identify transcription factor binding, reporter gene assays to evaluate promoter activity, and analysis of epigenetic modifications across different cell types and activation states.

What is the evidence linking IFNA1 to autoimmune disease pathogenesis?

IFNA1, along with other type I interferons, has been implicated in several autoimmune conditions, most notably systemic lupus erythematosus (SLE) . The evidence includes:

  • Type I interferon signature: Elevated expression of interferon-stimulated genes is observed in peripheral blood cells of many SLE patients, creating what is known as the "interferon signature."

  • Genetic associations: Polymorphisms in genes related to type I interferon signaling pathways are associated with increased risk of autoimmune disorders.

  • Pathogenic mechanisms: Type I interferons promote autoimmunity through multiple mechanisms:

    • Activation of antigen-presenting cells

    • Enhanced B-cell activation and autoantibody production

    • Promotion of T helper cell differentiation toward inflammatory phenotypes

    • Prolonged survival of autoreactive immune cells

  • Therapeutic implications: The development of anti-interferon therapies, including antibodies targeting the type I interferon receptor, has shown efficacy in SLE clinical trials, supporting the pathogenic role of these cytokines .

  • Type I interferonopathies: A group of monogenic disorders characterized by aberrant activation of type I interferon signaling leads to autoinflammation and autoimmunity, further supporting the causal role of interferons in autoimmune pathology .

Research methodologies to investigate these connections include genetic association studies, transcriptomic analysis of patient samples, functional assays with patient-derived cells, and animal models of interferon-driven autoimmunity.

How does IFNA1 contribute to antiviral immunity against specific pathogens?

IFNA1 contributes to antiviral immunity through multiple mechanisms that vary in importance depending on the specific viral pathogen:

  • Direct antiviral effects:

    • Induction of interferon-stimulated genes (ISGs) that directly inhibit viral replication

    • Activation of antiviral enzymes such as protein kinase R (PKR), oligoadenylate synthetase (OAS), and MX proteins

    • Establishment of an antiviral state in neighboring uninfected cells

  • Pathogen-specific relationships:

    • Hepatitis B virus (HBV): IFN-α is used as part of first-line therapies for chronic hepatitis B (CHB), with genetic polymorphisms in the IFNAR1 promoter affecting clinical outcomes of HBV infection

    • COVID-19: IFNA1 has been associated with COVID-19 susceptibility and severity

    • Zika virus: IFNA1 plays a role in immunity against Zika virus infection

  • Immunomodulatory effects:

    • Enhancement of natural killer (NK) cell cytotoxicity

    • Promotion of dendritic cell maturation

    • Augmentation of CD8+ T cell responses

    • Modulation of B cell responses and antibody production

  • Differential effectiveness: Evidence suggests that different IFN-α subtypes may have varying potency against specific viruses, potentially linked to their evolutionary diversification .

Research approaches include viral infection models in cells with IFNA1 knockdown or overexpression, analysis of ISG induction patterns, assessment of viral replication kinetics in the presence of recombinant IFN-α1, and clinical correlation studies examining associations between IFNA1 polymorphisms and infection outcomes.

What are the key differences in signal transduction pathways activated by IFNA1 compared to other type I interferons?

While all type I interferons signal through the same heterodimeric receptor complex (IFNAR1/IFNAR2), subtle differences in signaling outcomes exist between IFNA1 and other type I interferons:

  • Receptor binding kinetics: IFNA1 may have distinct binding affinities and kinetics with the IFNAR complex compared to other subtypes, potentially affecting signal strength and duration.

  • JAK-STAT pathway utilization: Although all type I IFNs activate the JAK-STAT pathway, IFNA1 may preferentially activate specific STAT proteins (STAT1, STAT2, STAT3, etc.) in different ratios.

  • Non-canonical pathways: Beyond JAK-STAT signaling, IFNA1 activates additional pathways including PI3K-AKT, which has been identified as significantly enriched in transcriptomic studies of IFNAR1 promoter variants . This pathway connects to MDM2 proto-oncogene and the apoptosis regulator Bcl-2, controlling cell cycle progression.

  • Transcriptional profiles: Despite signaling through the same receptor, different IFN subtypes induce partially distinct sets of interferon-stimulated genes, suggesting subtype-specific signaling outcomes .

  • Cell type-specific responses: The response to IFNA1 versus other subtypes may vary based on cell type, potentially due to differences in receptor expression levels or the presence of cell-specific signaling components .

Research methodologies should include phospho-flow cytometry to measure activation of specific signaling proteins, RNA-seq to compare transcriptional responses, proximity ligation assays to detect protein-protein interactions, and single-cell analyses to capture cellular heterogeneity in responses.

How does the truncated IFNAR1 transcript affect IFNA1 signaling and what are its functional implications?

Research has identified a truncated IFNAR1 transcript with potentially significant functional implications:

  • Structural characteristics: The truncated IFNAR1 transcript results from alternative splicing or the influence of promoter polymorphisms, particularly the -77VNTR (GT repeat polymorphism) .

  • Signaling consequences: This truncated form appears to modify downstream gene expression profiles, potentially through altered receptor function or competitive inhibition of the full-length receptor .

  • Gene expression effects: Transcriptomic analyses suggest that the presence of the truncated transcript correlates with modifications in the "Interferome" (the network of interferon-regulated genes), particularly affecting genes involved in the PI3K-AKT signaling pathway .

  • Cancer relevance: The PI3K-AKT pathway links to critical oncogenic processes through the MDM2 proto-oncogene and the apoptosis regulator Bcl-2, suggesting the truncated IFNAR1 may influence cancer susceptibility or progression .

  • Methodological investigation: To study this phenomenon, researchers should:

    • Perform knock-down experiments of the truncated transcript

    • Transfect cells with plasmids expressing the truncated form

    • Conduct quantitative analysis of both the full-length and truncated transcripts

    • Measure activation of downstream signaling pathways

    • Assess functional outcomes like antiviral activity and cell proliferation

The truncated IFNAR1 transcript represents an important regulatory mechanism potentially linking interferon signaling to both antiviral immunity and cancer development .

What evolutionary forces have shaped the diversity of interferon alpha subtypes including IFNA1?

The evolution of multiple distinct interferon alpha subtypes, including IFNA1, appears to be driven by several selective pressures:

  • Gene duplication and conversion: The human IFNA genes likely arose through gene conversion or recent duplication events. Genome-wide association studies reveal that the IFNA gene families among placental mammals have undergone significant gene duplication and conversion processes .

  • Pathogen diversity: The diversification of IFN-α subtypes is theorized to be driven by the need to combat diverse viral pathogens throughout evolutionary history. Different subtypes may have evolved specialized activities against specific pathogen types .

  • Functional gains: The maintenance of multiple highly similar but distinct IFN-α genes suggests functional gains for the individual subtypes rather than simple redundancy. These genetic duplications likely conferred evolutionary advantages through enhanced or specialized immune responses .

  • Balancing selection: The need to maintain robust antiviral immunity while preventing autoimmunity likely created balancing selection pressures on the IFN-α gene family.

  • Cross-species evidence: Comparative studies across mammalian species show that while the number of IFN-α subtypes varies, the principle of having multiple subtypes is conserved, supporting their non-redundant functions .

Research approaches should include phylogenetic analyses across species, tests for signatures of positive selection at specific codons, functional comparisons of orthologous interferons from different species, and comparative genomics to identify conserved regulatory elements.

How do mouse models reflect or diverge from human IFNA1 functions in research applications?

Mouse models provide valuable but imperfect representations of human IFNA1 functions:

  • Evolutionary divergence: While the basic principles of type I interferon signaling are conserved between mice and humans, the specific interferon alpha gene repertoires differ significantly between species.

  • Functional evidence: Mouse models support the concept that IFN subtypes, rather than serving merely as amplifiers of a single biological activity, qualitatively modify the "antiviral state" in distinct ways . This principle likely applies to human IFNA1 as well.

  • Translational considerations:

    • Mouse interferon alpha genes are not direct orthologs of human IFNA genes

    • Receptor binding characteristics may differ between species

    • Downstream signaling pathways are generally conserved but may have species-specific components

    • Cell type-specific responses may vary between humans and mice

  • Experimental advantages: Despite these differences, mouse models offer unique advantages for studying interferon biology:

    • Genetic manipulation (knockout, knockin, conditional expression)

    • In vivo infection models

    • Analysis of tissue-specific responses

    • Developmental studies

  • Complementary approaches: To maximize translational relevance, researchers should complement mouse studies with:

    • Humanized mouse models expressing human IFNA genes

    • Ex vivo studies with human primary cells

    • Validation in human clinical samples

    • Comparative analyses between mouse and human responses

When interpreting results from mouse models, researchers must consider these species differences and validate key findings in human experimental systems.

What cutting-edge technologies are advancing our understanding of IFNA1 biology?

Several cutting-edge technologies are transforming IFNA1 research:

  • Single-cell RNA sequencing (scRNA-seq): This technology enables precise characterization of IFNA1 expression at the single-cell level, revealing previously unappreciated heterogeneity in producing cells and responding populations. It allows for identification of rare IFN-producing cells and analysis of cell-specific responses.

  • CRISPR-Cas9 gene editing: Precise genome editing enables:

    • Creation of IFNA1 knockout or knock-in models

    • Introduction of specific promoter polymorphisms

    • Engineering of reporter systems for live-cell imaging

    • High-throughput screening of factors regulating IFNA1 expression

  • Mass cytometry (CyTOF): This technique allows simultaneous measurement of multiple signaling pathways activated by IFNA1 at the single-cell level, providing insight into signaling heterogeneity across cell populations.

  • Spatial transcriptomics: These methods preserve spatial information about IFNA1 expression within tissues, critical for understanding its function in complex tissue environments during infection or inflammation.

  • Advanced computational approaches: Sophisticated bioinformatics techniques for analyzing the "Interferome" include:

    • Network analysis of interferon-regulated genes

    • Machine learning algorithms to predict interferon responses

    • Integration of multi-omics data (transcriptomics, proteomics, metabolomics)

    • Systems biology approaches to model interferon signaling dynamics

Future research should leverage these technologies in integrated approaches to understand IFNA1 biology at multiple scales, from molecular interactions to whole-organism phenotypes.

What are the most promising research directions for therapeutic applications targeting IFNA1?

Several promising research directions for therapeutic applications involving IFNA1 include:

  • Subtype-specific targeting: Development of technologies to selectively modulate IFNA1 versus other interferon subtypes could enable more precise therapeutic approaches:

    • Subtype-specific antibodies or aptamers for selective neutralization

    • Gene therapy approaches for selective expression

    • Small molecules that modulate specific subtype-receptor interactions

  • Genetic biomarkers: Further characterization of IFNAR1 promoter polymorphisms and their impact on treatment responses could enable personalized medicine approaches:

    • Predictive biomarkers for interferon therapy response in viral hepatitis and cancers

    • Genetic risk assessment for interferon-related adverse effects

  • Combined immunotherapies: Integration of IFNA1-based approaches with other immunotherapies:

    • Combination with checkpoint inhibitors in cancer

    • Sequential therapy regimens for chronic viral infections

    • Adjuvant applications for vaccines

  • Delivery innovations: Development of targeted delivery systems for IFNA1:

    • Tumor-targeting nanoparticles for cancer therapy

    • Liver-directed delivery for hepatitis treatment

    • Cell-specific targeting to minimize systemic side effects

  • Autoimmune applications: Further understanding of the role of IFNA1 in autoimmunity could lead to:

    • Selective antagonists for treating interferon-driven autoimmune diseases

    • Diagnostic tests based on interferon signatures

    • Therapeutic strategies targeting downstream effectors of interferon signaling

These approaches require interdisciplinary collaboration between molecular biologists, immunologists, geneticists, computational biologists, and clinicians to translate basic science discoveries into clinically applicable therapies.

Product Science Overview

Definition and Classification

Interferon-alpha 1a (IFN-α1a) is a type of recombinant protein that belongs to the family of Type I interferons. Interferons are cytokines with potent antiviral, antiproliferative, and immunomodulatory properties . IFN-α1a is produced using recombinant DNA technology, which involves inserting the human gene encoding IFN-α1a into a host cell, such as E. coli or human embryonic kidney cells (HEK293), to produce the protein .

Biological Properties and Functions

IFN-α1a exhibits several biological properties:

  • Antiviral Activity: It inhibits viral replication within host cells by inducing the expression of antiviral proteins .
  • Antiproliferative Effects: It can inhibit the proliferation of various cell types, including tumor cells .
  • Immunomodulatory Functions: It enhances the immune response by increasing the activity of natural killer (NK) cells and macrophages, and by promoting the differentiation of T cells .
Mode of Action

The mode of action of IFN-α1a involves binding to specific cell surface receptors known as interferon-alpha receptors (IFNAR1 and IFNAR2). This binding triggers a signaling cascade through the JAK-STAT pathway, leading to the transcription of interferon-stimulated genes (ISGs). These genes encode proteins that mediate the antiviral, antiproliferative, and immunomodulatory effects of IFN-α1a .

Production and Purification

Recombinant IFN-α1a is typically produced in host cells such as E. coli or HEK293 cells. The protein is then purified using chromatographic techniques to achieve high purity levels, often exceeding 95% . The final product is lyophilized and can be reconstituted in phosphate-buffered saline (PBS) for use in various applications .

Applications

IFN-α1a is used in research and clinical settings for its antiviral and anticancer properties. It is also employed in studies investigating the immune response and the mechanisms of interferon signaling .

Stability and Storage

Recombinant IFN-α1a is stable when stored at -20°C to -70°C in its lyophilized form. After reconstitution, it should be stored under sterile conditions at 2-8°C for short-term use or at -20°C to -70°C for long-term storage .

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.