IFNA7 Human, Sf9 is a recombinant interferon-alpha protein expressed in Spodoptera frugiperda (Sf9) insect cells via the baculovirus expression system. It belongs to the type I interferon family and is glycosylated, with a theoretical molecular weight of 20.7 kDa and an observed size of 18–28 kDa on SDS-PAGE due to post-translational modifications . This protein is produced for research purposes, with applications in studying antiviral mechanisms, interferon signaling, and glycoprotein engineering .
The Sf9 cell line, derived from S. frugiperda pupal ovarian tissue, is optimized for high-yield protein expression. Key advantages include:
Scalability: Suspension culture compatibility for large-scale production .
Glycoengineering: Modified Sf9 strains (e.g., SfSWT-5) enable human-like N-glycan processing, though IFNA7 retains insect-specific glycosylation .
Promoter Efficiency: The 39K promoter enhances glycogene expression compared to the ie1 promoter, improving processing efficiency .
IFNA7 is supplied as a sterile-filtered solution in PBS (pH 7.4) with 10% glycerol, ensuring stability during storage . Purity exceeds 90% as verified by SDS-PAGE .
IFNA7 activates interferon-alpha/beta (IFN-α/β) signaling, inducing:
Protein kinase R (PKR): Inhibits viral translation initiation .
Oligoadenylate synthetase: Activates ribonuclease L, degrading viral RNA .
Pathogens like vesicular stomatitis virus (VSV) and hepatitis C virus (HCV) suppress IFN-α signaling by:
Phosphorylation of IFNAR1: Leads to ubiquitination and degradation, reducing STAT1 activation .
PERK-dependent UPR: ER stress pathways attenuate IFN-α efficacy, as demonstrated in PERK knockdown studies .
Viral Strategy | Impact on IFN-α Signaling | Source |
---|---|---|
IFNAR1 degradation | Reduced STAT1 phosphorylation | |
PERK activation | Inhibition of antiviral responses |
IFNA7 is included in interferon-alpha sampler sets (e.g., PBL Assay Science’s Human IFN-Alpha Sampler) to evaluate subtype-specific antiviral efficacy . Subtype nomenclature and gene associations are summarized below:
Subtype | Gene | Key Feature |
---|---|---|
IFN-α-J1 | IFNA7 | Macrophage-derived, antiviral |
IFN-α-2a | IFNA2 | Therapeutic use in viral infections |
While Sf9-produced IFNA7 retains insect glycosylation, engineered strains (e.g., SfSWT-5) improve N-glycan processing, enabling human-like sialylation for therapeutic proteins . This system is critical for producing interferons with enhanced stability and bioactivity .
Interferon alpha-7, IFN-alpha-7, Interferon alpha-J, LeIF J, Interferon alpha-J1, IFN-alpha-J1, IFNA7, IFNA-J, IFN-alphaJ.
ADPCDLPQTH SLRNRRALIL LAQMGRISPF SCLKDRHEFR FPEEEFDGHQ FQKTQAISVL HEMIQQTFNL FSTEDSSAAW EQSLLEKFST ELYQQLNDLE ACVIQEVGVE ETPLMNEDFI LAVRKYFQRI TLYLMEKKYS PCAWEVVRAE IMRSFSFSTN LKKGLRRKDH HHHHH.
IFNA7 (Interferon alpha-7) is a member of the alpha/beta interferon family with significant antiviral properties. Also known as IFN-alpha-7, Interferon alpha-J, LeIF J, Interferon alpha-J1, or IFN-alphaJ, this cytokine is primarily produced by macrophages in response to viral infections . The protein contains 175 amino acids (residues 24-189) with a molecular mass of approximately 20.7 kDa, though the apparent molecular weight can vary due to post-translational modifications .
IFNA7 exerts its antiviral effects by binding to specific cell surface receptors, which triggers a signaling cascade that promotes the production of two key enzymes: a protein kinase and an oligoadenylate synthetase . These enzymes establish an antiviral state within cells, inhibiting viral replication through various mechanisms including protein synthesis inhibition and RNA degradation. The biological activity of IFNA7 is typically measured through anti-viral assays, with research showing effective doses (ED50) as low as 1.50-30.0 pg/mL when tested against encephalomyocarditis virus in HeLa cells .
Beyond antiviral activity, interferons like IFNA7 possess immunomodulatory properties, affecting cell proliferation, differentiation, and immune cell function. This makes IFNA7 relevant not only to infectious disease research but also to immunology and cancer biology investigations.
Sf9 cells, derived from the fall armyworm (Spodoptera frugiperda), represent a powerful eukaryotic expression system for recombinant proteins like IFNA7 for several important reasons. First, the baculovirus expression system in Sf9 cells enables high-level protein production, significantly higher than many mammalian cell systems . This is particularly valuable for obtaining sufficient quantities of IFNA7 for research purposes.
Second, while bacterial expression systems might offer higher yields, Sf9 cells provide crucial post-translational modifications necessary for proper protein folding and function. Although these modifications differ somewhat from human cells, Sf9 cells can perform O-glycosylation and form disulfide bridges essential for IFNA7 activity . The search results indicate that IFNA7 expressed in Sf9 cells undergoes O-glycosylation at the same position as natural human IFN-alpha 2, though with different glycan compositions .
Additionally, Sf9 cells grow rapidly at room temperature without requiring CO2 incubation, making them relatively easy and cost-effective to maintain compared to mammalian cells . The cells can be grown in suspension culture, allowing for scalable production processes that are advantageous for research settings.
The baculovirus expression system utilizes recombinant baculoviruses to deliver and express foreign genes in insect cells. For IFNA7 production, the human IFNA7 gene is first inserted into a baculovirus transfer vector under control of a strong promoter (typically the polyhedrin promoter), which is then used to generate recombinant baculoviruses through homologous recombination or transposition .
A critical parameter in this system is the multiplicity of infection (MOI) - the ratio of virus particles to cells. Interestingly, research suggests that lower MOI values between 0.1 and 1 are most effective for protein expression, with higher MOIs potentially decreasing productivity . As noted in the search results regarding a related protein expression study: "the expression with MOI 0.1 was similar to MOI 1 and three times higher than MOI 10" .
Harvest timing represents another crucial optimization parameter. The search results indicate that maximum enzyme activity is typically obtained at 96 hours post-transfection, though when considering productivity per hour, the 72-hour timepoint (4.03 mU mL⁻¹ h⁻¹) outperformed the 96-hour mark (3.62 mU mL⁻¹ h⁻¹) . This suggests that earlier harvest may be advantageous for balancing yield and quality.
Cell morphology provides valuable indicators of successful infection and expression. Infected cells typically show a marked increase in size compared to uninfected cells (which measure around 18.0 ± 0.67 μm) . Western blot analysis can confirm increasing expression levels, with protein accumulation typically peaking between 72-96 hours post-infection .
Optimizing these parameters allows researchers to maximize IFNA7 yield while maintaining protein quality, ensuring sufficient material for downstream applications and experiments.
Assessing purity and identity of IFNA7 expressed in Sf9 cells requires multiple complementary analytical techniques. SDS-PAGE analysis serves as the primary method for purity assessment, with commercial standards typically requiring >90% purity visualized by silver staining and quantitative densitometry with Coomassie Blue staining . Pure IFNA7 appears as bands between 18-28 kDa on SDS-PAGE, with the variation reflecting different glycoforms of the protein .
Western blotting provides specificity through antibody recognition, confirming the identity of expressed IFNA7 while also detecting potential degradation products or truncated forms. The search results indicate that Western blot analysis can reveal increasing expression levels of the target protein over time in infected Sf9 cells, while showing minimal changes in uninfected control cells .
More detailed characterization employs mass spectrometry techniques. Plasma-desorption mass spectroscopy analysis can confirm protein identity and detect modifications such as glycosylation . The search results describe how this technique revealed two types of O-linked carbohydrates in Sf9-expressed interferon: the major component being galactosyl(β1-3)-N-acetylgalactosamine and the minor component being N-acetylgalactosamine .
Reverse-phase HPLC peptide mapping provides additional information about protein composition and can detect variants with different post-translational modifications . For biological products, endotoxin testing using the LAL (Limulus Amebocyte Lysate) method ensures levels below regulatory thresholds, typically <0.10 EU per 1 μg of protein for research-grade material .
N-terminal sequencing confirms correct processing of the protein, with IFNA7 typically beginning at Cys24 as indicated in the product specifications from commercial sources . This comprehensive analytical approach ensures that researchers are working with correctly identified, high-purity IFNA7 suitable for their experimental needs.
Maintaining IFNA7 stability requires careful attention to storage conditions that prevent degradation and preserve biological activity. According to the search results, purified IFNA7 protein solution is typically formulated in Phosphate Buffered Saline (pH 7.4) containing 10% glycerol at a concentration of approximately 0.25 mg/ml . The glycerol serves as a cryoprotectant that helps maintain protein structure during freeze-thaw cycles.
Short-term storage recommendations indicate that IFNA7 can be kept at 4°C if the entire vial will be used within 2-4 weeks . This reduces the risk of degradation that might occur during repeated freeze-thaw cycles. For longer periods, storage at -20°C is recommended to minimize proteolytic degradation and preserve activity .
For extended storage periods, the addition of a carrier protein such as 0.1% Human Serum Albumin (HSA) or Bovine Serum Albumin (BSA) is advised . These carrier proteins help prevent non-specific binding to storage containers and protect the target protein from degradation through competitive inhibition of proteases and oxidative damage.
Multiple freeze-thaw cycles should be strictly avoided as they can lead to protein denaturation, aggregation, and loss of biological activity . Researchers should consider aliquoting the protein into single-use volumes before freezing to eliminate this risk. Commercial preparations of IFNA7 may come as lyophilized powder, which should be reconstituted at approximately 100 μg/mL in PBS before use .
Following these storage guidelines ensures that IFNA7 maintains its structural integrity and biological activity throughout the research timeline, providing consistent and reliable experimental results.
IFNA7 expressed in Sf9 cells exhibits several distinctive post-translational modifications compared to human-derived protein. First, Sf9-expressed IFNA7 undergoes heterogeneous O-glycosylation, with approximately 60% of the protein being glycosylated and 40% remaining non-glycosylated . The glycosylated fraction contains two types of O-linked carbohydrates: a major component consisting of galactosyl(β1-3)-N-acetylgalactosamine (disaccharide) and a minor component of N-acetylgalactosamine (monosaccharide) .
Importantly, Sf9-expressed IFNA7 lacks sialylation, with the search results specifically noting "no evidence for sialylation was found" . This contrasts with human-derived interferons, which typically contain terminal sialic acid residues that extend half-life in circulation. Despite these glycosylation differences, the O-glycosylation occurs at the same position as in natural human IFN-alpha 2 .
Disulfide bond formation represents another area of difference. The search results indicate incomplete disulfide bridge formation between Cys1 and Cys98 in a portion of the Sf9-expressed protein . This leads to the formation of "reduction-sensitive dimers" due to the lack of proper intramolecular disulfide bridges . Additionally, a minor species contains Cys1 and Cys98 in modified forms that prevent disulfide bridge formation .
Proteolytic processing also differs, with Sf9-expressed IFNA7 showing partial C-terminal truncation. Approximately 40% of the non-glycosylated material lacks the C-terminal three amino acids . This heterogeneity in primary structure may affect certain protein properties and should be considered when designing experiments.
Despite these differences in post-translational modifications, the search results suggest that Sf9-expressed interferon can exhibit "biological activity identical to that of leukocyte-derived 'natural' IFN-alpha 2" , indicating that core functionality may be preserved despite structural differences.
Multiple factors significantly impact both yield and quality of IFNA7 expression in the Sf9 baculovirus system. The multiplicity of infection (MOI) represents a critical parameter, with research suggesting that lower MOI values between 0.1 and 1 produce optimal results . Interestingly, higher MOIs do not proportionally increase yield and may actually decrease productivity, as demonstrated in the search results where "the expression with MOI 0.1 was similar to MOI 1 and three times higher than MOI 10" .
Harvest timing significantly affects both yield and quality. The data shows maximum enzyme activity at 96 hours post-transfection, but when productivity per hour is calculated, the 72-hour timepoint (4.03 mU mL⁻¹ h⁻¹) outperforms the 96-hour mark (3.62 mU mL⁻¹ h⁻¹) . This suggests that earlier harvest may optimize the balance between quantity and quality, as proteolytic degradation increases with longer culture times.
Cell density and viability during infection also impact expression outcomes. The search results note the importance of monitoring cell morphology, with infected cells showing distinct size increases compared to uninfected cells (measured at approximately 18.0 ± 0.67 μm) . Optimal infection typically occurs at mid-log phase growth when cells are most metabolically active.
Culture conditions including temperature, pH, and dissolved oxygen can significantly influence glycosylation patterns and protein folding. While the search results don't specifically address these parameters for IFNA7, they are known factors affecting recombinant protein expression in insect cells.
Proteolytic degradation presents a major challenge, with the search results confirming that Sf9-expressed IFNA7 can be "partially truncated by proteolysis" . This degradation can be mitigated through addition of protease inhibitors during purification and by optimizing harvest timing to collect protein before extensive cell lysis releases cellular proteases.
Optimizing these factors through systematic experimentation allows researchers to develop protocols that maximize both the quantity and quality of IFNA7 expressed in the Sf9 system.
Heterogeneous glycosylation of IFNA7 expressed in Sf9 cells presents a significant challenge for researchers requiring consistent protein preparations. The search results indicate that Sf9-expressed IFNA7 is "heterogeneously glycosylated," with approximately 60% glycosylated forms (containing two different types of O-linked carbohydrates) and 40% non-glycosylated material .
Several strategies can address this heterogeneity. First, researchers can implement post-purification glycoform separation using lectin affinity chromatography. Lectins such as jacalin or peanut agglutinin bind specifically to O-linked glycans, allowing separation of glycosylated from non-glycosylated forms. Elution with specific sugars (galactose or N-acetylgalactosamine) can then release the bound glycoproteins .
Hydrophobic interaction chromatography (HIC) offers another separation approach, as different glycoforms often display distinct hydrophobicity profiles. Shallow salt gradients can effectively resolve glycoforms with minor differences in surface properties. Size exclusion chromatography can further refine separation based on the slight differences in hydrodynamic radius between glycoforms.
For applications where glycosylation heterogeneity must be eliminated entirely, enzymatic deglycosylation presents a viable option. Treatment with specific O-glycosidases can remove the O-linked glycans, creating a homogeneous non-glycosylated population. This approach is particularly valuable for structural studies where glycan heterogeneity might complicate crystallization or NMR analysis.
Culture optimization represents a preventative approach to reducing heterogeneity. Lower culture temperatures (24-25°C instead of 27-28°C), controlled pH (6.2-6.4), and nutrient supplementation with glycosylation precursors can promote more complete and uniform glycosylation. Harvesting at optimal timepoints (typically earlier rather than later) may also reduce heterogeneity by limiting exposure to glycosidases released during cell lysis.
For applications where absolute glycoform homogeneity is required, researchers might consider alternative expression systems such as HEK293 cells, which provide more human-like and consistent glycosylation patterns .
Assessing the biological activity of IFNA7 expressed in Sf9 cells requires methods that confirm proper folding and functionality. The gold standard approach is the antiviral protection assay, which directly measures the protein's primary function. According to the search results, this involves using HeLa human cervical epithelial carcinoma cells infected with encephalomyocarditis (EMC) virus . The effective dose providing 50% protection (ED50) for active IFNA7 typically falls within 1.50-30.0 pg/mL range .
Researchers should include appropriate controls in these assays, particularly a reference standard such as commercially available IFNA7 or another well-characterized alpha interferon. This enables calculation of specific activity (units/mg) and facilitates comparison between different preparations or batches.
Reporter gene assays provide a faster alternative to viral protection assays. These utilize cells engineered with interferon-stimulated response elements (ISREs) controlling expression of reporter genes like luciferase. When treated with biologically active IFNA7, these cells produce quantifiable reporter signals proportional to interferon activity. These assays typically require only 6-24 hours compared to 24-72 hours for viral protection assays.
For signaling pathway analysis, researchers can assess JAK-STAT pathway activation using Western blotting for phosphorylated STAT1/STAT2 or flow cytometry for phospho-STAT1. These assays evaluate a critical early event in the interferon signaling cascade and can detect signaling-specific defects that might not be apparent in other assays.
Gene induction analysis examines the ability of IFNA7 to upregulate interferon-stimulated genes (ISGs). RT-qPCR can quantify expression of key ISGs like MX1, OAS1, or PKR in response to IFNA7 treatment. This approach provides insight into the physiological response to the interferon rather than just receptor binding or initial signaling events.
A comprehensive activity assessment would ideally combine multiple methods to gain a complete understanding of the biological activity profile of Sf9-expressed IFNA7.
IFNA7 produced in Sf9 cells differs in several important aspects from HEK293-derived IFNA7, with implications for various research applications. The most significant differences relate to post-translational modifications, particularly glycosylation patterns. Sf9-expressed IFNA7 exhibits heterogeneous O-glycosylation without sialylation, containing mainly galactosyl(β1-3)-N-acetylgalactosamine and N-acetylgalactosamine . In contrast, HEK293-expressed IFNA7 contains mammalian-type glycosylation including potential sialylation, resulting in more homogeneous products as indicated by narrower SDS-PAGE bands (18-22 kDa) compared to Sf9-expressed protein (18-28 kDa) .
Despite these structural differences, core antiviral functionality appears preserved across expression systems. The search results note that Sf9-expressed interferon alpha exhibited "biological activity identical to that of leukocyte-derived 'natural' IFN-alpha 2" . The HEK293-derived IFNA7 demonstrates potent antiviral activity with an ED50 of 1.50-30.0 pg/mL in standard assays .
For applications requiring higher purity, HEK293-derived IFNA7 typically shows advantages. Commercial preparations from HEK293 cells achieve >95% purity by SDS-PAGE, while Sf9-derived material typically reaches >90% purity . HEK293-derived material also shows more consistent batch-to-batch activity profiles due to more homogeneous post-translational modifications.
The choice between expression systems depends on specific research requirements. For basic antiviral screening assays or preliminary studies, Sf9-derived IFNA7 provides a cost-effective option with adequate biological activity. For detailed receptor binding studies, in vivo applications, or therapeutic development, HEK293-derived IFNA7 offers advantages due to its more human-like post-translational modifications and greater homogeneity.
Structural biology applications typically benefit from the more homogeneous HEK293-derived material, though for certain crystallization approaches, the lack of glycosylation or simplified glycosylation in Sf9-derived material might actually be advantageous.
Purifying IFNA7 from Sf9 cells requires a strategic approach addressing the unique challenges presented by this expression system. Based on the search results and established purification principles, a multi-step strategy is recommended for optimal results.
The initial capture step typically employs immunoaffinity chromatography, which provides high selectivity for the target protein. The search results indicate that immunoaffinity chromatography is used for purifying IFNA7 from Sf9 cells . This approach rapidly separates IFNA7 from most cellular proteins and concentrates the target protein. Alternatively, if the construct contains a His-tag (as mentioned in the search results: "IFNA7 is fused to a 9 amino acid IgG His-Tag at C-terminus"), immobilized metal affinity chromatography (IMAC) can serve as an effective initial capture method .
Intermediate purification steps address specific challenges like heterogeneous glycosylation and incomplete disulfide formation. Ion exchange chromatography can separate truncated forms based on charge differences, while lectin affinity chromatography using jacalin or similar lectins can separate glycoforms if needed. For separating correctly folded monomers from improperly disulfide-bonded forms, non-reducing size exclusion chromatography under carefully controlled conditions proves effective.
A final polishing step using size exclusion chromatography removes aggregates, dimers, and remaining impurities to achieve the high purity levels required for research applications (>90% as determined by SDS-PAGE according to the search results) . This step also facilitates buffer exchange into the final formulation.
The purified IFNA7 is typically formulated in phosphate-buffered saline (pH 7.4) containing 10% glycerol . For long-term storage, addition of a carrier protein (0.1% HSA or BSA) is recommended . This formulation maintains protein stability while preventing adsorption to storage containers.
Throughout the purification process, maintaining low temperature (4°C), including protease inhibitors, and minimizing process time helps preserve protein integrity and activity, addressing the challenge of proteolytic degradation noted in the search results .
Scaling up IFNA7 production in Sf9 cells presents several technical challenges that must be addressed to maintain yield and quality. Cell culture scale-up introduces oxygen transfer limitations not present in smaller-scale cultures. As culture volume increases, the surface-to-volume ratio decreases, potentially leading to oxygen depletion in regions of the culture. Since oxygen availability affects cell metabolism and protein glycosylation, maintaining consistent dissolved oxygen levels becomes critical for product quality.
Infection homogeneity presents another significant challenge during scale-up. In small-scale cultures, achieving uniform virus-to-cell ratios (MOI) is relatively straightforward, but larger cultures may experience virus distribution heterogeneity, leading to asynchronous infection and variable protein expression. The search results indicate that MOI optimization is critical, with lower MOI values (0.1-1) providing optimal expression . Maintaining this optimal MOI consistently throughout larger cultures requires careful mixing strategies and potentially different infection approaches.
Nutrient gradients become more pronounced in larger-scale cultures, potentially affecting glycosylation patterns of IFNA7. The search results indicate that Sf9-expressed IFNA7 already exhibits heterogeneous glycosylation , which may be exacerbated by nutrient gradients in scaled-up cultures. This could lead to increased product heterogeneity and variable post-translational modifications.
Proteolytic degradation increases with scale due to longer processing times. The search results note that Sf9-expressed IFNA7 is "partially truncated by proteolysis" . In larger-scale operations, the time from cell harvest to completed purification typically increases, providing more opportunity for proteolytic damage. This necessitates robust protease inhibition strategies throughout the process.
Purification scale-up introduces additional challenges including column packing consistency, pressure limitations, and increased processing times. The immunoaffinity chromatography method mentioned in the search results may face capacity limitations and increased cost at larger scales, potentially requiring alternative purification strategies.
Addressing these challenges requires systematic process development, including bioreactor optimization, improved mixing strategies, enhanced monitoring capabilities, and potentially different infection methodologies suited to larger-scale operations.
Detecting subtle batch-to-batch variations in Sf9-expressed IFNA7 requires sophisticated analytical methods that can characterize the protein at multiple levels. Multi-angle light scattering (MALS) coupled with size exclusion chromatography provides high-resolution analysis of protein aggregation, oligomerization states, and molecular weight distribution. This technique can detect differences in the proportion of monomeric versus dimeric forms, which is particularly relevant given the search results' mention of "reduction-sensitive dimers" in Sf9-expressed IFNA7 .
Mass spectrometry offers the most comprehensive characterization tool, with several complementary approaches. Intact mass analysis can identify differences in glycoforms, truncation variants, and other post-translational modifications. Peptide mapping combined with LC-MS/MS provides sequence coverage and identifies specific modification sites. The search results mention that "plasma-desorption mass spectroscopy analysis revealed the presence of two types of O-linked carbohydrates" in Sf9-expressed IFNA7 , demonstrating the power of mass spectrometry for detailed characterization.
Charge variant analysis using capillary isoelectric focusing (cIEF) or ion exchange chromatography can detect subtle differences in protein charge profiles arising from deamidation, terminal processing variations, or differential sialylation. While Sf9-expressed IFNA7 lacks sialylation according to the search results , other charge-altering modifications may still occur.
Circular dichroism (CD) spectroscopy provides sensitive detection of secondary structure differences that might affect protein function. Subtle changes in alpha-helical or beta-sheet content between batches can indicate variations in folding that might not be apparent in activity assays with large margins.
Differential scanning calorimetry (DSC) measures thermal stability differences between batches, with shifts in melting temperature (Tm) potentially indicating structural variations. This can be particularly important for detecting differences in disulfide bond formation, which the search results indicate is incomplete in some portion of Sf9-expressed IFNA7 .
Glycan analysis using hydrophilic interaction liquid chromatography (HILIC) with fluorescence detection provides detailed profiles of carbohydrate structures. This is especially relevant for Sf9-expressed IFNA7, which exhibits heterogeneous O-glycosylation with two different types of O-linked carbohydrates according to the search results .
Implementing these complementary analytical methods creates a comprehensive characterization package that can detect even subtle batch-to-batch variations in Sf9-expressed IFNA7, ensuring consistent research outcomes.
Interferon-alpha 7 (IFNA7) is a member of the alpha/beta interferon family, which plays a crucial role in the immune response against viral infections. Interferons are cytokines produced by host cells in response to pathogens such as viruses, bacteria, and parasites. They are named for their ability to “interfere” with viral replication within host cells.
The recombinant form of Interferon-alpha 7 is produced in Sf9 Baculovirus cells. Sf9 cells are derived from the fall armyworm (Spodoptera frugiperda) and are commonly used in biotechnology for the production of recombinant proteins. The recombinant IFNA7 produced in these cells is a single, glycosylated polypeptide chain containing 175 amino acids, with a molecular mass of approximately 20.7 kDa .
The recombinant IFNA7 protein includes a 6 amino acid His-Tag at the C-terminus, which facilitates its purification through chromatographic techniques. The protein is typically formulated in a phosphate-buffered saline solution with 10% glycerol to ensure stability. It is recommended to store the protein at 4°C for short-term use and at -20°C for long-term storage to avoid multiple freeze-thaw cycles .
Interferon-alpha 7 exhibits antiviral activities by promoting the production of two key enzymes: a protein kinase and an oligoadenylate synthetase. These enzymes play a role in inhibiting viral replication and enhancing the immune response. IFNA7 is produced by macrophages and is involved in various immune processes, including B cell activation and lymphocyte activation .
Recombinant IFNA7 is used extensively in laboratory research to study its antiviral properties and its role in the immune response. It is also used in various assays to investigate the signaling pathways mediated by interferons and their potential therapeutic applications in treating viral infections and certain types of cancer .