IFN b 1a Human

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

Description

Introduction to IFN Beta-1a Human

IFN Beta-1a Human (Interferon Beta-1a) is a recombinant glycoprotein belonging to the type I interferon family. Produced in mammalian cell systems such as Chinese Hamster Ovary (CHO) or Human Embryonic Kidney (HEK) cells, it shares identical amino acid sequences with natural human interferon beta . This cytokine is primarily used to treat relapsing-remitting multiple sclerosis (MS), with brand names including Avonex and Rebif . Its molecular weight ranges between 18–23 kDa due to glycosylation, distinguishing it from non-glycosylated variants like IFN Beta-1b .

Mechanism of Action

IFN Beta-1a exerts therapeutic effects via the following pathways:

  • Receptor Binding: Binds to IFNα/β receptor (IFNAR), activating JAK/STAT signaling .

  • Immunomodulation:

    • Downregulates pro-inflammatory cytokines (IL-1β, IL-23) and upregulates anti-inflammatory mediators (IL-10, IL-27) .

    • Suppresses Th17 cell differentiation, reducing IL-17A and retinoic acid-related orphan receptor gamma (RORc) expression .

  • Antiviral Activity: Enhances MHC class I antigen presentation to inhibit viral replication .

Multiple Sclerosis (MS)

  • Relapse Reduction: IFN Beta-1a reduces annualized relapse rates by 27–32% compared to placebo .

  • Disability Progression: Delays progression to EDSS milestones (e.g., 4.0 or 6.0) by 37% over 2 years .

  • MRI Outcomes: Decreases gadolinium-enhancing lesions by 67% .

Table 1: Key Clinical Trials

Trial NameDesignKey FindingsSource
PRISMSRCT, 2 years44 µg SC tiw reduced relapse rate by 32% vs. placebo
EVIDENCEHead-to-head (44 µg SC tiw vs. 30 µg IM qw)44 µg group had 1.9x higher relapse-free rate at 24 weeks
SPECTRIMSSPMS cohortSlowed disability progression in higher-dose groups

Exploratory Uses

  • COVID-19: A 2020 trial showed potential efficacy in severe cases, though results were inconclusive .

Table 2: Molecular and Formulation Details

ParameterSpecification
Molecular Weight18–23 kDa (glycosylated)
Production SystemCHO/HEK cells
FormulationLyophilized with stabilizers (e.g., BSA, mannitol)
Bioactivity6 IU/mg (via A549/EMCV assay)
Half-life10–24 hours (route-dependent)
Storage-18°C (lyophilized); 4°C (reconstituted)
Common Dosages22–44 µg SC tiw (Rebif); 30 µg IM qw (Avonex)

Research Insights and Limitations

  • Dose-Frequency Dependency: Higher efficacy with subcutaneous 44 µg tiw vs. intramuscular 30 µg qw .

  • Th17 Pathway Suppression: Identified as a key mechanism in MS therapy .

  • Limitations: Non-response in 30% of MS patients, often linked to neutralizing antibodies or aggressive disease subtypes .

Product Specs

Introduction
Interferon beta (IFN-beta) is a cytokine with antiviral, antibacterial, and anticancer properties.
Description
Recombinant Human IFN-beta 1a, produced in Chinese Hamster Ovarian (CHO) cells, is a single, glycosylated polypeptide chain consisting of 166 amino acids. It has a molecular weight of 22.5 kDa. Purification is achieved using proprietary chromatographic methods.
Formulation
Lyophilized from a solution containing 91 mg Human Albumin, 620 mg mannitol, and 50mM Acetate acid at pH 3.8.
Solubility
For reconstitution, it is recommended to dissolve the lyophilized IFN beta 1a in sterile 18 MΩ-cm H2O at a concentration of at least 100 µg/ml. This solution can then be further diluted in other aqueous solutions.
Stability
Lyophilized IFN-beta 1a remains stable at room temperature for up to 3 weeks. However, for long-term storage, it is recommended to store the desiccated product below -18°C. After reconstitution, IFN-Beta1a can be stored at 4°C for 2-7 days. For prolonged storage, store below -18°C. Repeated freeze-thaw cycles should be avoided.
Purity
Purity exceeds 99.0% 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, determined through a viral resistance assay using the human "Wish" cell line and Vesicular Stomatitis Virus (VSV) or the monkey VERO cell line with Encephalomyocarditis Virus (EMCV), was found to be 270 x 106 IU/mg.
Synonyms

Leukocyte IFN, B cell IFN, Type I IFN, IFNB1, IFB, IFF, IFNB, IFN-b 1a ,MGC96956.

Source
CHO (Chinese Hamster Ovarian) cells.

Q&A

What is IFN-β-1a and how does it differ from other type I interferons?

IFN-β-1a is a recombinant human interferon beta-1a, a glycosylated protein of approximately 22 kDa belonging to the type I interferon family. It is primarily produced by fibroblasts but can also be expressed by dendritic cells, macrophages, and endothelial cells in response to pathogen exposure . Unlike other type I interferons, IFN-β-1a has distinctive structural and functional characteristics:

  • Glycosylation pattern: IFN-β-1a maintains the natural N-glycosylation of human interferon-β, which significantly impacts its stability, half-life, and biological activity .

  • Receptor binding dynamics: While all type I interferons signal through the heterodimeric IFN-α/β receptor (IFNAR1/IFNAR2), IFN-β-1a demonstrates unique binding characteristics that influence its potency and signaling dynamics .

  • Transcriptional regulation: IFN-β is specifically regulated by transcription factors including TRAF3, IRF3, IRF7, and NF-κB, creating distinct expression patterns compared to other interferon subtypes .

For experimental purposes, researchers should note that IFN-β-1a signals through the JAK/STAT pathway and can induce hundreds of interferon-stimulated genes, making it a potent immunomodulator with antiviral and antiproliferative properties .

How should glycosylation heterogeneity be considered in IFN-β-1a experiments?

Glycosylation heterogeneity significantly impacts IFN-β-1a's biological properties and should be carefully considered when designing experiments:

  • Antennarity impacts: Research demonstrates that IFN-β-1a glycoforms with higher antennarity (more branched glycan structures) better sustain bioactivity over time, which can substantially affect experimental outcomes when measuring prolonged responses .

  • Sialylation effects: The degree of sialylation influences protein stability, receptor binding kinetics, and circulation half-life, affecting both in vitro and in vivo experimental results .

  • Batch considerations: Different preparations or commercial sources of IFN-β-1a may contain varying glycoform distributions, making standardization and characterization essential for experimental reproducibility .

When conducting comparative studies with IFN-β-1a, researchers should characterize the glycosylation profile of their preparation using techniques such as electrospray ionization-mass spectrometry to ensure experimental consistency and appropriate interpretation of results . Consider including multiple bioassays (antiviral, antiproliferative, and immunomodulatory) since different glycoforms may exhibit variable potencies across different biological activities.

What methodologies are most suitable for measuring IFN-β-1a biological activity?

IFN-β-1a activity can be evaluated through several complementary methodologies:

Table 1: Methodologies for Assessing IFN-β-1a Activity

Assay TypeMethodologyMeasured ParameterAdvantagesLimitations
AntiviralCytopathic effect reductionViral replication inhibitionDirect measurement of natural functionVariable cell sensitivity
AntiproliferativeMTT/XTT assaysCell growth inhibitionQuantitative, reproducibleCell type-dependent responses
JAK/STAT SignalingPhospho-flow cytometrySTAT1/STAT2 phosphorylationSingle-cell resolution, rapidMeasures early events only
Gene InductionqPCR/RNA-seqISG expression levelsComprehensive pathway analysisTime-dependent variability
Reporter SystemsLuciferase assaysTranscriptional activationHigh-throughput capabilityArtificial construct limitations

When selecting methodologies:

  • Consider employing multiple complementary assays to comprehensively characterize IFN-β-1a activity.

  • Include standardized reference materials (e.g., WHO International Standards) for calibration.

  • Design time-course experiments to capture both early (0-4h) and late (12-24h) responses to IFN-β-1a .

  • Document specific cell types and passage numbers, as receptor density and signaling components may vary between cell systems.

These approaches provide robust evaluation of IFN-β-1a's complex biological activities while ensuring experimental reproducibility across different research settings.

How do evolutionary aspects of IFN-β-1a inform experimental design in cross-species studies?

Understanding the evolutionary conservation of IFN-β-1a is critical when designing cross-species experiments:

Human IFN-β-1a shares approximately 47% amino acid sequence identity with mouse IFN-β-1a and 46% with rat IFN-β-1a . This moderate conservation has significant implications for experimental design:

  • Species-specific receptor interactions: Substitutions at IFNAR1 and IFNAR2 contact points affect binding affinity across species, potentially altering downstream signaling dynamics and biological outcomes .

  • Cross-reactivity limitations: Human IFN-β-1a typically shows limited activity in non-primate experimental systems, necessitating species-matched interferons for most rodent studies .

  • Evolutionary selection pressure: Certain regions of IFN-β have undergone selection against nonsynonymous variants, suggesting functional conservation that should be considered when selecting experimental models .

For translational research, these evolutionary differences demand careful consideration in model selection. When human IFN-β-1a must be studied in non-human systems, researchers should verify cross-reactivity and activity using species-appropriate bioassays before proceeding with comprehensive experiments. Humanized mouse models expressing human IFNAR components may provide alternatives for studying human IFN-β-1a in vivo.

What factors influence the development of neutralizing antibodies against IFN-β-1a in experimental systems?

The development of neutralizing antibodies (NAbs) against IFN-β-1a is a significant concern in both clinical applications and experimental systems:

  • Administration route: Studies demonstrate that subcutaneous (SC) administration of IFN-β-1a leads to higher immunogenicity compared to intramuscular (IM) delivery, with SC IFN-β-1a showing higher NAb development than IM IFN-β-1a .

  • Formulation differences: Antigenicity varies among commercial products, with hierarchical immunogenicity observed: SC IFN-β-1b > SC IFN-β-1a > IM IFN-β-1a .

  • Contributing factors: Differences in immunogenicity stem from production techniques, additives, administration methods, pH variations, and in vivo protein aggregation .

  • Temporal considerations: Risk of NAb development is highest within the first 9-18 months of treatment, with clinical efficacy potentially compromised beginning 18-24 months after initiation .

For researchers conducting long-term experiments with IFN-β-1a:

  • Monitor for NAb development using standardized bioassays, particularly in longitudinal studies.

  • Consider formulation and administration route when designing experiments and interpreting results.

  • Include appropriate controls to differentiate NAb-mediated effects from other experimental variables.

  • Document batch information and administration protocols to enhance experimental reproducibility.

Understanding these factors is crucial for accurate interpretation of experimental results, particularly in longitudinal studies where NAb development might confound observations.

How can researchers effectively analyze IFN-β-1a signaling pathway crosstalk with other cytokine networks?

Investigating IFN-β-1a pathway crosstalk requires sophisticated experimental approaches:

  • Sequential stimulation experiments: Treat cells with IFN-β-1a before or after other cytokines (e.g., IL-1β, TNF-α, IL-6) to identify synergistic, additive, or antagonistic effects on downstream signaling and gene expression .

  • Receptor competition studies: Quantify changes in receptor availability and signaling component phosphorylation when multiple cytokines are present simultaneously to detect competition or cooperative effects .

  • Pathway inhibition approaches:

    • Selective JAK inhibitors to differentiate IFN-dependent from cytokine-dependent signals

    • CRISPR-based knockout of pathway-specific components to isolate direct vs. indirect effects

    • Phosphatase inhibitors to extend signal duration and enhance detection of weak interactions

  • Multi-dimensional data analysis:

    • Time-resolved proteomics to capture dynamic changes in signaling networks

    • Network analysis algorithms to identify key nodes of pathway convergence

    • Machine learning approaches to predict pathway interactions from large datasets

Table 2: Key Pathway Crosstalk Nodes for IFN-β-1a Research

Signaling NodeInteracting PathwaysDetection MethodsBiological Significance
STAT1/STAT2JAK/STAT, TLR, NF-κBPhospho-flow, IP-MSCentral to antiviral response coordination
IRF9ISGF3 complex, IRF7ChIP-seq, RNA-seqTranscriptional regulation node
SOCS proteinsJAK/STAT, IL-6, IL-10qPCR, Western blotNegative feedback regulation
mTORPI3K, MAPK, JAK/STATPhospho-antibodies, kinase assaysMetabolic integration point
USP18ISG15, JAK/STATDeubiquitination assaysPathway desensitization regulator

These approaches can reveal how IFN-β-1a signaling is modulated in complex inflammatory environments, providing insights into both basic biology and therapeutic scenarios where multiple cytokines are present simultaneously .

What are the most effective protocols for studying behavioral conditioning with IFN-β-1a in human subjects?

Behavioral conditioning studies with IFN-β-1a in humans require rigorous experimental design:

Based on published research, key methodological considerations include:

  • Study design framework: Double-blind, placebo-controlled designs are essential. A successful paradigm involved administering IFN-β-1a (6MIU of REBIF) as an unconditioned stimulus (UCS) paired with a novel drink as the conditioned stimulus (CS) .

  • Sample collection schedule: Baseline measurements should be established, followed by strategic timepoints (e.g., 4, 8, and 24 hours post-administration) to capture both immediate and delayed immune parameters .

  • Measured parameters:

    • Cellular changes: Track peripheral granulocytes, monocytes, lymphocytes, T-cells, B-cells, and NK cell numbers

    • Physiological markers: Monitor body temperature and heart rate

    • Biochemical indicators: Measure norepinephrine and IL-6 plasma levels

  • Conditioning protocol: A common approach involves:

    • Initial pairing: IFN-β-1a injection with novel stimulus

    • Washout period: Typically 8 days

    • Test phase: Placebo injection with re-exposure to conditioned stimulus

    • Control groups: Include subjects receiving placebo without CS re-exposure and untreated controls

  • Statistical analysis: Plan for adequate power to detect conditioning effects, which can be subtle. Repeated measures ANOVA with appropriate post-hoc testing is typically employed.

While a one-trial learning paradigm has shown limited success in conditioning IFN-β-1a effects , researchers should consider multiple conditioning sessions, which have demonstrated efficacy with other immune-active compounds. Careful control of environmental factors and standardization of administration protocols are essential for valid results.

How should researchers control for experimental variables when comparing different IFN-β-1a formulations?

Comparing different IFN-β-1a formulations requires strict control of experimental variables:

  • Activity standardization: Use international units (IU) based on standardized bioassays rather than protein concentration to normalize potency across formulations .

  • Glycoform characterization: Analyze glycosylation profiles using techniques such as:

    • Mass spectrometry to determine glycan antennarity and branching

    • Sialic acid content measurement to assess charge heterogeneity

    • Thermal denaturation studies to evaluate stability differences

  • Comparative assessment framework:

    • Employ multiple bioassays (antiviral, antiproliferative, immunomodulatory) simultaneously

    • Conduct reporter gene assays to measure signaling pathway activation

    • Perform receptor binding studies to determine affinity constants

  • Standardized testing conditions:

    • Use consistent cell lines at defined passage numbers

    • Maintain identical incubation times and conditions

    • Apply parallel processing of all formulations to minimize technical variation

Table 3: Critical Parameters for IFN-β-1a Formulation Comparison

ParameterMeasurement MethodImportanceTarget Value or Range
Specific ActivityAntiviral assayPrimary potency indicator200-300 MIU/mg
Sialic Acid ContentHPAEC-PAD analysisStability and PK markerFormulation-dependent baseline
Thermal StabilityDSC analysisStorage impact predictorTm > 62°C
Receptor BindingSurface plasmon resonanceBiological activity determinantKD < 100 nM for IFNAR2
Glycan AntennarityMS analysisSustained activity predictorBi- and higher-antennarity > 80%

Evidence indicates that higher-antennarity glycoforms better sustain IFN-β-1a bioactivity over time, suggesting that glycoform distribution should be carefully documented when comparing formulations . For formulations with different routes of administration (SC vs. IM), researchers should additionally consider pharmacokinetic differences that may influence experimental outcomes .

How should researchers approach contradictory findings in IFN-β-1a signaling pathway studies?

When faced with contradictory findings in IFN-β-1a signaling studies, researchers should systematically evaluate:

  • Preparation differences: Various IFN-β-1a products (REBIF, AVONEX) have distinct formulations, glycosylation patterns, and potencies that can substantially impact experimental outcomes . Document exact product sources, lot numbers, and activity measurements.

  • Experimental system variations:

    • Cell type differences: Primary cells vs. cell lines have varying receptor densities and signaling component expressions

    • Species distinctions: Human vs. murine systems may exhibit different pathway dynamics

    • Administration routes: SC vs. IM delivery affects pharmacokinetics and subsequent signaling intensity

  • Methodological factors:

    • Timing: Early (0-4h) vs. late (12-24h) measurements can yield opposing results due to feedback mechanisms

    • Dose considerations: Concentration-dependent effects may show bell-shaped rather than linear responses

    • Detection sensitivity: Different assay platforms have varying detection thresholds and dynamic ranges

  • Resolution strategies:

    • Comprehensive time-course experiments to capture temporal dynamics

    • Dose-response studies across wide concentration ranges (typically 0.1-1000 IU/mL)

    • Multi-assay approaches to measure both proximal (STAT phosphorylation) and distal (gene expression) events

    • Side-by-side comparison of contradictory protocols under identical conditions

  • Statistical considerations:

    • Apply appropriate models for complex biological responses

    • Consider population heterogeneity in cellular responses

    • Report effect sizes alongside p-values to better contextualize findings

When contradictions persist despite controlled conditions, consider the possibility of genuine biological complexity rather than experimental error, as IFN-β-1a signaling involves numerous feedback loops and pathway interactions .

What approaches are most effective for analyzing the immune cell population changes induced by IFN-β-1a?

Analyzing IFN-β-1a-induced immune cell population changes requires sophisticated approaches:

  • High-dimensional phenotyping techniques:

    • Flow cytometry with 15+ parameters to simultaneously track multiple cell populations

    • Mass cytometry (CyTOF) for 40+ parameter analysis without spectral overlap concerns

    • Spectral cytometry for enhanced discrimination of subtle phenotypic changes

  • Temporal analysis frameworks:

    • Early timepoints (4-8h): Capture immediate redistribution effects where IFN-β-1a causes significant decreases in circulating monocytes, lymphocytes, T-cells, B-cells, and NK cells, with concurrent increases in granulocytes

    • Intermediate timepoints (24-48h): Monitor recovery patterns and activation-induced phenotypic changes

    • Late timepoints (72h+): Assess sustained effects on immune cell functionality and memory formation

  • Functional assessment alongside phenotypic changes:

    • Ex vivo stimulation assays to measure responsiveness to secondary challenges

    • Cytokine production profiling before and after IFN-β-1a exposure

    • Migration and adhesion assays to correlate with circulation patterns

  • Data analysis strategies:

    • Unsupervised clustering approaches (SPADE, viSNE, FlowSOM) to identify novel cell populations

    • Trajectory analysis methods to map developmental relationships

    • Mixed-effects statistical models to account for inter-individual variability

Table 4: IFN-β-1a Effects on Major Immune Cell Populations

Cell PopulationEarly Effect (0-8h)Later Effect (24h+)Key Markers for IdentificationFunctional Significance
GranulocytesSignificant increaseReturn toward baselineCD66b+, CD16+Enhanced tissue migration
MonocytesSignificant decreaseActivation phenotypeCD14+, HLA-DRAltered antigen presentation
LymphocytesSignificant decreaseSubset-specific recoveryCD3+/CD19+/CD56+Reversible lymphopenia
NK cellsSignificant decreaseActivity enhancementCD56+, CD16+Increased cytotoxic potential
T cellsSubset-specific changesTh1/Th2 balance shiftCD3+, CD4/CD8Modified adaptive responses

Research indicates that these population changes reflect redistribution rather than apoptosis, with cells marginating to vessel walls or migrating into tissues following IFN-β-1a administration . This understanding is essential for correct interpretation of observed changes.

What emerging technologies hold the most promise for understanding IFN-β-1a receptor interactions at the molecular level?

Several cutting-edge technologies are advancing our understanding of IFN-β-1a receptor interactions:

  • Cryo-electron microscopy (Cryo-EM): Enabling high-resolution (2-3Å) visualization of IFN-β-1a/IFNAR complexes in near-native states, revealing conformational changes upon binding that influence downstream signaling specificity .

  • Single-molecule techniques:

    • Single-molecule FRET to measure receptor dimerization kinetics in real-time

    • Super-resolution microscopy (STORM/PALM) to visualize receptor clustering and diffusion dynamics at 20-30nm resolution

    • Optical tweezers to quantify binding/unbinding forces between IFN-β-1a and its receptors

  • Advanced structural biology approaches:

    • Hydrogen-deuterium exchange mass spectrometry (HDX-MS) to map conformational changes upon receptor binding

    • Site-directed spin labeling combined with electron paramagnetic resonance (EPR) spectroscopy to track dynamic receptor movements

    • AlphaFold2 and RoseTTAFold computational modeling to predict structure-function relationships

  • Receptor signaling visualization:

    • CRISPR-based endogenous tagging of receptor components with fluorescent proteins

    • Biosensor technologies for real-time visualization of JAK-STAT activation in living cells

    • Proximity labeling approaches (BioID, APEX) to map receptor interaction partners

  • Glycosylation-focused methodologies:

    • Glycoengineered IFN-β-1a variants with defined glycan structures

    • Site-specific incorporation of unnatural amino acids for precise structural studies

    • Native mass spectrometry to analyze intact glycoprotein-receptor complexes

These technologies promise to resolve longstanding questions about how IFN-β-1a's unique binding characteristics to IFNAR1/2 translate into specific signaling outcomes and biological effects . Understanding these molecular details may eventually enable the design of IFN-β-1a variants with enhanced therapeutic properties or selective activity profiles.

How might systems biology approaches enhance our understanding of IFN-β-1a's diverse biological activities?

Systems biology approaches offer powerful frameworks for comprehending IFN-β-1a's complex effects:

  • Multi-omics integration strategies:

    • Combine transcriptomics, proteomics, metabolomics, and phospho-proteomics data from IFN-β-1a-treated samples

    • Perform temporal profiling to capture dynamic system responses across multiple molecular levels

    • Apply network analysis algorithms to identify key regulatory hubs and feedback mechanisms

  • Mathematical modeling approaches:

    • Ordinary differential equation (ODE) models of IFN-β-1a signaling cascades

    • Agent-based models to simulate cellular population responses

    • Bayesian network models to infer causal relationships between pathway components

  • In silico perturbation analysis:

    • Computational prediction of system responses to receptor or pathway component modifications

    • Simulation of dose-dependent effects across varying receptor densities and glycoform distributions

    • Modeling of pathway adaptation and desensitization mechanisms

  • Translational systems approaches:

    • Integration of clinical response data with molecular profiling to identify biomarkers

    • Prediction of patient-specific responses based on baseline immune parameters

    • Identification of optimal combination therapies through network-based drug interaction models

  • Advanced visualization tools:

    • Interactive pathway maps incorporating temporal dynamics

    • Multi-scale visualizations linking molecular events to cellular and organismal responses

    • Comparative visualization of species-specific pathway architectures

These approaches enable researchers to move beyond reductionist views of IFN-β-1a biology toward a comprehensive understanding of how molecular interactions generate complex physiological responses . Systems-level insights may ultimately explain the varying efficacy of IFN-β-1a across different disease contexts and patient populations, enabling more personalized therapeutic applications.

What are the key methodological considerations for ensuring reproducibility in IFN-β-1a research?

Ensuring reproducibility in IFN-β-1a research requires attention to several critical methodological factors:

  • Preparation characterization:

    • Document exact product sources (e.g., REBIF, AVONEX) and lot numbers

    • Report specific activity in international units (IU) rather than protein concentration

    • Characterize glycosylation profiles and antennarity distribution when possible

  • Experimental design rigor:

    • Include appropriate positive and negative controls in all experiments

    • Employ standardized protocols for cell culture, treatment, and analysis

    • Perform power calculations to determine adequate sample sizes

    • Report all experimental conditions in detail, including cell passage numbers and density

  • Time and dose considerations:

    • Conduct comprehensive time-course experiments to capture the full spectrum of responses

    • Perform dose-response studies covering at least 3-4 log concentration ranges

    • Document exact timing of measurements relative to IFN-β-1a administration

  • Antibody development monitoring:

    • For longitudinal studies, regularly assess neutralizing antibody development

    • Document administration routes as they significantly impact immunogenicity

    • Report changes in bioactivity over time that might indicate NAb effects

  • Data reporting standards:

    • Share raw data alongside processed results when possible

    • Provide detailed statistical analysis methods

    • Document software versions and parameters used for data analysis

    • Report both positive and negative findings to avoid publication bias

By adhering to these methodological considerations, researchers can enhance the reproducibility of IFN-β-1a studies, facilitating more reliable knowledge accumulation in this important field. These practices are particularly crucial given the complex and sometimes contradictory nature of findings in interferon biology .

How can researchers best integrate findings from different experimental models to advance IFN-β-1a research?

Effective integration of findings across experimental models requires structured approaches:

  • Cross-model validation framework:

    • Systematically test key findings in multiple systems (cell lines, primary cells, animal models)

    • Verify critical pathways using complementary methodologies (genetic, pharmacological, biochemical)

    • Establish concordance metrics to quantify agreement between different experimental systems

  • Translational research pipeline:

    • Begin with mechanistic studies in defined cell systems

    • Validate in more complex primary human cells

    • Extend to appropriate animal models with careful consideration of species differences

    • Correlate with clinical observations when possible

  • Comparative biology approach:

    • Leverage evolutionary understanding of IFN-β conservation across species

    • Document species-specific differences in receptor binding and signaling

    • Develop conversion factors to translate findings between experimental systems

  • Meta-analysis strategies:

    • Conduct systematic reviews of methodologically similar studies

    • Perform quantitative meta-analyses when appropriate data are available

    • Identify factors contributing to heterogeneity across studies

  • Collaborative research networks:

    • Establish multi-laboratory validation studies for key findings

    • Develop shared repositories of well-characterized reagents and protocols

    • Create standardized reporting formats to facilitate cross-study comparisons

Product Science Overview

Production and Structure

IFN-Beta 1a is produced by mammalian cells, typically Chinese Hamster Ovary (CHO) cells, which have been genetically modified to express the human interferon beta gene . This recombinant protein is glycosylated and has a molecular weight of approximately 22.5 kDa . The amino acid sequence of IFN-Beta 1a is identical to that of natural human interferon beta .

Mechanism of Action

IFN-Beta 1a exerts its effects by binding to the interferon alpha/beta receptor (IFNAR), which is a heterodimeric receptor composed of IFNAR1 and IFNAR2 subunits . Upon binding, it activates the JAK-STAT signaling pathway, leading to the transcription of interferon-stimulated genes (ISGs) that play a crucial role in antiviral defense, immune regulation, and cell proliferation .

Clinical Applications

The primary clinical application of IFN-Beta 1a is in the treatment of multiple sclerosis (MS). It has been shown to reduce the frequency and severity of MS relapses by modulating the immune response and reducing inflammation . IFN-Beta 1a is also being investigated for its potential use in other autoimmune diseases and certain types of cancer .

Research and Development

Research on IFN-Beta 1a continues to evolve, with ongoing studies exploring its broader applications and optimizing its production. Advances in biotechnology have led to the development of more efficient and cost-effective methods for producing recombinant IFN-Beta 1a with lower endotoxin levels .

Storage and Stability

Recombinant IFN-Beta 1a is typically stored at -20 to -70 °C to maintain its stability and bioactivity . It is important to avoid repeated freeze-thaw cycles to prevent degradation of the protein .

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.