IFNAR1 Mouse

Interferon Alpha And Beta Receptor Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Definition and Genetic Background of IFNAR1 Mouse Models

The IFNAR1 mouse refers to genetically engineered murine models used to study the role of the interferon-alpha/beta receptor 1 (IFNAR1), a critical component of the type I interferon (IFN-I) receptor complex. IFNAR1 pairs with IFNAR2 to mediate signaling downstream of IFN-α/β, enabling antiviral, immunomodulatory, and antitumor responses. These models include knockout (KO) and humanized variants, designed to elucidate IFN-I biology or mimic human receptor interactions.

FeatureIFNAR1 KO MiceHumanized IFNAR-hEC Mice
Genetic modificationDeletion of exon 2 in Ifnar1 gene Humanized extracellular domains in Ifnar1/2
PurposeStudy IFN-I deficiency and immune dysfunctionEnable human IFN-I signaling in mice
Species-specificityRetains mouse IFN-I signaling Responds to human IFN-α/β
Immune phenotypeImpaired antiviral responses, increased myeloid cells Functional human IFN-I responses

IFNAR1 Knockout (KO) Mice

  • Immune Dysregulation:

    • Myeloid Cell Expansion: Increased CD11b+ myeloid cells and granulocytes in bone marrow, but no changes in T, B, or NK cells .

    • Impaired Tumor Immunity: Accelerated tumor growth due to defective neutrophil-mediated T-cell activation .

    • Reduced IFN-λ Efficacy: Tumor growth in KO mice is partially rescued by IFN-λ, which bypasses IFNAR1 .

ParameterWild-Type MiceIFNAR1 KO Mice
Tumor GrowthSlowerFaster
Neutrophil-T Cell InteractionsHighLow
IFN-γ ProductionNormalReduced

Humanized IFNAR-hEC Mice

  • Human IFN-I Responsiveness:

    • STAT1/2 Phosphorylation: Concentration-dependent activation in hepatocytes treated with human IFN-α .

    • Subtype-Specific Effects:

      • IFN-α14 > IFN-α2: Greater STAT1/2 activation, ISG expression, and antigen-specific CD8+ T-cell expansion .

      • Synergy with IFN-γ: IFN-α14 enhances IFN-γ signaling, potentiating antiviral and immunogenic effects .

IFN-α SubtypeSTAT1/2 ActivationISG ExpressionCD8+ T-Cell Response
IFN-α2ModerateModerateModerate
IFN-α14HighHighHigh

Antiviral and Immunomodulatory Studies

  • HBV Infection: Humanized IFNAR-hEC mice enable testing of pegylated human IFN-α2 for HBV suppression .

  • Neuroinflammation: Ifnar1 antisense oligonucleotides (ASOs) reduce cerebral interferonopathy in transgenic mice, preserving blood-brain barrier integrity .

Cancer Immunology

  • Tumor Microenvironment: IFNAR1 deficiency correlates with reduced T-cell activation and increased metastatic potential, underscoring the role of type I IFNs in anti-tumor immunity .

Antibody Development

  • MAR1-5A3 Antibody: A monoclonal antibody blocking mouse IFNAR1, used to inhibit type I IFN signaling in vivo and in vitro .

Limitations and Challenges

  • Species-Specificity: Mouse IFNAR1 does not bind human IFN-I, necessitating humanized models for translational studies .

  • Complex Signaling: IFN-I subtypes exhibit distinct potencies, requiring tailored therapeutic approaches .

  • Off-Target Effects: Systemic IFNAR1 blockade may compromise antiviral defense in wild-type mice .

Product Specs

Introduction
The Interferon Alpha And Beta Receptor, also known as IFNAR1, is a member of the class II cytokine receptor family. This receptor is composed of two chains, one of which is the IFNAR1 protein. IFNAR1 binds to and activates the receptor in response to interferons alpha and beta. This activation triggers the stimulation of Janus protein kinases, which then phosphorylate various proteins, including STAT1 and STAT2. Notably, IFNAR1 also exhibits antiviral properties.
Description
Recombinant IFNAR1 from mice, produced in Sf9 Baculovirus cells, is a single, glycosylated polypeptide chain. It comprises 411 amino acids (27-429a.a) and exhibits a molecular mass of 46.8 kDa. On SDS-PAGE, the apparent molecular size will be approximately 40-57 kDa. The IFNAR1 protein is fused to a 6 amino acid His-tag at the C-terminus and undergoes purification using proprietary chromatographic methods.
Physical Appearance
A clear, sterile solution.
Formulation
The IFNAR1 protein solution has a concentration of 0.5 mg/ml and is prepared in phosphate-buffered saline (pH 7.4) with 10% glycerol.
Stability
For short-term storage (2-4 weeks), the solution should be kept at 4°C. For longer storage, it should be frozen at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for extended storage. Repeated freezing and thawing should be avoided.
Purity
The purity is greater than 90.0% as assessed by SDS-PAGE.
Synonyms
IFN-R-1, IFN-alpha/beta receptor 1, Type I interferon receptor 1, Ifar, Ifnar, Infar CD118 and Ifnar1.
Source
Sf9, Baculovirus cells.
Amino Acid Sequence

ENLKPPENID VYIIDDNYTL KWSSHGESMG SVTFSAEYRT KDEAKWLKVP ECQHTTTTKC EFSLLDTNVY IKTQFRVRAE EGNSTSSWNE VDPFIPFYTA HMSPPEVRLE AEDKAILVHI SPPGQDGNMW ALEKPSFSYT IRIWQKSSSD KKTINSTYYV EKIPELLPET TYCLEVKAIH PSLKKHSNYS TVQCISTTVA NKMPVPGNLQ VDAQGKSYVL KWDYIASADV LFRAQWLPGY SKSSSGSRSD KWKPIPTCAN VQTTHCVFSQ DTVYTGTFFL HVQASEGNHT SFWSEEKFID SQKHILPPPP VITVTAMSDT LLVYVNCQDS TCDGLNYEII FWENTSNTKI SMEKDGPEFT LKNLQPLTVY CVQARVLFRA LLNKTSNFSE KLCEKTRPGS FSTLEHHHHH H

 

Q&A

What is IFNAR1 and why are IFNAR1 mouse models important in research?

IFNAR1 is a critical component of the type I interferon receptor complex that mediates cellular responses to type I interferons (IFN-I). This receptor is crucial for the first line of host defense against invading viruses and plays significant roles in various immunological processes . IFNAR1 mouse models, including knockout and transgenic variants, enable researchers to study the specific roles of IFN-I signaling in different cell types and disease contexts.

These models are particularly valuable because they allow dissection of cell-specific effects of type I interferons. For example, the IFNAR1 Texcl transgenic mouse model expresses IFNAR1 exclusively on T cells, providing a unique in vivo system for studying direct activities of IFN-I on T cells in various autoimmune, infectious, or neoplastic inflammatory conditions .

What are the main types of IFNAR1 mouse models available for research?

Several distinct IFNAR1 mouse models exist for different research applications:

  • Global IFNAR1 knockout mice (Ifnar1-/-): These mice lack functional IFNAR1 in all cells, resulting in complete absence of type I IFN signaling throughout the body .

  • Cell-specific IFNAR1 transgenic models: These include models like IFNAR1 Texcl mice, which express IFNAR1 exclusively on T cells while lacking expression in all other cell types .

  • Humanized IFNAR mice (HyBNAR): These transgenic mice harbor humanized type I interferon receptors, containing human extracellular domains fused to mouse transmembrane and cytoplasmic segments. This allows for the study of human type I interferons in a mouse system .

Each model offers unique advantages for specific research questions, ranging from examining global effects of IFN-I deficiency to isolating cell-specific responses.

How do IFNAR1 knockout mice respond to viral infections compared to wild-type mice?

IFNAR1 knockout mice exhibit dramatically increased susceptibility to viral infections. The search results demonstrate that these mice show:

  • Accelerated disease progression: In SARS-CoV-2 infection models, hACE2-expressing Ifnar1-/- mice lost weight significantly faster, developed hypothermia more rapidly, and displayed severe clinical symptoms much earlier than control mice .

  • Higher viral loads: Significantly higher viral replication is typically observed in the tissues of IFNAR1-deficient mice during infection .

  • Enhanced tissue damage: These mice frequently show more severe inflammation, increased immune cell infiltration, and greater pathology in infected organs .

  • Increased mortality: For example, Usutu virus (USUV) is lethal in Ifnar1-/- mice at doses as low as 20 pfu per mouse, whereas immunocompetent mice typically resist the infection .

These enhanced susceptibility phenotypes make IFNAR1 knockout mice valuable for studying viral pathogenesis mechanisms and for testing antiviral interventions that might compensate for the lack of IFN-I signaling.

What insights have IFNAR1 mouse models provided about autoimmune diseases?

IFNAR1 mouse models have revealed complex and sometimes paradoxical roles of type I interferons in autoimmune diseases:

In experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis, IFNAR1 Texcl mice (expressing IFNAR1 only on T cells) showed a significantly delayed onset and milder progression of disease compared to complete Ifnar1-/- mice . This finding revealed a previously unknown protective effect of endogenous IFN-I when acting directly on T cells during the early phase of EAE.

This research highlights how cell-specific IFNAR1 signaling can have protective effects in certain contexts, while global IFNAR1 deficiency might exacerbate disease, illustrating the complex role of type I interferons in autoimmunity.

How have IFNAR1 mouse models contributed to our understanding of SARS-CoV-2 pathogenesis?

IFNAR1 mouse models have been instrumental in establishing the critical protective role of type I interferons during SARS-CoV-2 infection:

Researchers developed hACE2-expressing Ifnar1-/- mice (with dampened IFN-I response) and compared them to hACE2; Irgm1-/- mice (with constitutively high IFN-I response) . The studies revealed:

  • Protective role of IFN-I: hACE2; Ifnar1-/- mice showed severe SARS-CoV-2 infection with enhanced immune cell infiltration, inflammatory response, and lung pathology .

  • Neuroinvasion in absence of IFN-I protection: These mice were highly susceptible to SARS-CoV-2 neuroinvasion, showing immune cell infiltration in the brain, microglia/astrocyte activation, cytokine response, and neuronal demyelination .

  • IFN-I-dependent protection mechanism: In contrast, mice with heightened IFN-I responses (hACE2; Irgm1-/- mice) were resistant to lethal SARS-CoV-2 infection and showed substantially reduced cytokine storm and immunopathology .

These findings definitively demonstrated that IFN-I protects from lethal SARS-CoV-2 infection and suggested that targeting pathways like Irgm1 could have therapeutic potential.

What are important considerations when determining viral challenge doses in IFNAR1-deficient mice?

When designing viral challenge experiments with IFNAR1-deficient mice, dose selection is critical due to their heightened susceptibility:

  • Start with significantly lower doses: Research with Usutu virus showed that doses as low as 20 pfu per mouse were sufficient for lethal infection in Ifnar1-/- mice, which is considerably lower than what might be used in wild-type mice .

  • Consider dose-dependent disease kinetics: Higher viral doses (10^3 pfu/mouse or above) typically cause very rapid lethality in Ifnar1-/- mice, which may not allow sufficient time to study disease progression or evaluate interventions .

  • Establish a dose-response curve: Researchers should conduct preliminary experiments with a range of doses (e.g., from 5×10^2 to 5×10^4 pfu/mouse) to determine the relationship between dose and survival time .

  • Select doses based on experimental objectives:

    • For comparative virulence studies between viral strains, lower doses that extend survival time allow better discrimination of subtle virulence differences.

    • For therapeutic efficacy studies, doses that provide a wider window between symptom onset and humane endpoint are preferable.

  • Account for strain-specific differences: Different viral strains may show varying lethality in Ifnar1-/- mice even at the same dose, so strain-specific dose optimization is recommended .

These considerations are essential for developing meaningful and interpretable experiments while minimizing animal distress.

How should researchers monitor disease progression in IFNAR1-deficient mice during viral challenge experiments?

Proper monitoring of disease progression in IFNAR1-deficient mice requires comprehensive assessment of multiple parameters:

  • Weight loss tracking: Daily weight measurements expressed as a percentage of initial weight provide a quantitative measure of disease severity. In the Usutu virus study, significant weight loss preceded other clinical signs and correlated with disease outcome .

  • Body temperature monitoring: Hypothermia is often observed in severely ill mice and can be a predictor of imminent mortality, as seen in SARS-CoV-2-infected hACE2; Ifnar1-/- mice .

  • Clinical scoring systems: Implement standardized scoring for symptoms such as:

    • Activity level/lethargy

    • Posture (hunched vs. normal)

    • Coat condition (ruffled vs. smooth)

    • Ocular discharge

    • Neurological signs (limb weakness, paralysis, ataxia)

    • Respiratory distress

  • Viremia assessment: Serial blood sampling (e.g., tail bleeds on alternating days) allows tracking viral load kinetics via RT-qPCR, which can be correlated with clinical progression .

  • Tissue-specific viral burden: Terminal tissue collection and viral load quantification in relevant organs (brain, lungs, liver, spleen, etc.) provides insights into viral tropism and dissemination .

  • Humane endpoints: Clearly defined criteria for euthanasia should be established before experiments begin, typically including weight loss exceeding 20%, severe neurological symptoms, or inability to access food/water .

This comprehensive monitoring approach enables more detailed phenotypic characterization and better comparison between experimental groups.

What control groups are essential when working with IFNAR1 transgenic and knockout mouse models?

Proper experimental design with IFNAR1 mouse models requires carefully selected controls:

  • Wild-type controls: Age-matched mice of the same genetic background without IFNAR1 modification are essential baselines. For instance, comparing Ifnar1-/- mice with wild-type counterparts reveals the full impact of IFN-I signaling absence .

  • Heterozygous controls: Ifnar1+/- mice can help determine if gene dosage effects exist in IFN-I responses.

  • Cell-specific controls: When using models like IFNAR1 Texcl (T-cell-specific expression), comparison with both wild-type and global Ifnar1-/- mice is necessary to distinguish cell-specific from systemic effects of IFN-I signaling .

  • Vehicle/mock infection controls: Mice receiving only the vehicle (e.g., DMEM medium) are necessary to control for stress, injury, or immune activation from the inoculation procedure itself .

  • Conditional knockout temporal controls: For inducible IFNAR1 deletion models, controls should include both pre-induction samples and non-induced transgenic mice.

  • Dose-matched controls: When comparing different viral strains or variants, identical inoculation doses should be used across groups to enable valid comparisons of virulence .

The IFNAR1 Texcl mouse study demonstrated the importance of proper controls by revealing that the protective effect of T cell-specific IFNAR1 expression in EAE was masked in wild-type mice due to counterbalancing effects of IFN-I signaling in non-T cell compartments .

How can researchers manage the high susceptibility of IFNAR1-deficient mice to opportunistic infections?

IFNAR1-deficient mice require special handling due to their compromised antiviral immunity:

  • Enhanced housing precautions:

    • Use individually ventilated caging systems

    • Maintain specific-pathogen-free conditions with enhanced microbiological monitoring

    • Consider using HEPA-filtered changing stations for cage maintenance

    • Implement strict barrier facility protocols

  • Health monitoring program:

    • Conduct regular screening for common mouse pathogens that might be subclinical in wild-type mice but pathogenic in Ifnar1-/- colonies

    • Monitor sentinel animals from the same housing room

    • Consider periodic testing of fecal pellets for viral pathogens

  • Prophylactic measures:

    • Maintain Ifnar1-/- mice on autoclaved food, bedding, and water

    • Consider prophylactic antibiotic treatment when conducting procedures that might compromise mucosal barriers

    • Minimize stress during handling and transportation

  • Experimental timing:

    • Complete experiments within shorter timeframes when possible

    • Be prepared for increased attrition rates in long-term studies

  • Breeding strategies:

    • Maintain breeder colonies in enhanced barrier facilities

    • Consider heterozygous breeding schemes to reduce opportunistic infection risk in breeders

These precautions are particularly important when working with models like the hACE2; Ifnar1-/- mice, which showed extreme susceptibility to SARS-CoV-2 infection even at low viral doses .

How can researchers differentiate between direct effects of IFNAR1 deficiency and secondary compensatory mechanisms?

Distinguishing primary effects of IFNAR1 deficiency from compensatory adaptations requires specialized approaches:

  • Acute IFNAR1 inhibition models:

    • Compare constitutive Ifnar1-/- mice with wild-type mice receiving anti-IFNAR1 blocking antibodies

    • Short-term antibody blockade prevents long-term compensatory changes

  • Inducible knockout systems:

    • Use tamoxifen-inducible or tetracycline-regulated IFNAR1 deletion systems

    • Analyze both early (primary effects) and late (compensatory) timepoints after induction

  • Combined deficiencies:

    • Generate double knockout models lacking both IFNAR1 and potential compensatory pathways

    • The study combining Irgm1-/- with Ifnar1-/- demonstrated how secondary pathways impact disease outcomes

  • Cell-specific models:

    • Compare phenotypes between global Ifnar1-/- and cell-specific models like IFNAR1 Texcl

    • This approach revealed distinct T cell-specific protective effects of IFNAR signaling that were masked in global knockouts

  • Developmental timing analysis:

    • Compare adult-induced IFNAR1 deficiency with congenital deficiency

    • Assess developmental milestones in immune system maturation

The IFNAR1 Texcl transgenic model represents a particularly valuable tool for this purpose, as it enables researchers to isolate T cell-specific responses to IFN-I in vivo while eliminating effects in other cell types .

How should researchers interpret seemingly contradictory findings between different IFNAR1 mouse models?

Resolving contradictory findings between IFNAR1 mouse models requires systematic analysis:

  • Consider cell type-specific effects: The IFNAR1 Texcl study revealed that IFN-I signaling in T cells has protective effects in EAE that are masked in global Ifnar1-/- mice, highlighting how cell-specific and systemic IFN-I effects can oppose each other .

  • Analyze temporal dynamics: Different models may reveal distinct roles of IFN-I at different disease stages. What appears contradictory may reflect time-dependent functions of the same pathway.

  • Evaluate genetic background influences: IFNAR1 models on different mouse strain backgrounds may show varying phenotypes due to modifier genes. Document the exact strain background of all models compared.

  • Assess environmental factors: Housing conditions, microbiota, and pathogen exposure can significantly influence outcomes in immunodeficient models.

  • Examine experimental methodologies:

    • Infection route (e.g., subcutaneous vs. intranasal)

    • Dose differences

    • Timing of measurements

    • Assay sensitivities

  • Consider viral strain differences: Studies of Usutu virus showed that different isolates varied in their virulence in Ifnar1-/- mice . Similarly, one Af-3-NL strain of USUV previously reported as non-lethal was found to be lethal in subsequent studies, highlighting potential viral adaptation during laboratory passage .

  • Triangulate with additional models: When findings conflict, adding a third model or approach can provide clarity. For example, combining Ifnar1-/- and Irgm1-/- models in SARS-CoV-2 research helped establish the protective role of IFN-I signaling .

Understanding these variables can transform seemingly contradictory findings into complementary insights about the complex roles of IFN-I signaling.

What approaches should be used to analyze the tissue-specific effects of IFNAR1 deficiency during viral infection?

Comprehensive analysis of tissue-specific effects in IFNAR1-deficient mice requires multi-dimensional approaches:

  • Quantitative viral load assessment:

    • Measure viral RNA by RT-qPCR in different tissues

    • Use standardized reference materials for absolute quantification

    • Express results as viral RNA copies per gram of tissue for cross-tissue comparison

  • Tissue compartmentalization analysis:

    • Assess viral distribution across anatomical subdivisions (e.g., brain regions)

    • The Usutu virus study examined viral loads in distinct brain regions (olfactory bulb, frontal lobe, cerebellum, cortex, brain stem) revealing region-specific vulnerability

  • Correlation with clinical signs:

    • Link tissue-specific viral loads with relevant clinical manifestations

    • For example, high viral loads in the brain stem correlated with neurological symptoms in USUV-infected mice

  • Histopathological assessment:

    • Perform histological analysis with scoring systems for inflammation, tissue damage, and cellular infiltrates

    • Use immunohistochemistry to identify infected cell types within tissues

  • Immune profiling by tissue:

    • Analyze infiltrating immune cell populations using flow cytometry

    • Measure local cytokine/chemokine production by tissue homogenate analysis

    • Compare systemic (serum) vs. local (tissue) immune responses

  • Temporal dynamics:

    • Sample tissues at multiple timepoints to track infection progression

    • Identify tissues that serve as initial targets versus sites of secondary spread

  • Comparison across models:

    • Contrast findings between global Ifnar1-/- mice and cell-specific models

    • The SARS-CoV-2 study demonstrated how IFN-I deficiency affected both pulmonary and neurological tissues differently

This multi-parametric approach provides a comprehensive understanding of how IFNAR1 deficiency affects tissue-specific viral pathogenesis and host responses.

Product Science Overview

Introduction

Interferon alpha and beta receptors (IFNAR) are critical components of the immune response in mammals. These receptors are part of the type I interferon receptor family and play a pivotal role in mediating the effects of interferons, which are cytokines involved in antiviral defense, immune regulation, and cell proliferation.

Structure and Function

The interferon alpha and beta receptor is a heterodimeric complex composed of two subunits: IFNAR1 and IFNAR2. These subunits belong to the class II cytokine receptor family. The receptor complex mediates the signaling of type I interferons, including interferon-alpha (IFN-α) and interferon-beta (IFN-β), by binding to these cytokines and initiating a cascade of intracellular events that lead to the expression of interferon-stimulated genes (ISGs).

  • IFNAR1: This subunit is approximately 100-130 kDa in size and is responsible for binding to the interferon molecules. It is essential for the receptor’s ability to mediate the biological effects of interferons .
  • IFNAR2: This subunit exists in multiple isoforms, including a long form that participates in signal transduction and a short form that acts as a decoy receptor. The long form of IFNAR2 is crucial for the activation of downstream signaling pathways .
Recombinant Mouse IFNAR

Recombinant mouse IFNAR proteins are produced using advanced biotechnological methods. These recombinant proteins are used in research to study the signaling mechanisms and biological effects of interferons in a controlled environment. The recombinant mouse IFNAR1 protein, for example, is derived from a mouse myeloma cell line and is purified to high standards to ensure its functionality in experimental settings .

Biological Significance

The interferon alpha and beta receptor is integral to the immune response against viral infections. Upon binding to their respective receptors, IFN-α and IFN-β trigger the activation of the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway. This leads to the transcription of ISGs, which encode proteins that inhibit viral replication, enhance antigen presentation, and modulate the activity of immune cells .

In addition to their antiviral properties, type I interferons and their receptors are involved in the regulation of cell growth and differentiation. They have been shown to suppress the proliferation of cancer cells and modulate the immune response in various autoimmune diseases .

Applications in Research and Medicine

Recombinant mouse IFNAR proteins are valuable tools in biomedical research. They are used to investigate the molecular mechanisms of interferon signaling, study the effects of interferons on different cell types, and develop therapeutic strategies for diseases such as multiple sclerosis, hepatitis, and certain cancers .

In clinical settings, recombinant interferons are used as therapeutic agents to treat viral infections and immune-related disorders. Understanding the interactions between interferons and their receptors is crucial for optimizing these treatments and developing new therapeutic approaches .

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.