TNFR Mouse

Tumor Necrosis Factor Receptor Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

TNFR Mouse Recombinant produced in E.coli is a single, non-glycosylated polypeptide chain containing 191 amino acids and having a molecular mass of 21.1kDa.
The TNFR is purified by proprietary chromatographic techniques.

Product Specs

Introduction
Tumor necrosis factor receptor 1 (TNFR1), a member of the TNF-receptor superfamily, binds to TNFSF2/TNF-alpha and homotrimeric TNFSF1/lymphotoxin-alpha. Two soluble TNF receptors, sTNFR-I and sTNFR-II, neutralize the biological activities of TNF-alpha and TNF-beta. These soluble receptors are thought to increase due to shedding of the extracellular domains of their membrane-bound counterparts. TNF-a, TNFR1, and TNFR2 play roles in cellular differentiation, while TNFR1 and TNFR2 are involved in cell type-specific renal injury. TNFR1 can signal both cell survival and apoptosis. Apoptosis induced by TNFR1 requires two sequential signaling complexes. Additionally, TNFR1 can activate NF-kappaB, mediate apoptosis, and regulate inflammation. Oxidative stress promotes TNFR1 and TNFR2 self-interaction, leading to ligand-independent and enhanced ligand-dependent TNF signaling. TNFR1 contributes to non-cytocidal TNF effects like the anti-viral state and activation of acid sphingomyelinase. Human TNFR1 possesses a major region that regulates cell surface expression. Amniotic fluid of pregnant women contains high levels of soluble TNF receptors. Germline mutations in the extracellular domains of TNFR1 are associated with the autosomal dominant periodic fever syndrome, with impaired receptor clearance suggested as a potential disease mechanism. Familial hibernian fever (FHF) is caused by defects in the TNFRSF1A gene.
Description
Recombinant Mouse TNFR, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 191 amino acids with a molecular weight of 21.1 kDa. The purification of TNFR is carried out using proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White lyophilized powder.
Formulation
The TNFR protein was lyophilized from a 0.2 µm filtered solution in phosphate-buffered saline (PBS) at pH 7.4.
Solubility
It is recommended to reconstitute the lyophilized TNFR in sterile 18 MΩ-cm H2O to a concentration of at least 100 µg/ml. This solution can be further diluted in other aqueous solutions.
Stability
Lyophilized TNFR, while stable at room temperature for 3 weeks, should be stored desiccated at temperatures below -18°C. After reconstitution, TNFR should be stored at 4°C for 2-7 days. For long-term storage, it is recommended to store at -18°C after adding a carrier protein (0.1% HSA or BSA). Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 97.0% by SDS-PAGE analysis.
Biological Activity
The ED₅₀, determined by the ability to inhibit TNF-α mediated cytotoxicity in L-929 cells, is less than 1.0 µg/ml. This corresponds to a specific activity greater than 1000 IU/mg in the presence of 0.1 ng/mL of recombinant murine TNF-α.
Synonyms
Tumor necrosis factor receptor superfamily member 1A, Tumor necrosis factor receptor 1, TNF-R1, Tumor necrosis factor receptor type I, TNF-RI, TNFR-I, p55, p60, CD120a, Tnfrsf1a, Tnfr-1, Tnfr1, FPF, TNF-R, TNFAR, TNFRI, p55-R, TNFR60, Tnfr-2, TNF-R-I, TNF-R55, TNFRp55, TNF-alphaR1, TNFalpha-R1.
Source
Escherichia Coli.
Amino Acid Sequence
IHPSGVTGLV PSLGDREKRD SLCPQGKYVH SKNNSICCTK CHKGTYLVSD CPSPGRDTVC RECEKGTFTA SQNYLRQCLS CKTCRKEMSQ VEISPCQADK DTVCGCKENQ FQRYLSETHF QCVDCSPCFN GTVTIPCKET QNTVCNCHAG FFLRESECVP CSHCKKNEEC MKLCLPPPLA NVTNPQDSGT A.

Q&A

What are the primary differences between TNFR1 and TNFR2 in mouse models?

TNFR1 and TNFR2 work antithetically to balance CNS immune responses in autoimmune diseases such as multiple sclerosis . While TNFR1 is widely expressed and primarily mediates pro-inflammatory responses and cell death pathways, TNFR2 expression is more restricted and generally promotes tissue repair and immune regulation. In mouse models, TNFR1 knockout mice show increased apoptosis following TNF treatment compared to wild-type mice, demonstrating TNFR1's protective role against TNF-induced cell death . Conversely, TNFR2 knockout mice exhibit impaired proliferative responses to TNF stimulation .

How are various TNFR knockout mouse models generated and validated?

TNFR knockout mice are typically generated through:

  • Traditional gene targeting via homologous recombination

  • Site-specific recombinase systems (e.g., Cre-loxP)

  • Tissue-specific knockout approaches

For validation, researchers utilize:

  • PCR-based genotyping (as described for TNFR KO mice )

  • Western blot analysis for protein expression

  • Functional assays to confirm receptor inactivation

Tissue-specific knockouts, like intestinal epithelium-specific Raf knockout (Raf KO IE) mice, are created by crossing floxed mice (e.g., Raf flx/flx) with tissue-specific Cre expressing lines (e.g., villin-Cre) .

How do mouse and human TNF-TNFR systems differ?

There are important species-specific differences in the TNF-TNFR system:

  • Human TNF-α can bind to mouse TNFR1, as demonstrated in Biacore chip experiments

  • Anti-human TNFR1 antibodies like ATROSAB do not recognize mouse TNFR1, necessitating the development of humanized mouse models for certain studies

  • Mouse models with chimeric human/mouse TNFR1 have been developed to evaluate human-specific therapeutics

These differences are critical when interpreting translational research findings and developing therapeutic strategies.

What methodological approaches are most effective for studying TNFR1 versus TNFR2 functions in mouse models?

To effectively distinguish between TNFR1 and TNFR2 functions, researchers should consider:

Genetic Approaches:

  • Using receptor-specific knockout models (TNFR1-KO, TNFR2-KO, and double-knockout mice)

  • Conditional tissue-specific knockouts for targeted analysis

Pharmacological Approaches:

  • Selective TNFR1 antagonists such as domain antibodies (e.g., DMS5540)

  • TNFR2-specific agonists

  • Sequential treatment protocols with both agents

Readout Methods:

  • Cell death assays (e.g., L929 cytotoxicity assay for TNFR1 function)

  • Proliferation assays for TNFR2 activity

  • Signaling pathway analysis (NF-κB activation, MAPK pathways)

How should researchers design experiments to evaluate TNFR-mediated apoptosis in mouse models?

For robust apoptosis studies:

  • In vivo approaches:

    • TUNEL staining in tissue sections, as demonstrated in lung tissues after M. avium infection

    • Active caspase-3 immunostaining

    • Combined with receptor-specific knockouts to attribute effects

  • In vitro approaches:

    • L929 cytotoxicity assays for TNF-α-mediated effects

    • Primary cell isolation from receptor-specific knockout mice

    • RNA interference approaches with Raf siRNA to investigate downstream signaling

  • Important controls:

    • Include all relevant genotypes (WT, TNFR1-KO, TNFR2-KO, double-KO)

    • Time-course experiments to capture both early and late apoptotic events

    • Appropriate positive controls (known apoptosis inducers)

What are the optimal pharmacokinetic/pharmacodynamic (PK/PD) protocols for evaluating anti-TNFR therapeutics in mice?

Based on studies with the anti-TNFR1 domain antibody DMS5540:

PK Study Design:

  • Test multiple dose levels (e.g., 0.1, 1.0, and 10 mg/kg)

  • Monitor clearance patterns (linear vs. non-linear)

  • Consider the impact of albumin-binding domains for half-life extension

PD Measurements:

  • Measure total soluble TNFR1 levels as a biomarker of target engagement

  • Assess functional outcomes with challenge studies (e.g., TNF-α-induced IL-6 production)

  • Monitor disease-relevant parameters in specific models

Study Duration:

  • Design sampling schedules based on expected half-life

  • Include both acute and chronic dosing protocols when appropriate

How can TNFR mouse models best advance understanding of autoimmune disease mechanisms?

TNFR mouse models have provided critical insights into autoimmune disease mechanisms:

Experimental Autoimmune Encephalomyelitis (EAE):

  • Humanized TNFR mice show amelioration of disease with anti-TNFR1 antibody treatment

  • Sequential treatment with TNFR2 agonist followed by TNFR1 antagonist significantly reduces paralysis symptoms and demyelination compared to single treatments

  • Treatment effects are not mediated by altering immune cell subset frequencies but rather by modulating their tissue infiltration patterns

Key Considerations:

  • Timing of interventions is critical - TNFR2 stimulation before symptom onset improves response to subsequent TNFR1 antagonism

  • Cellular mechanisms include reduced immune cell infiltration across the blood-brain barrier

  • Evaluate both clinical scores and histopathological outcomes

How do TNFR1 and TNFR2 signaling pathways interact in inflammatory contexts?

The interplay between TNFR1 and TNFR2 signaling is complex:

Signaling Crosstalk:

  • TNFR1 promotes cell survival through activation of NF-κB downstream pathways

  • Raf kinase is a critical mediator of TNF-induced anti-apoptotic signaling in a Ras-independent manner

  • TNFR2 activation can modulate TNFR1-mediated inflammatory responses

Methodological Approaches:

  • Biochemical assays for pathway activation (Ras-GTP pulldown, Raf kinase assays)

  • Western blot analysis for phosphorylated signaling molecules

  • Combined genetic and pharmacological interventions to dissect pathway interactions

What are the current limitations of TNFR-humanized mouse models and how might they be addressed?

TNFR-humanized mouse models face several challenges:

Current Limitations:

  • Species-specific differences in downstream signaling components

  • Potential alterations in receptor cross-talk

  • Incomplete humanization of the entire TNF-TNFR system

Potential Solutions:

  • Development of more comprehensive humanized models incorporating multiple pathway components

  • Validation studies comparing responses in humanized mice to human cells/tissues

  • Combined in vitro human systems with in vivo mouse models for translational research

How can researchers overcome challenges in targeting TNFR1 without inducing receptor agonism?

Anti-TNFR1 antibodies can potentially induce receptor agonism, complicating research approaches. Solutions include:

  • Use of monovalent domain antibodies (dAbs) like DMS5540, which demonstrated pure antagonist properties

  • Careful epitope selection - targeting sites that don't induce receptor clustering

  • Testing for agonist activity in sensitive bioassays before proceeding to in vivo studies

Interestingly, some anti-TNFR1 dAbs don't compete with TNF-α for receptor binding but still function as antagonists, suggesting allosteric inhibition mechanisms .

What strategies help distinguish between TNFR-mediated effects in different cell populations?

To attribute TNFR effects to specific cell populations:

  • Conditional knockout approaches:

    • Cell-type specific Cre drivers (e.g., villin-Cre for intestinal epithelium)

    • Inducible systems for temporal control

  • Ex vivo analysis:

    • Flow cytometry to assess receptor expression and responses in specific cell subsets

    • Cell sorting followed by functional assays

  • In vitro validation:

    • Primary cell cultures from relevant knockout models

    • Reconstitution studies (e.g., TNFR1 addback cells generated through retroviral infection)

How should researchers address contradictory findings between mouse TNFR studies and human clinical outcomes?

When faced with translational discrepancies:

  • Critical analysis of model systems:

    • Consider species-specific differences in TNFR expression and signaling

    • Evaluate the fidelity of disease models to human pathology

  • Complementary approaches:

    • Use humanized mouse models when appropriate

    • Confirm key findings in human cells/tissues

    • Consider combinatorial approaches (e.g., sequential TNFR targeting)

  • Mechanistic investigations:

    • Focus on shared downstream pathways rather than receptor-level effects

    • Identify biomarkers that translate between species

How might selective TNFR targeting advance therapeutic approaches for inflammatory diseases?

Selective TNFR targeting offers several advantages over pan-TNF inhibition:

Current Evidence:

  • Anti-TNFR1 therapies effectively modulate immune responses in autoimmune disease models

  • Sequential TNFR2 agonism followed by TNFR1 antagonism provides superior efficacy in EAE compared to either approach alone

  • TNFR1 antagonism may preserve beneficial TNFR2-mediated tissue repair functions

Potential Applications:

  • Multiple sclerosis and other CNS inflammatory conditions

  • Inflammatory bowel diseases

  • Rheumatoid arthritis and related disorders

What novel tools are being developed to study TNFR biology in mouse models?

Emerging research tools include:

  • Advanced antibody formats:

    • Domain antibodies with albumin-binding domains for extended half-life

    • Bispecific antibodies targeting multiple pathway components

  • Genetic engineering approaches:

    • CRISPR/Cas9-mediated receptor modifications

    • Reporter systems for real-time monitoring of TNFR activation

  • Imaging technologies:

    • In vivo imaging of TNFR activation and signaling

    • Spatial transcriptomics to map receptor activities in tissues

How do TNFR-mediated mechanisms contribute to tissue-specific disease processes?

TNFR signaling exhibits tissue-specific effects:

CNS Autoimmunity:

  • TNFR1 antagonism reduces immune cell infiltration across the blood-brain barrier

  • TNFR2 agonism promotes Treg accumulation in the CNS, while TNFR1 antagonism increases T-cell infiltration and B-cell cuffing at perivascular sites

Intestinal Inflammation:

  • TNFR1 activation promotes colon epithelial cell survival through NF-κB

  • Raf expression is required for TNFR-induced cell survival in intestinal epithelium

Infectious Disease:

  • TNFR1-mediated apoptosis is protective against Mycobacterium avium infection in lung tissues

Understanding these tissue-specific mechanisms is crucial for developing targeted therapeutic approaches.

Product Science Overview

Structure and Function

TNFR belongs to the tumor necrosis factor receptor superfamily, which includes several receptors that bind to tumor necrosis factors (TNFs). These receptors are involved in various cellular processes, including cell proliferation, differentiation, and death. The TNFR family is characterized by the presence of cysteine-rich domains in their extracellular regions, which are crucial for ligand binding.

Types of TNFR

There are two main types of TNFRs:

  1. TNFR1 (p55/p60): This receptor is ubiquitously expressed and can bind both soluble and membrane-bound TNF-α. It is involved in mediating most of the TNF-α-induced cellular responses, including apoptosis and inflammation.
  2. TNFR2 (p75/p80): This receptor is primarily expressed on immune cells and binds preferentially to membrane-bound TNF-α. It plays a role in immune regulation and tissue regeneration.
Recombinant TNFR in Mice

Recombinant TNFR proteins are produced using genetic engineering techniques, where the TNFR gene is inserted into an expression vector and introduced into host cells (such as bacteria or mammalian cells) to produce the protein. The recombinant TNFR (mouse) is used in various research applications to study its role in immune responses and disease mechanisms.

Applications in Research and Medicine
  1. Cancer Research: TNFRs are studied for their role in tumor progression and metastasis. For instance, TNFRSF25, a member of the TNFR superfamily, has been shown to promote tumor regression when targeted by agonistic antibodies .
  2. Inflammatory Diseases: TNFRs are involved in the pathogenesis of several inflammatory diseases. Recombinant TNFRs are used to investigate their potential as therapeutic targets for conditions like rheumatoid arthritis and inflammatory bowel disease .
  3. Immunotherapy: TNFRs are explored for their potential in immunotherapy. For example, TNFRSF25 agonists have shown promise in enhancing T-cell responses and inhibiting tumor growth .

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 2025 TheBiotek. All Rights Reserved.