TNFR2 Human, His is available in multiple formats:
Glycosylation: Mammalian-expressed variants (e.g., HEK293) are glycosylated, increasing molecular weight (~55 kDa vs. ~24.45 kDa for E. coli variants) .
Functionality: Fc fusion proteins improve solubility and enable bivalent binding, enhancing TNFR2 signaling .
TNFR2 Human, His is pivotal in studying TNFR2’s role in immune modulation:
SPR Studies: Used to measure binding kinetics between TNFR2 and TNF-α or engineered ligands (e.g., scR2agoTNF-Fc) .
Treg Activation: TNFR2 Human, His is immobilized to assess ligand-induced signaling in Tregs, enhancing IL2-driven proliferation via non-canonical NF-κB pathways .
Treg Expansion: TNFR2 agonists (e.g., D18, scR2agoTNF-Fc) expand Tregs ex vivo, improving suppressive function in autoimmune models .
Cancer Immunology: TNFR2 Human, His helps study Treg infiltration in tumors, where TNFR2+ Tregs correlate with poor prognosis .
TNFR2 Human, His has driven the development of:
TNFR2 Agonists: To treat autoimmune diseases (e.g., type 1 diabetes) by expanding Tregs .
TNFR2 Antagonists: To block Treg infiltration in tumors, enhancing antitumor immunity .
Adoptive Treg Therapy: Optimizing ex vivo Treg expansion using TNFR2 agonists for graft-versus-host disease (GvHD) or organ rejection .
TNFR2 (also known as TNFRSF1B or CD120b) is a type I transmembrane protein and member of the TNF receptor superfamily (TNFRSF). It belongs to the TRAF (TNF-receptor-associated factor)-interacting subgroup of TNFRSF . Human TNFR2 protein typically comprises amino acids 23-257 of the sequence encoded by GenBank Accession No. NM_001066 . Unlike TNFR1, TNFR2 lacks an intracellular death domain, which results in fundamentally different signaling outcomes. His-tagged versions often include a C-terminal histidine tag to facilitate purification and detection, with recombinant forms frequently designed as fusion proteins with molecular weights around 55 kDa .
While TNFR1 is the dominant receptor for pro-inflammatory responses, TNFR2 has more complex and context-dependent signaling outcomes . TNFR2 activates both canonical and non-canonical NF-κB pathways. In the canonical pathway, TNFR2 ligation leads to IκBα degradation . Additionally, TNFR2 induces transcription of NFKB2 and RELB genes, which encode proteins forming the non-canonical NF-κB transcription factor . TNFR2 signaling in regulatory T cells (Tregs) enhances their proliferation and stability through the PI3-kinase (PI3K)-Akt-mammalian target of rapamycin (mTOR) pathway . Unlike TNFR1, TNFR2 is preferentially activated by membrane-bound TNF rather than soluble TNF .
TNFR2 expression is typically high in myeloid cells but is also found in certain T and B cell subsets and some non-immune cells such as endothelial cells, glial cells, and cardiomyocytes . Regulatory T cells (Tregs) constitutively express high levels of TNFR2 . In unstimulated conditions, TNFR2 is primarily expressed by CD4+Foxp3+ Treg cells, but not by other evaluated lymphocyte populations . Following stimulation with PHA-P/IL-2, TNFR2 is additionally expressed by CD4+Foxp3− and CD8+ effector T cells, and NK cells . Antigen-specific CD8+ T cells, such as those responding to CMV pp65, can display particularly high intensity TNFR2 expression . Mouse NK cell TNFR2 expression differs from human patterns, highlighting important species differences in TNFR2 biology .
His-tagged human TNFR2 can be efficiently purified using immobilized metal affinity chromatography (IMAC). For optimal purification:
Express the protein in an appropriate system (HEK293 cells are commonly used for TNFR2)
Use a buffer system containing 8 mM phosphate, pH 7.4, with 110 mM NaCl for stability
Verify purity using SDS-PAGE (TNFR2-Fc-His fusion proteins typically appear around 55 kDa)
Validate functionality through binding assays with TNF-alpha
Essential validation steps include functional assessment using chemiluminescent binding assays with biotinylated TNF-alpha to generate inhibition curves, confirming proper folding and biological activity of the purified protein .
Multiple complementary approaches can be used to detect TNFR2 expression in various sample types:
Flow cytometry:
Immunohistochemistry:
Use validated anti-TNFR2 antibodies
Consider dual staining with cell-type specific markers
Particularly valuable for tissue localization studies
Western blotting:
For cerebrospinal fluid (CSF) analysis, measuring soluble TNFR2 (sTNFR2) can provide insights into TNFR2-related processes in neurological conditions like Alzheimer's disease .
When designing experiments with His-tagged TNFR2, several critical controls should be included:
Binding specificity controls:
Irrelevant His-tagged protein of similar size
Unlabeled TNF for competitive binding
TNFR1-specific controls to distinguish receptor-specific effects
Functional assay controls:
Pathway inhibitor controls (NF-κB, PI3K-Akt-mTOR)
Cell-type controls (TNFR2-expressing vs. non-expressing cells)
Multiple timepoints to capture both early and late signaling events
Technical controls:
Endotoxin testing to prevent confounding inflammatory effects
Concentration gradient to establish dose-response relationships
Lot-to-lot consistency validation
TNFR2: TNF-alpha binding inhibition assays using biotinylated TNF-alpha with chemiluminescent detection provide a robust readout for functional validation .
TNFR2 plays multiple critical roles in Treg biology through several mechanisms:
NF-κB pathway activation:
Canonical pathway: TNFR2 ligation activates canonical NF-κB signaling via IκBα degradation
Non-canonical pathway: TNFR2 induces transcription of NFKB2 and RELB genes
TNFR2 ligation in combination with IL-2 stabilizes NF-κB-inducing kinase (NIK) protein
RelB translocation from cytosol to nucleus occurs following TNFR2 stimulation
Epigenetic regulation:
Proliferative effects:
Inhibition of RelB nuclear translocation blocks the proliferative response, demonstrating that the non-canonical NF-κB pathway is crucial for TNFR2-mediated Treg expansion .
Developing effective TNFR2-specific agonists involves several strategic approaches:
Fusion protein design:
Structure-guided design:
Validation methodology:
In vivo validation:
The design of NewSTAR2 exemplifies this approach, demonstrating how a single injection can dramatically enhance Treg numbers and function in vivo .
TNFR2 signaling exhibits significant cell type-specific differences:
Expression patterns:
Functional outcomes:
In Tregs: TNFR2 activation enhances suppressive function and proliferation
In CD8+ T cells: TNFR2 can mediate activation-induced cell death (AICD) through reactive oxygen species production
In activated CD8+ effector T cells: TNFR2 activation increases memTNF expression, which can in turn promote Treg proliferation via TNFR2
Molecular mechanisms:
These differences create a complex regulatory network where TNFR2 can simultaneously enhance Treg-mediated suppression and modulate effector T cell responses in a context-dependent manner.
TNFR2 plays complex roles in cancer biology with therapeutic implications:
Expression patterns in tumors:
Functional effects:
Immunotherapy approaches:
Researchers have found that TNFR2 emerged as a key target from unbiased screens, suggesting its fundamental importance in tumor immunology and potential as a therapeutic target .
TNFR2 shows emerging significance in neurodegenerative diseases, particularly Alzheimer's disease:
Genetic associations:
Mechanistic insights:
Clinical implications:
These findings highlight TNFR2 as an important focus for understanding and potentially treating neurodegenerative diseases through novel mechanisms beyond traditional amyloid and tau pathways .
TNFR2-targeted therapies show promise for autoimmune and inflammatory conditions:
Graft-versus-host disease (GvHD):
Inflammatory modulation:
Cell therapy applications:
Other immune-regulatory cell types:
These findings suggest that targeted TNFR2 activation could provide novel therapeutic avenues for treating inflammatory and autoimmune conditions through multiple cellular mechanisms.
Reconciling contradictory findings on TNFR2 requires systematic consideration of experimental variables:
Cell type considerations:
TNFR2 functions differently across cell types (Tregs vs. effector T cells vs. non-immune cells)
Expression levels vary dramatically between populations
Cell purity is critical - minor contaminating populations can skew results
Experimental conditions:
TNF concentration affects TNFR1 vs. TNFR2 activation balance
Soluble vs. membrane-bound TNF preferentially activates different receptors
Culture conditions (serum, cytokines) modulate TNFR2 expression and signaling
Species differences:
Methodological approach to reconciliation:
These considerations help explain why TNF has been variably reported to promote, inhibit, or have no effect on Treg function in different experimental systems .
Investigating TNFR2 signaling pathway cross-talk presents several technical challenges:
Receptor specificity:
Temporal dynamics:
TNFR2 activates multiple pathways with different kinetics
Canonical NF-κB signaling occurs rapidly
Non-canonical NF-κB and other pathways develop more slowly
Comprehensive temporal analysis is required
Pathway interconnections:
In vivo translation:
Addressing these challenges requires innovative approaches combining genetic models, highly selective pharmacological tools, and advanced analytical techniques.
Several promising research directions are emerging for TNFR2:
Enhanced therapeutic design:
Cell-specific targeting:
Exploring differential effects on Tregs vs. effector cells
Developing strategies to selectively target TNFR2 on specific cell populations
Understanding cell type-specific signaling networks
Combinatorial approaches:
Novel disease applications:
These emerging directions highlight the continued importance of TNFR2 as a research focus with significant therapeutic potential across multiple disease areas.
Tumor Necrosis Factor Receptor Type 2 (TNFR2), also known as Tumor Necrosis Factor Receptor Superfamily Member 1B (TNFRSF1B) and CD120b, is one of the two primary receptors that bind to Tumor Necrosis Factor-alpha (TNFα). TNFR2 is a membrane receptor that plays a crucial role in various physiological and pathological processes, including immune response, inflammation, and tumor progression .
TNFR2 consists of an extracellular region with four cysteine-rich domains that facilitate binding to TNFα. Unlike its counterpart, Tumor Necrosis Factor Receptor Type 1 (TNFR1), TNFR2 lacks a death domain (DD) in its intracellular structure, which results in different signaling pathways and functions when bound to TNFα .
TNFR2 is expressed in various tumor cells and certain immune cells, such as regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). It contributes significantly to the tumor microenvironment by promoting tumor cell growth, activating immunosuppressive cells, and supporting immune escape . TNFR2’s activation mechanism involves recruiting a complex composed of adapter proteins, such as TNF receptor-associated factor 2 (TRAF2) and apoptosis-related markers like cIAP1/2 .
The dual role of TNFR2 in promoting both tumor progression and immune regulation makes it a promising target for cancer therapy. Antagonists and agonists targeting TNFR2 have shown significant anti-tumor activity in preclinical studies . These therapeutic agents can either inhibit the immunosuppressive functions of TNFR2 or enhance its ability to stimulate cytotoxic T-cells, thereby providing a multifaceted approach to cancer treatment .
Human recombinant TNFR2 with a His tag is a laboratory-produced version of the receptor, designed for research and therapeutic purposes. The His tag, a sequence of histidine residues, facilitates the purification and detection of the recombinant protein. This recombinant form is used in various studies to understand the receptor’s function, signaling pathways, and potential as a therapeutic target .