Recombinant Human Tumor necrosis factor receptor superfamily member 9 (TNFRSF9), partial (Active)

Shipped with Ice Packs
In Stock

Description

Functional Characteristics

The protein retains high biological activity, demonstrated through ligand-binding assays:

ParameterDetails
Ligand BindingBinds TNFSF9 (4-1BBL) with EC₅₀ of 1.011–2.429 ng/mL
Receptor DimerizationDisulfide-linked dimerization enhances signaling via CRD4
Signaling PathwaysActivates TRAF1/2, promoting Bcl-2 family survival genes and inhibiting Bim

Crystallographic studies reveal TNFRSF9 forms a 2D network with TNFSF9, amplifying co-stimulatory signals in T cells . This structural insight supports its role in immune modulation.

Applications in Research and Therapeutics

TNFRSF9 is pivotal in cancer immunotherapy and autoimmune studies:

Cancer Immunotherapy

  • CAR-T Cell Therapy: Enhances T-cell persistence and anti-tumor efficacy by incorporating TNFRSF9 costimulation .

  • Bispecific Antibodies: Targets TNFRSF9 to activate tumor-infiltrating T cells while blocking PD-1/PD-L1 .

Autoimmune and Inflammatory Diseases

  • Modulation of Regulatory T Cells: Balances immune responses by influencing Treg activity .

Key Research Findings

  1. Structural Insights:

    • TNFRSF9 forms a disulfide-linked dimer, enabling cross-linking with TNFSF9 trimers to create a 2D signaling network .

    • The DE loop lacks a conserved tyrosine, suggesting a unique binding mechanism compared to other TNFRs .

  2. Clinical Relevance:

    • Deficiencies in TNFRSF9 impair EBV-specific T-cell responses, linking it to chronic viral infections .

    • Overexpression enhances T-cell proliferation and CD25 expression, critical for anti-tumor immunity .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered 1xPBS, pH 7.4.
Form
Lyophilized powder
Lead Time
Generally, we can ship the products within 5-10 business days after receiving your orders. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery times.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend that this vial be briefly centrifuged prior to opening to bring the contents to the bottom. Please reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final concentration of glycerol is 50%. Customers can use this as a reference.
Shelf Life
The shelf life is influenced by various factors, including storage conditions, buffer ingredients, storage temperature, and the intrinsic stability of the protein.
Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. The shelf life of the lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt, aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
C-terminal hFc-tagged
Synonyms
4 1BB; 4 1BB ligand receptor; 4-1BB ligand receptor; 4-1BB Ligand Receptor T Cell; 4-1BB, mouse, homolog of; Antigen 4-1BB Homolog; CD 137; CD137; CD137 antigen; CDw137; HLDA VI; Homolog of mouse 4 1BB; ILA; induced by lymphocyte activation (ILA); Induced by lymphocyte activation; Interleukin activated receptor homolog of mouse Ly63; Ly63, mouse, homolog of; MGC2172; OTTHUMP00000044294; Receptor protein 4 1BB; T cell antigen 4 1BB homolog; T cell antigen ILA; T-cell antigen 4-1BB homolog; T-cell antigen ILA; TNF receptor superfamily member 9; TNFRSF9; TNR9_HUMAN; Tumor necrosis factor receptor superfamily member 9
Datasheet & Coa
Please contact us to get it.
Expression Region
24-186aa
Mol. Weight
44.2 kDa
Protein Length
Extracellular Domain
Purity
Greater than 95% as determined by SDS-PAGE.
Research Area
Cancer
Source
Mammalian cell
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function
Receptor for TNFSF9/4-1BBL. It may play a role in T cell activation.
Gene References Into Functions
  1. A study on a northern Chinese Han population investigated the association of 3 SNPs (rs161827, rs161818, and rs161810) in the CD137 gene with ischemic stroke. rs161827 showed a significant difference between patients with and without diabetes and the controls. rs161818 and rs161810 differed significantly between patients without diabetes and the controls. All 3 SNPs were statistically significant in the combined stroke group. PMID: 28755037
  2. This study introduces LOAd703, a designed adenovirus equipped with trimerized CD40L and 4-1BBL, which activates the CD40 and 4-1BB pathways, respectively. PMID: 28536305
  3. Continuous 4-1BB costimulation in chimeric antigen receptors hinders T cell survival and is vector-dependent. PMID: 28978471
  4. Cetuximab-mediated NK-cell expression of CD137 on tumor-infiltrating lymphocytes is dependent on FcgammaRIIIa polymorphism. In patients with head and neck cancer receiving neoadjuvant cetuximab treatment, upregulation of CD137 by intratumoral, cetuximab-activated NK cells correlated with FcgammaRIIIa V/F polymorphism and predicted clinical response. PMID: 27496866
  5. This research systematically assessed a series of CAR constructs targeting glypican-3 (GPC3), which is specifically expressed on various solid tumors. The study compared GPC3-specific CARs encoding CD3zeta (Gz) alone or with costimulatory domains derived from CD28 (G28z), 4-1BB (GBBz), or CD28 and 4-1BB (G28BBz). PMID: 27530312
  6. 4-1BB and 4-1BBL are potential markers for predicting patient outcomes and represent a valuable target for screening patients with acute myeloid leukemia at the initial diagnosis. PMID: 27388616
  7. This study further investigated the role of CD137-CRDI (cysteine rich domain I) in the binding of CD137-CD137L. PMID: 27430526
  8. The cell surface proteins LAG-3 and 4-1BB, driven by Egr2, can identify dysfunctional tumor antigen-specific CD8(+) TIL. PMID: 28115575
  9. These findings indicate that the CD137 antigen is a valuable marker for identifying Mycobacterium tuberculosis (Mtb)-reactive CD4(+) T cells (Mtb-reactive CD4(+) T cells) using flow cytometry. PMID: 28218958
  10. Anti-4-1BB single chain variable fragments enhanced surface CD69 expression and interleukin-2 production in stimulated CCRF-CEM cells, confirming the agonistic effect of the selected single chain variable fragments. The data from this study provide a basis for further investigations involving the biological functions of anti-4-1BB single chain variable fragments in future research. PMID: 28347235
  11. Studies suggest that adoptive T cell therapy and CD137 antigen hold considerable potential to enhance the efficacy of current cancer immunotherapies. PMID: 26970765
  12. In complex with T cell receptor, it promotes memory T cells, cell respiration, fatty acid oxidation, and mitochondrial biogenesis. PMID: 26885860
  13. These studies provide the first direct evidence that ligation of tumor necrosis factor superfamily members and their cognate receptors is crucial for controlling viral lytic replication. PMID: 26467721
  14. Our findings reveal a novel, TNFRSF9-positive, reactive astrocytic phenotype in human gliomas. PMID: 24606203
  15. Human genetic evidence supports the involvement of CD137 in atherosclerosis. PMID: 25032953
  16. Upon activation, transferred human T lymphocytes express the inducible surface antigens hPD-1 and hCD137 on their plasma membrane. PMID: 26113085
  17. Our findings provide biological explanations for the antitumor effects of CD19 CARs and for the observations that CD19 CAR T cells incorporating the 4-1BB costimulatory domain are more persistent than those incorporating CD28 in clinical trials. PMID: 25939063
  18. Upregulation of CD137 expression through LMP1 by EBV promotes cell survival in T or NK cells. PMID: 25409517
  19. Based on CD137 or CD154 expression. PMID: 25367298
  20. High expression of CD137 is associated with type 1 diabetes. PMID: 24797972
  21. DENV C disrupts Daxx and NF-kappaB interaction to induce CD137-mediated apoptosis during DENV infection. PMID: 25019989
  22. The action of agonist anti-4-1BB in suppressing autoimmune and allergic inflammation was completely dependent on Galectin-9 (Gal-9). Gal-9 directly bound to 4-1BB, in a site distinct from the binding site of antibodies and the natural ligand of 4-1BB. PMID: 24958847
  23. CD137 plays a role in the immunobiology of human cancer where it is preferentially expressed on a tumor-reactive subset of tumor-infiltrating lymphocytes. PMID: 24045181
  24. Monocytes interact with iNKT cells to increase expression of 4-1BBL and 4-1BB, and in conjunction with this pathway, maintain their numbers at baseline. PMID: 24639347
  25. Findings show that immunohistochemistry for CD137L is capable of reliably distinguishing small B-cell lymphomas from reactive lymphoid aggregates. PMID: 24746207
  26. Dengue virus induces CD137 signaling to enhance apoptosis by increasing TNF-alpha production via activation of p38 MAPK. PMID: 23557259
  27. This is the first study to demonstrate that this member of the TNF superfamily, CD137, is modulated by SAHA treatment in breast cancer cells. PMID: 22797667
  28. The CD137 multi-parameter flow cytometry fast assay allows for phenotypic and functional determination of alloreactive precursor frequencies of both CD4+ and CD8+ T cells with high sensitivity and specificity. PMID: 23750604
  29. Co-stimulation through 4-1BB/CD137 improves the expansion and function of CD8(+) melanoma tumor-infiltrating lymphocytes for adoptive T-cell therapy. PMID: 23560068
  30. Taken together, these data provide evidence that the 4-1BB signal is an important regulator of gammadelta T cells. PMID: 23640752
  31. The mechanisms that account for the effect of CD137 signaling on TNF-alpha production were based on a decrease of TNF-alpha production by antigen presenting cells (APCs) and, potentially, on an increase in APC apoptosis. PMID: 23437083
  32. Our results uncover a new regulatory mechanism for CD137L expression that mediates immune escape by HRS cells, and they identify CD137 as a potential target for immunotherapy of Hodgkin lymphoma. PMID: 23204227
  33. Patients with head and neck cancer have reduced levels of alternative co-stimulatory receptors OX40 and 4-1BB. PMID: 22204816
  34. 4-1BB (CD137), along with CD103, marks mesenteric lymph node dendritic cells (DCs) with the highest level of retinal dehydrogenase (RALDH) activity, and ligation of 4-1BB maintains RALDH expression in these gut DCs. PMID: 22896640
  35. CD137 protein is expressed by a specific group of hematolymphoid tumors, including classical Hodgkin lymphoma, T-cell and NK/T-cell lymphomas, and follicular dendritic cells neoplasms. PMID: 22901750
  36. Treatment with a CD137 agonistic antibody induces CCL21 expression and DC accumulation near lymphatic vessels. Collectively, our results demonstrate that the inflammatory function of lymphatic vessels can be regulated by CD137. PMID: 22593548
  37. CD137:CD137L interactions regulate the innate and adaptive immune response of the host against M. tuberculosis. PMID: 21747409
  38. There is a significantly positive correlation between CD137 expression and complex coronary stenosis morphology. PMID: 21396356
  39. Data suggest that 4-1BBL mediates NK-cell immunosubversion in CLL, which may contribute to the reported limited efficacy of Rituximab in inducing NK-cell reactivity in the disease. PMID: 22144129
  40. CD137 activity is directly proportional to colorectal cancer stage. Surgical resection of the tumor results in increased CD134 and CD137 expression. PMID: 22343199
  41. We demonstrate that the inflammatory and cytotoxic function of CD4(+)CD28(null) T cells can be inhibited by blocking OX40 and 4-1BB costimulatory receptors. PMID: 22282196
  42. The sCD137 levels correlate with the likelihood of complications and mortality. The association of sCD137, a product of activated T cells, with the severity of acute pancreatitis suggests that T cells contribute to the pathogenesis of acute pancreatitis. PMID: 21963611
  43. CD137 plays a role in breast cancer, and its specific antibody can be used to enhance trastuzumab efficacy. PMID: 22326955
  44. Conditioned medium from Lewis Lung Carcinoma cells caused significant upregulation of 4-1BB in mast cells. PMID: 22343053
  45. Data indicate that ex4-1BBL augments 4-1BB expression not only on the primed T cell but also on DCs. PMID: 21745658
  46. The measurement of a single gene expressed by tumor cells (LMO2) and a single gene expressed by the immune microenvironment (TNFRSF9) strongly predicts overall survival in patients with diffuse large B-cell lymphoma. PMID: 21670469
  47. This work is the first to demonstrate the contribution of CD137 signaling to DENV-mediated apoptosis. PMID: 21669186
  48. CD137 ligand can also be expressed as a transmembrane protein on the cell surface and transmit signals into the cells on which it is expressed (reverse signaling). PMID: 20643812
  49. Results suggest a two-step model of M cell differentiation, with initial CD137-independent commitment to the M cell lineage followed by CD137-CD137L interaction of M cells with CD137-activated B cells or dendritic cells for functional maturation. PMID: 20616340
  50. Data support a role for CD137 in the recruitment of monocytes to inflammatory tissues. PMID: 20347151

Show More

Hide All

Database Links

HGNC: 11924

OMIM: 602250

KEGG: hsa:3604

STRING: 9606.ENSP00000366729

UniGene: Hs.738942

Subcellular Location
Membrane; Single-pass type I membrane protein.
Tissue Specificity
Expressed on the surface of activated T-cells.

Q&A

What is TNFRSF9 and what are its alternative nomenclatures in scientific literature?

TNFRSF9, also known as 4-1BB, CD137, CDw137, and ILA (Induced By Lymphocyte Activation), is an inducible co-stimulatory receptor primarily expressed on activated T cells. It functions as a critical mediator in T cell activation, proliferation, differentiation, and apoptosis processes. TNFRSF9 also plays significant roles in the pathogenesis of certain autoimmune diseases and mediates anti-tumor immune responses of T cells and NK cells .

Researchers should be aware that literature searches may require using multiple nomenclatures to ensure comprehensive review of existing research. When reporting experimental findings, it is recommended to include all relevant nomenclatures in keywords and abstracts to enhance discoverability.

What is the molecular structure of recombinant human TNFRSF9 protein used in research applications?

Commercially available recombinant human TNFRSF9 typically consists of amino acids Leu24-Gln186 of the native protein. The recombinant form often contains an N-terminal linker and a C-terminal tag (commonly a 10xHis-tag) when expressed in mammalian cells. The protein has an apparent molecular weight of approximately 28 kDa on gel electrophoresis and demonstrates high purity (>95%) with low endotoxin content (<1EU/μg) in research-grade preparations .

For functional validation, recombinant TNFRSF9 can bind to its ligand TNFSF9 with EC50 values ranging from 1.011-3.702 ng/mL in ELISA assays, confirming biological activity suitable for immune and antibody screening experiments .

How should researchers assess functional activity of recombinant TNFRSF9 preparations?

Methodological approach for functional assessment:

  • Perform binding assays with natural ligand TNFSF9 using ELISA to determine EC50 values

  • Verify protein integrity via Western blot analysis under reducing and non-reducing conditions

  • Conduct cellular activation assays using TNFRSF9-responsive T cell lines

  • Analyze downstream signaling events (NF-κB activation, cytokine production)

  • Compare activity with established reference standards

Functional recombinant TNFRSF9 should demonstrate consistent binding kinetics with its ligand and induce expected biological responses in relevant cell systems .

How does TNFRSF9 deficiency impact immune responses, particularly against viral pathogens?

TNFRSF9 deficiency presents a critical research model for understanding this receptor's immunological significance. Case studies of biallelic TNFRSF9 mutations reveal that CD137 deficiency severely compromises host defense against viral infections, particularly Epstein-Barr virus (EBV).

Immunological consequences include:

  • Markedly reduced or abrogated expression of CD137 on activated T, B, and NK cells

  • Impaired CD8+ T cell activation and function

  • Reduced expression and release of critical cytokines (IFN-γ, TNF-α)

  • Diminished expression of cytolytic molecules (perforin, granzyme B)

  • Compromised cytotoxic activity against infected cells

These observations highlight TNFRSF9's essential role in anti-viral immunity, particularly for controlling persistent viral infections. Researchers investigating primary immunodeficiencies should consider TNFRSF9 deficiency in patients presenting with severe EBV-associated lymphoproliferative disorders (LPD) .

What experimental approaches can elucidate TNFRSF9 signaling mechanisms in immune cells?

To comprehensively investigate TNFRSF9 signaling pathways, researchers should employ a multi-modal approach:

  • Genetic manipulation techniques:

    • CRISPR/Cas9-mediated gene editing of TNFRSF9 or downstream effectors

    • Overexpression systems using wildtype or mutant TNFRSF9 constructs

    • siRNA/shRNA knockdown for transient signaling studies

  • Biochemical signaling analysis:

    • Phosphoproteomic analysis of downstream effectors (p38MAPK pathway)

    • Co-immunoprecipitation to identify novel interaction partners

    • Western blot analysis of activated signaling components

  • Functional readouts:

    • Cytokine production profiling (IFN-γ, TNF-α, IL-2)

    • Cytotoxicity assays against relevant targets

    • Proliferation and survival assays following TNFRSF9 engagement

  • Advanced imaging:

    • Live-cell imaging of TNFRSF9 clustering and immunological synapse formation

    • Super-resolution microscopy to visualize receptor distribution patterns

Research has established that TNFRSF9 signaling impacts p38MAPK phosphorylation, which subsequently regulates PAX6 expression. This signaling axis represents a targetable pathway in diseases where TNFRSF9 function is dysregulated .

How does TNFRSF9 expression influence tumor progression and what are the underlying mechanisms?

TNFRSF9 demonstrates significant tumor-suppressive functions in several cancer models, particularly breast cancer. The mechanisms through which TNFRSF9 regulates tumor progression include:

  • Regulation of the p38MAPK/PAX6 signaling axis:

    • TNFRSF9 downregulation increases phosphorylated p38 (p-p38) levels

    • Elevated p-p38 subsequently upregulates PAX6 expression

    • PAX6 promotes tumor cell proliferation and invasion

    • P38 phosphorylation inhibitors can reverse these effects, suppressing proliferation and invasion while promoting apoptosis

  • Expression patterns in tumor microenvironments:

    • TNFRSF9 is frequently downregulated in breast cancer tissues and cell lines

    • Knockdown of TNFRSF9 promotes breast cancer cell development

    • In melanoma brain metastases, TNFRSF9 is expressed on tumor cells, endothelial cells, and tumor-infiltrating lymphocytes

  • Spatial expression patterns:

    • In some tumors, TNFRSF9 expression increases with distance from blood vessels

    • This pattern may be linked to hypoxia-driven regulation of TNFRSF9

    • The relationship between hypoxia and TNFRSF9 expression warrants further investigation

These findings position TNFRSF9 as a potential biomarker and therapeutic target in cancer research.

What methodological approaches should researchers use to study TNFRSF9 expression in tumor samples?

When investigating TNFRSF9 expression in tumor tissues, researchers should implement a comprehensive analysis strategy:

  • Tissue preparation and analysis:

    • Formalin-fixed paraffin-embedded (FFPE) and fresh-frozen tissue processing

    • Laser capture microdissection for isolation of specific cellular compartments

    • Multiplex immunohistochemistry to simultaneously assess TNFRSF9 expression with other markers

    • Spatial transcriptomics to map expression patterns relative to vascular structures and hypoxic regions

  • Expression quantification:

    • Digital pathology with automated quantification algorithms

    • Cell-type specific scoring systems (tumor cells vs. endothelial cells vs. immune infiltrates)

    • Gradient analysis to assess expression patterns relative to blood vessels or hypoxic regions

  • Correlation with clinical parameters:

    • Tumor size, grade, and stage

    • Mutation status of relevant cancer genes (e.g., BRAF V600E in melanoma)

    • Patient survival and treatment response data

    • Immune infiltration patterns and markers

  • Epigenetic analysis:

    • DNA methylation profiling of the TNFRSF9 promoter

    • Chromatin accessibility studies (ATAC-seq)

    • Histone modification analysis at the TNFRSF9 locus

This systematic approach enables comprehensive characterization of TNFRSF9 expression patterns in tumors and facilitates identification of clinically relevant associations .

How is TNFRSF9 being utilized in CAR-T cell therapy design and what are the key considerations?

TNFRSF9 (CD137/4-1BB) has emerged as a crucial component in second-generation chimeric antigen receptor (CAR) T cell therapy designs. Researchers should consider the following aspects when incorporating TNFRSF9 into CAR constructs:

  • Structural integration:

    • Position of the TNFRSF9 co-stimulatory domain relative to CD3ζ signaling domain

    • Optimization of linker sequences between domains

    • Potential for domain mutations to enhance or modulate signaling strength

  • Functional characteristics of TNFRSF9-containing CARs:

    • Enhanced T cell persistence compared to CD28-based CARs

    • Balanced effector-memory phenotype development

    • Reduced T cell exhaustion during repeated antigen exposure

    • Distinct cytokine production profiles compared to other co-stimulatory domains

  • Experimental validation approaches:

    • In vitro cytotoxicity against target cells expressing varying antigen densities

    • Long-term persistence assays in serial re-stimulation models

    • Detailed phenotypic characterization using multiparameter flow cytometry

    • In vivo efficacy and persistence in relevant preclinical models

  • Clinical translation considerations:

    • Optimized manufacturing protocols specific for TNFRSF9-containing CARs

    • Monitoring for unique toxicity profiles

    • Biomarkers to predict and assess response to TNFRSF9-based CAR-T cells

TNFRSF9 incorporation in CAR-T cells has demonstrated significant advantages for treating certain malignancies, particularly in terms of long-term persistence and memory formation, making it a valuable component in current CAR designs .

What are the emerging strategies for therapeutic targeting of TNFRSF9 in cancer immunotherapy?

Several innovative approaches are being explored for therapeutic targeting of TNFRSF9 in cancer immunotherapy:

  • Agonistic antibodies:

    • Development of antibodies with optimal binding kinetics and epitope specificity

    • Engineering of antibody Fc regions to enhance or minimize FcγR engagement

    • Bispecific antibody formats linking TNFRSF9 engagement to tumor targeting

  • Targeted delivery strategies:

    • Antibody-cytokine fusion proteins (immunocytokines)

    • Tumor-targeting scFv:TNF fusion proteins that enhance TNFRSF9 signaling

    • Protease-activated prodrug approaches using tumor-selective proteases

    • Hypoxia-responsive TNFRSF9 targeting agents based on observed hypoxia-driven expression

  • Combination therapeutic approaches:

    • Synergistic pairing with checkpoint inhibitors (anti-PD-1, anti-CTLA-4)

    • Combination with conventional therapies (radiation, chemotherapy)

    • Sequential timing strategies to optimize immune activation

    • Coupling with NF-κB inhibitors or proteasome inhibitors to modulate signaling outcomes

  • Biomarker development:

    • TNFRSF9 DNA methylation as a predictive biomarker for immunotherapy response

    • Expression patterns on tumor and immune cells as response indicators

    • Soluble TNFRSF9 as a potential blood-based biomarker

These approaches highlight the complexity and potential of TNFRSF9-targeted immunotherapeutic strategies, with particular emphasis on overcoming previous clinical challenges related to toxicity and efficacy .

How do genetic variations in TNFRSF9 influence protein function and disease susceptibility?

Genetic variations in TNFRSF9 have significant implications for protein function and disease susceptibility, as evidenced by recent discoveries:

  • Pathogenic mutations:

    • Biallelic heterozygous mutations (e.g., NM_001561.5: c.208+1→AT and c.452C>A [p.T151K]) can cause CD137 deficiency

    • These hypomorphic mutations result in markedly reduced or abrogated CD137 expression on activated immune cells

    • Functional consequences include impaired T cell activation, reduced cytokine production, and diminished cytotoxic activity

    • Clinical manifestations include increased susceptibility to severe EBV-associated lymphoproliferative disease

  • Experimental approaches to study genetic variations:

    • Site-directed mutagenesis to generate TNFRSF9 variants

    • Stable cell line creation expressing wild-type vs. mutant TNFRSF9

    • CRISPR/Cas9 knock-in models recapitulating human mutations

    • Patient-derived primary cell analyses

  • Structure-function relationship analyses:

    • Mapping mutations to functional domains

    • Computational modeling of protein structural changes

    • Ligand binding assays for variant proteins

    • Signaling pathway activation assessments

  • Clinical correlation strategies:

    • Genotype-phenotype correlation studies

    • Response to immunomodulatory therapies based on variant status

    • Development of personalized therapeutic approaches for patients with TNFRSF9 mutations

Understanding these genetic variations provides critical insights for researchers investigating primary immunodeficiencies and developing targeted therapeutics for patients with TNFRSF9-related disorders .

What is the significance of TNFRSF9 DNA methylation patterns in cancer biology and immunotherapy response?

TNFRSF9 DNA methylation represents an emerging epigenetic biomarker with significant implications for cancer biology and immunotherapy response prediction:

  • Methylation analysis methodologies:

    • Bisulfite sequencing of TNFRSF9 promoter and regulatory regions

    • Methylation-specific PCR for targeted analysis

    • Genome-wide methylation arrays incorporating TNFRSF9 CpG sites

    • Integration with transcriptomic data to correlate methylation with expression

  • Cancer-specific methylation patterns:

    • Differential methylation between tumor and normal adjacent tissues

    • Correlation with TNFRSF9 expression levels in various tumor types

    • Association with tumor progression and clinical outcomes

    • Relationship to tumor immune microenvironment characteristics

  • Predictive value for immunotherapy:

    • Correlation between TNFRSF9 methylation status and response to immune checkpoint inhibitors

    • Integration into multiparameter predictive models

    • Potential as a companion diagnostic for TNFRSF9-targeted therapies

    • Longitudinal assessment during treatment to monitor response

  • Mechanistic insights:

    • Relationship between hypoxia and TNFRSF9 methylation

    • Impact of epigenetic modifying drugs on TNFRSF9 expression

    • Interaction with transcription factors and chromatin remodeling complexes

These findings provide rationale for further investigating TNFRSF9 DNA methylation as a predictive biomarker for immunotherapy response, potentially enabling more precise patient selection for various immunotherapeutic approaches .

What are the optimal storage and handling conditions for recombinant TNFRSF9 protein?

Proper storage and handling of recombinant TNFRSF9 protein is critical for maintaining biological activity and experimental reproducibility:

  • Storage recommendations:

    • Store lyophilized recombinant TNFRSF9 at -20°C

    • Avoid repeated freeze-thaw cycles of reconstituted protein

    • For reconstituted protein, prepare single-use aliquots

    • Typical formulation includes PBS (pH 7.4) with stabilizers such as 0.01% SKL and 5% trehalose

  • Reconstitution protocols:

    • Use sterile, filtered buffer to reconstitute lyophilized protein

    • Allow protein to equilibrate to room temperature before reconstitution

    • Gently mix by swirling rather than vortexing to prevent protein denaturation

    • Briefly centrifuge to collect all material at the bottom of the vial

  • Quality control measures:

    • Verify protein concentration after reconstitution

    • Perform activity testing using standardized binding assays

    • Check for aggregation using dynamic light scattering

    • Monitor endotoxin levels for experiments sensitive to bacterial contaminants

  • Shipping and transfer conditions:

    • Transport on blue ice to maintain temperature

    • Validate activity after shipping or transfer between laboratories

    • Document freeze-thaw cycles and storage durations

Adherence to these storage and handling guidelines will ensure optimal experimental outcomes when working with recombinant TNFRSF9 protein .

How can researchers effectively design experiments to investigate the TNFRSF9/p38MAPK/PAX6 signaling axis?

Investigation of the TNFRSF9/p38MAPK/PAX6 signaling axis requires a systematic experimental approach:

  • Pathway validation experiments:

    • siRNA/shRNA knockdown of TNFRSF9 followed by assessment of p-p38 and PAX6 levels

    • Overexpression of TNFRSF9 with concurrent monitoring of downstream components

    • P38 inhibitor studies (e.g., SB203580) to verify the dependence of PAX6 upregulation on p38 phosphorylation

    • Rescue experiments using constitutively active or dominant-negative constructs

  • Signaling kinetics analysis:

    • Time-course experiments following TNFRSF9 activation

    • Quantitative assessment of phosphorylation status at multiple timepoints

    • Single-cell analysis to account for heterogeneity in signaling responses

    • Correlation between signaling dynamics and functional outcomes

  • Cell models and conditions:

    • Comparison across multiple cell lines with varying baseline TNFRSF9 expression

    • Primary cells versus established cell lines

    • Consideration of microenvironmental factors (hypoxia, inflammatory cytokines)

    • 3D culture systems to better recapitulate in vivo signaling

  • Functional readouts:

    • Cell proliferation, invasion, and apoptosis assays

    • Gene expression profiling to identify additional pathway components

    • Interaction with other signaling pathways (crosstalk mapping)

    • Systems biology approaches to model pathway dynamics

This comprehensive approach enables detailed characterization of this important signaling axis and provides opportunities to identify novel therapeutic targets for diseases where TNFRSF9 function is dysregulated .

What are the emerging technologies for studying TNFRSF9 function in complex tissue microenvironments?

Cutting-edge technologies are revolutionizing the study of TNFRSF9 function within complex tissue microenvironments:

  • Spatial multi-omics approaches:

    • Spatial transcriptomics to map TNFRSF9 expression patterns relative to vascular structures and cell types

    • Multiplexed ion beam imaging (MIBI) for simultaneous detection of multiple proteins

    • Imaging mass cytometry for high-dimensional spatial phenotyping

    • Digital spatial profiling to quantify protein and RNA abundance with spatial context

  • Advanced organoid and tissue models:

    • Patient-derived tumor organoids with preserved immune components

    • Microfluidic organ-on-chip systems modeling tumor-immune interactions

    • 3D bioprinting of tissue models with controlled TNFRSF9 expression

    • Ex vivo tissue slice cultures maintaining native architecture

  • Single-cell analysis platforms:

    • Single-cell RNA sequencing with TNFRSF9 pathway gene signatures

    • CyTOF mass cytometry for high-parameter protein profiling

    • Cellular indexing of transcriptomes and epitopes (CITE-seq)

    • Spatial single-cell sequencing technologies

  • In vivo imaging technologies:

    • Intravital microscopy with fluorescently labeled TNFRSF9 antibodies

    • PET imaging with radiolabeled TNFRSF9-targeting agents

    • Photoacoustic imaging for deep tissue visualization

    • Bioluminescence resonance energy transfer (BRET) systems for pathway activation monitoring

These technologies provide unprecedented insights into TNFRSF9 biology within native tissue contexts, enabling more translational research approaches .

How might TNFRSF9-based therapies be optimized to overcome current limitations in clinical applications?

Optimization of TNFRSF9-based therapies requires innovative approaches to address current clinical limitations:

  • Reducing systemic toxicity:

    • Development of tumor-selective activation strategies

    • Incorporation of protease-sensitive linkers activated in tumor microenvironments

    • Engineering of prodrug-like antibody formats with inhibitory domains shed at tumor sites

    • Spatially restricted activation through tumor-targeting bispecific constructs

  • Enhancing efficacy:

    • Rational combination with checkpoint inhibitors or conventional therapies

    • Optimization of dosing schedules and administration routes

    • Development of TNFRSF9 agonists with optimized receptor crosslinking properties

    • Engineering of novel scaffold formats beyond traditional antibodies

  • Precision medicine approaches:

    • Implementation of TNFRSF9 methylation as a biomarker for patient selection

    • Development of companion diagnostics to identify optimal responders

    • Monitoring of tumor immune microenvironment changes during treatment

    • Adaptive trial designs with biomarker-guided treatment modifications

  • Manufacturing and stability improvements:

    • Enhanced production systems for complex biologics

    • Formulation optimization for improved stability and delivery

    • Development of extended-release or depot formulations

    • Novel delivery systems for improved tumor penetration

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.