TNFAIP8L2 Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery times may vary depending on the purchase method and location. For specific delivery times, please consult your local distributor.
Synonyms
AW610835 antibody; FLJ23467 antibody; Inflammation factor 20 antibody; Inflammation factor protein 20 antibody; TIPE2 antibody; TNF alpha-induced protein 8-like protein 2 antibody; TNFAIP8-like protein 2 antibody; TNFAIP8L2 antibody; TP8L2_HUMAN antibody; Tumor necrosis factor alpha induced protein 8 like 2 antibody; Tumor necrosis factor alpha-induced protein 8-like protein 2 antibody
Target Names
TNFAIP8L2
Uniprot No.

Target Background

Function
TNFAIP8L2 acts as a negative regulator of both innate and adaptive immunity by maintaining immune homeostasis. It functions as a negative regulator of Toll-like receptor and T-cell receptor activity. TNFAIP8L2 prevents hyperresponsiveness of the immune system and maintains immune homeostasis. It inhibits JUN/AP1 and NF-kappa-B activation. Additionally, TNFAIP8L2 promotes Fas-induced apoptosis.
Gene References Into Functions
  1. Our research indicated that TIPE2 is a novel tumor suppressor gene that inhibits Gastric cancer growth, potentially through AKT and IkappaBalpha phosphorylated activation. PMID: 30157801
  2. Low TIPE2 expression is associated with Gastric cancer. PMID: 30015980
  3. Our findings suggest that TIPE2 acts as an inhibitor of hepatocellular carcinoma cell growth and aggressiveness, promoting apoptosis. This indicates that TIPE2 may inhibit the metastasis-associated PI3K/AKT signaling cascade and potentially arrest the tumor cell cycle. PMID: 29568863
  4. The expression of TIPE2 in THP1 cells can be upregulated by Poly I:C. PMID: 28849057
  5. In conclusion, our study revealed that the Th2 milieu upregulates TIPE2 expression in macrophages, facilitating a change in macrophage phenotype and function. This, in turn, potentially exacerbates eosinophilic inflammation and disease progression in chronic rhinosinusitis with nasal polyps. PMID: 28665518
  6. TIPE2 may participate in T2DM by regulating TNF-alpha production. PMID: 28626770
  7. Our research elucidates the molecular mechanisms underlying the interplay of TNF-alpha, TIPE2, and apoptosis during allograft rejection. This suggests that both TNF-alpha and TIPE2 could be potential targets for successfully grafted corneal endothelium. PMID: 29480366
  8. TIPE2 inhibited the expression of asthma-related inflammatory factors in hyperstretched BEAS-2B cells by suppressing the Wnt/beta-catenin signaling pathway. PMID: 28188409
  9. TIPE2 might be associated with immune clearance of patients with chronic hepatitis B. PMID: 28390195
  10. TIPE2 could play significant roles in maintaining maternal-fetal tolerance. Decreased TIPE2 expression in the decidua may be associated with the development of missed abortion. PMID: 28851386
  11. TIPE2 suppressed tumor invasiveness and angiogenesis in non-small cell lung cancer by inhibiting the activation of Rac1, subsequently weakening its downstream effects, including F-actin polymerization and VEGF expression. PMID: 27556698
  12. Further MD simulations confirmed the dynamic stability of these lipids in the TH domain. This computational analysis provides insight into the binding mode of phospholipids in the TH domain of the TIPE family of proteins. PMID: 27783229
  13. The expression of TIPE2 protein could be a predictor of better prognosis for DLBCL. PMID: 27578327
  14. Insufficient expression of TIPE2 might be involved in the hyperreactivity of monocyte to Toll-like receptor ligands in primary biliary cirrhosis. PMID: 26644386
  15. Our data provides the first evidence that TIPE2 inhibits gastric cancer cell migration, invasion, and metastasis, likely through reversal of EMT. This suggests that TIPE2 may be a novel therapeutic target for human gastric cancer EMT and metastasis. PMID: 28186089
  16. Our study demonstrated that TIPE2 overexpression may suppress proliferation, migration, and invasion in prostate cancer cells by inhibiting the PI3K/Akt signaling pathway. PMID: 27712587
  17. TIPE2 suppressed breast cancer tumorigenesis, growth, and metastasis potentially through regulation of the AKT and p38 signaling pathways. PMID: 27779698
  18. Our data suggests that TIPE2 overexpression inhibited hypoxia-induced Wnt/beta-catenin pathway activation and EMT in glioma cells. PMID: 27656836
  19. This study shows that TIPE2 contributes to the pathogenesis of ankylosing spondylitis. PMID: 27816498
  20. TIPE2 expression was significantly decreased in human breast cancer tissue and cell lines. Overexpression of TIPE2 inhibited proliferation in vitro and tumor xenograft growth in vivo. TIPE2 also inhibited the migration/invasion of breast cancer cells by preventing the epithelial-to-mesenchymal transition (EMT) phenotype. PMID: 28081733
  21. Low expression of TIPE2 is associated with hepatocellular carcinogenesis. PMID: 27696294
  22. Our data suggests a previously unappreciated role of TIPE2 in the crosstalk between skin SCC and TAMs, highlighting TIPE2 as a promising novel target for skin SCC treatment. PMID: 26577853
  23. Our results provide evidence that TIPE2 acts as an inhibitor of gastric cancer cell growth and triggers an IRF4-associated signaling cascade that promotes p27 expression and restores control of cell proliferation. PMID: 26781452
  24. The TIPE2-elicited antimetastatic effect in gastric cancer was closely associated with the inhibition of AKT signaling and enhancement of GSK3b activity, followed by the degradation and decreased translocation to the nucleus of b-catenin. PMID: 26530498
  25. TIPE2 promoted lung cancer cell apoptosis by affecting apoptosis-related molecules caspase-3, caspase-9, Bcl-2, and Bax, potentially through regulating P38 and Akt pathways. PMID: 25946186
  26. Tumor necrosis factor-alpha-induced protein-8 like-2 (TIPE2) upregulates p27 to decrease gastric cancer cell proliferation. PMID: 25536447
  27. TIPE2 mRNA showed a sensitivity of 74.63%, specificity of 90.24%, positive predictive value of 92.5%, and negative predictive value of 67.3% for predicting 3-month mortality in Acute-on-Chronic Hepatitis B Liver Failure. PMID: 26426653
  28. These findings strongly suggest that TIPE2 plays a crucial role in shifting L-arginase metabolism from the production of NO to urea during the host inflammatory response. PMID: 24806446
  29. Both TNF-alpha and TIPE2 could be potential targets for the treatment of hepatocellular carcinoma metastasis. PMID: 25339267
  30. TIPE2 might serve as a tumor suppressor in non-small cell lung cancer progression. PMID: 25542151
  31. TIPE2 can inhibit caspase-8 activity in colon cancer cells. TIPE2 can regulate TLR4 inflammatory effects and inhibit further amplification of the cascade reaction via caspase-8. PMID: 24934366
  32. TIPE2 plays a significant role in regulating hepatitis B virus-specific CD8(+) T cell functions in patients with hepatitis B. PMID: 25499447
  33. Human TIPE2 is an endogenous inhibitor of Rac1 in hepatocellular carcinoma (HCC), attenuating invasion and metastasis of HCC. PMID: 24274578
  34. TIPE2 mRNA and protein expression were decreased in children with asthma compared with healthy controls. PMID: 24107080
  35. Letter: Detection of TIPE2 in blood samples may be used as a diagnostic molecular marker in clinical monitoring of kidney chronic rejection. PMID: 23009108
  36. TIPE2 is an inhibitor of both inflammation and cancer, and a potential drug target for inflammatory and neoplastic diseases. PMID: 22326055
  37. The unique expressional profile of human TIPE2 suggests new functions beyond controlling innate and adaptive immunity. PMID: 21459448
  38. These findings indicate that TIPE2 plays a significant role in taming Hepatitis B virus-induced hepatic inflammation. PMID: 21466895
  39. High levels of TIPE2 were detected in monocyte/macrophage-derived cell lines and ovarian adenocarcinoma cells, but not detectable or weakly expressed in most human carcinoma cell lines. PMID: 20663561

Show More

Hide All

Database Links

HGNC: 26277

OMIM: 612112

KEGG: hsa:79626

STRING: 9606.ENSP00000357906

UniGene: Hs.432360

Protein Families
TNFAIP8 family, TNFAIP8L2 subfamily

Q&A

What is TNFAIP8L2/TIPE2 and why is it important in immunological research?

TNFAIP8L2 (Tumor Necrosis Factor Alpha-Induced Protein 8-Like 2), also known as TIPE2, is a 184 amino acid protein that functions as a critical negative regulator of both innate and adaptive immunity . It plays an essential role in maintaining immune homeostasis by regulating Toll-like receptor and T-cell receptor function . TIPE2 is preferentially expressed in lymphoid tissues including spleen, thymus, small intestine, and lymph nodes, with lower expression in colon, lung, and skin .

Research significance:

  • Acts as a negative regulator that prevents excessive inflammatory responses

  • Implicated in autoimmune disorders when downregulated

  • Has emerging roles in tumor progression through RAS signaling pathway interaction

  • Involved in cerebral ischemia inflammatory response

  • Associated with diseases including skin squamous cell carcinoma

The protein's role in immune regulation makes TIPE2-targeting antibodies valuable tools for studying inflammation, autophagy, and immune response mechanisms.

What applications are FITC-conjugated TNFAIP8L2 antibodies validated for?

FITC-conjugated TNFAIP8L2 antibodies have been validated for multiple applications across various experimental systems:

ApplicationValidated DilutionsSample Types
Immunofluorescence (IF/ICC)1:50-1:200HepG2 cells, RAW 264.7 cells, C6, HeLa, L929
Flow Cytometry (FC)0.40 μg per 10^6 cells in a 100 μl suspensionTHP-1 cells and other suspension cells
Western Blotting (WB)1:300-5000THP-1 cells, mouse tissues (spleen, small intestine, cerebellum)
Immunohistochemistry (IHC-P/F)1:50-500Human lymphoma tissue, ovary tumor tissue, mouse spleen tissue

For optimal results in immunofluorescence applications, researchers should begin with the recommended dilution range and optimize based on their specific experimental conditions, cell lines, and detection systems .

How should FITC-conjugated TNFAIP8L2 antibodies be stored to maintain reactivity?

Proper storage is crucial for maintaining antibody performance across multiple experiments:

Recommended storage conditions:

  • Store at -20°C in aliquots to avoid repeated freeze-thaw cycles

  • Some antibody preparations contain 50% glycerol to prevent freezing damage

  • Protect from light exposure to prevent photobleaching of the FITC fluorophore

  • Valid for approximately 12 months when stored properly

Practical methodology:

  • Upon receipt, immediately aliquot the antibody into smaller volumes based on typical experimental usage (20-50 μl per aliquot)

  • Store aliquots in opaque containers or wrapped in aluminum foil

  • Thaw only the required amount for each experiment

  • When working with the antibody, keep it on ice and protected from direct light

  • Return to -20°C promptly after use

This storage protocol helps maintain the structural integrity of both the antibody and the FITC conjugate, ensuring consistent signal intensity across experiments .

How should researchers design proper controls when using FITC-conjugated TNFAIP8L2 antibodies?

Robust experimental design requires appropriate controls to validate specificity and reduce false positives:

Essential controls for immunofluorescence/flow cytometry:

Control TypePurposeImplementation
Isotype ControlControls for non-specific bindingUse FITC-conjugated rabbit IgG (matched to antibody host/isotype) at the same concentration
Secondary-only ControlControls for non-specific binding of secondary antibody (if using indirect detection)Omit primary antibody but include all other staining steps
Unstained ControlEstablishes baseline autofluorescenceProcess cells through all steps except antibody incubation
Positive ControlValidates staining protocolUse cells/tissues known to express TNFAIP8L2 (e.g., spleen tissue, THP-1 cells)
Negative ControlConfirms specificityUse TNFAIP8L2 knockout cells or tissues or cells known not to express the target
Peptide CompetitionVerifies epitope specificityPre-incubate antibody with immunizing peptide before staining

For western blotting validation, additional controls using siRNA knockdown or CRISPR knockout of TNFAIP8L2 can further confirm antibody specificity .

What is the optimal sample preparation protocol for detecting TNFAIP8L2 in different cellular compartments?

TNFAIP8L2/TIPE2 localizes to multiple cellular compartments including cytoplasm and lysosomal membranes , requiring specific fixation and permeabilization protocols:

For immunofluorescence microscopy (cultured cells):

  • Grow cells on sterile coverslips to 70-80% confluency

  • Wash cells twice with PBS (pH 7.4)

  • Fix with 4% paraformaldehyde for 15 minutes at room temperature

  • Wash 3x with PBS

  • Permeabilize with 0.1% Triton X-100 in PBS for 10 minutes

  • Block with 5% normal serum (from same species as secondary antibody) for 1 hour

  • Incubate with FITC-conjugated TNFAIP8L2 antibody at 1:50-1:200 dilution overnight at 4°C

  • Wash 3x with PBS

  • Counterstain nuclei with DAPI

  • Mount with anti-fade mounting medium

For tissue sections:

  • For paraffin sections, antigen retrieval with TE buffer (pH 9.0) is recommended

  • Alternatively, citrate buffer (pH 6.0) can be used

For lysosomal localization studies:

  • Co-stain with lysosomal markers such as LAMP1 to visualize colocalization of TNFAIP8L2 with lysosomes

  • Based on research findings, TNFAIP8L2 can localize to LAMP1-positive structures and affect lysosomal RAC1 levels

What are the optimal imaging parameters for FITC-conjugated TNFAIP8L2 in fluorescence microscopy?

Optimizing imaging parameters ensures high-quality data acquisition while minimizing photobleaching:

FITC fluorophore characteristics:

  • Excitation maximum: ~495 nm

  • Emission maximum: ~520 nm

  • Filter set: Standard FITC/GFP filter set (typically 480/30 ex, 535/40 em)

Imaging recommendations:

  • Exposure time: Start with short exposures (200-500ms) and adjust based on signal intensity

  • Laser/lamp power: Use minimum power necessary to visualize signal (typically 10-30% of maximum)

  • Gain settings: Begin with moderate gain settings and increase if necessary (trade-off with noise)

  • Pixel dwell time (confocal): 1-2 μs for initial scans, increase for final images if needed

  • Z-stack parameters: 0.3-0.5 μm step size for high-resolution 3D reconstruction

  • Dynamic range: Capture using 12-16 bit depth for maximum information retention

  • Sequential scanning: When performing multi-color imaging, use sequential scanning to prevent bleed-through

Advanced optimization:

  • Employ deconvolution algorithms for improved signal-to-noise ratio

  • For co-localization studies with lysosomal markers, consider super-resolution techniques such as STED or SIM microscopy to resolve structures below the diffraction limit

How can TNFAIP8L2 antibodies be used to investigate MTOR-dependent autophagy regulation?

Research has revealed that TNFAIP8L2 plays a critical role in autophagy regulation through the RAC1-MTOR axis, offering several experimental approaches:

Experimental design for studying TNFAIP8L2-autophagy regulation:

  • Autophagic flux assessment:

    • Treat cells with TNFAIP8L2 overexpression/knockdown vectors

    • Monitor LC3B-II and SQSTM1 levels by western blotting

    • Use bafilomycin A1 (autophagosome-lysosome fusion inhibitor) to determine which step of autophagy is affected

  • MTOR activity measurement:

    • Evaluate phosphorylation status of MTOR and its downstream targets (p-RPS6) by western blotting

    • Use rapamycin as a control for MTOR inhibition

  • Visualization of autolysosome reformation:

    • Track LAMP1+LC3B+ puncta under starvation conditions

    • Monitor lysosome regeneration using LAMP+ vesicle quantification

    • Compare wild-type and TNFAIP8L2-overexpressing cells during prolonged starvation (8-10h)

  • RAC1-MTOR-TNFAIP8L2 interaction studies:

    • Perform co-immunoprecipitation to assess RAC1-MTOR binding in presence/absence of TNFAIP8L2

    • Use immunofluorescence to visualize colocalization of RAC1 and MTOR on lysosomal membranes

Research has shown that TNFAIP8L2 competes with MTOR for binding to GTP-bound RAC1, thereby inhibiting MTORC1 activity during prolonged starvation conditions .

What methodological approaches can resolve contradictory data regarding TNFAIP8L2 localization patterns?

Researchers may encounter apparently contradictory data regarding TNFAIP8L2 subcellular localization due to various factors including cell type differences, experimental conditions, and technical limitations:

Resolution approaches:

  • Multi-technique validation:

    • Combine immunofluorescence with subcellular fractionation and western blotting

    • Use both N and C-terminal targeting antibodies to account for potential cleavage products

    • Employ epitope-tagged constructs (GFP/FLAG-TNFAIP8L2) for verification

  • Advanced imaging techniques:

    • Super-resolution microscopy (STED, STORM, SIM) to precisely define localization

    • Live-cell imaging with photoactivatable fluorescent protein fusions to track dynamic localization

    • Correlative light and electron microscopy for ultrastructural localization

  • Context-dependent localization studies:

    • Examine localization under different cellular states:

      • Normal growth conditions

      • Starvation (amino acid/serum deprivation)

      • Inflammatory stimulation (LPS treatment)

      • Cell cycle phases

  • Functional domain mapping:

    • Create deletion/mutation constructs to identify localization signal sequences

    • Particularly focus on the RAC1-binding domain (K15,16 region) which affects protein-protein interactions

Research indicates that TNFAIP8L2 can translocate between cytoplasmic and lysosomal compartments, necessitating careful experimental design to capture its dynamic localization patterns .

How can TNFAIP8L2 antibodies be applied in studies of immunometabolism and inflammation?

TNFAIP8L2/TIPE2 has emerged as a key regulator at the intersection of inflammation and metabolism, offering several research applications:

Experimental approaches:

  • In vitro macrophage inflammation models:

    • Isolate bone marrow-derived macrophages (BMDMs) from wild-type and Tnfaip8l2-knockout mice

    • Stimulate with LPS to trigger inflammatory response

    • Measure pro-inflammatory cytokine production (IL6, TNF) by ELISA

    • Assess TNFAIP8L2 expression changes using the antibody

  • Metabolic reprogramming analysis:

    • Measure mitochondrial respiration rate using Seahorse metabolic analyzer

    • Compare oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) in wild-type versus Tnfaip8l2-deficient macrophages

    • Evaluate lipid biosynthesis pathway activation

  • In vivo inflammation models:

    • LPS-induced endotoxemia model in wild-type and Tnfaip8l2-knockout mice

    • Monitor body temperature and clinical scores

    • Assess tissue inflammation via histology (H&E staining)

    • Measure circulating pro-inflammatory cytokines

    • Evaluate MTOR activity in tissues using p-RPS6 immunohistochemistry

  • Gene expression profiling integration:

    • Perform RNA-seq on macrophages with varied TNFAIP8L2 expression

    • Analyze enrichment for inflammatory and metabolic pathways

    • Focus on leukocyte activation and lipid biosynthesis gene sets

Research has demonstrated that Tnfaip8l2 deficiency exacerbates inflammatory responses by upregulating MTOR activity, and TNFAIP8L2 serves as a "brake" for immunometabolism that must be released for effective inflammatory responses .

How can researchers troubleshoot weak or non-specific FITC-TNFAIP8L2 antibody signals in immunofluorescence?

Several factors can contribute to suboptimal staining results with FITC-conjugated TNFAIP8L2 antibodies:

Common issues and solutions:

IssuePotential CausesTroubleshooting Approaches
Weak signalInsufficient antibody concentrationIncrease antibody concentration within recommended range (1:50-1:200)
Inadequate antigen retrievalFor FFPE tissues, optimize antigen retrieval using TE buffer (pH 9.0) or citrate buffer (pH 6.0)
Low target expressionVerify expression in your sample type; consider positive control tissues (spleen, thymus)
PhotobleachingProtect samples from light; use anti-fade mounting medium; image promptly
High backgroundNon-specific bindingIncrease blocking time/concentration; use 5% BSA or normal serum
Insufficient washingExtend wash steps (3x 5min); use gentle agitation
AutofluorescenceUse Sudan Black B (0.1-0.3%) treatment to reduce autofluorescence
Fixation artifactsOptimize fixation time; consider alternative fixatives (methanol vs. PFA)
No signalAntibody degradationCheck storage conditions; use fresh aliquot
Target denaturationEnsure epitope is preserved during fixation/processing
Wrong filter setVerify microscope filter settings for FITC (Ex:~495nm, Em:~520nm)

Advanced troubleshooting:

  • Perform titration experiments to determine optimal antibody concentration for specific cell types

  • Consider alternative detection methods (e.g., indirect immunofluorescence with amplification)

  • For co-localization studies, address potential fluorophore interactions and spectral overlap

What quality control metrics should be established for FITC-conjugated TNFAIP8L2 antibody validation?

Implementing comprehensive quality control is essential for reliable TNFAIP8L2 antibody applications:

Validation metrics:

  • Specificity assessment:

    • Western blot showing single band at expected molecular weight (18-20 kDa)

    • Absence of signal in TNFAIP8L2 knockout/knockdown samples

    • Signal reduction in peptide competition assays

    • Consistent staining pattern across multiple antibody clones targeting different epitopes

  • Sensitivity measurements:

    • Limit of detection determination using serial dilutions of recombinant protein

    • Signal-to-noise ratio quantification in immunofluorescence images

    • Dynamic range assessment across expression levels

  • Reproducibility standards:

    • Intra-assay variability (<10% CV preferred)

    • Inter-assay variability (<15% CV preferred)

    • Lot-to-lot consistency validation

  • Application-specific validation:

    • For flow cytometry: Staining index calculation, resolution of positive/negative populations

    • For microscopy: Colocalization coefficients with known markers (e.g., LAMP1 for lysosomal localization)

    • For quantitative applications: Standard curve linearity, dynamic range assessment

Documentation best practices:

  • Maintain detailed records of validation experiments including all controls

  • Include positive control samples in each experimental run

  • Document lot numbers and validation data for antibody batches

How can live-cell imaging approaches be optimized for studying TNFAIP8L2 dynamics during autophagy?

Live-cell imaging offers unique insights into the dynamic role of TNFAIP8L2 in autophagy regulation, particularly during autolysosome reformation:

Methodological approach:

  • Expression construct design:

    • Create fluorescent protein fusion constructs (e.g., TNFAIP8L2-mEGFP or mCherry-TNFAIP8L2)

    • Validate fusion protein functionality through rescue experiments in TNFAIP8L2-knockout cells

    • Consider using CRISPR knock-in approaches for endogenous tagging to maintain physiological expression levels

  • Multi-color imaging system:

    • Combine with lysosomal markers (LAMP1-mRFP) and autophagosomal markers (GFP-LC3 or mCherry-GFP-LC3 tandem reporter)

    • Use spectrally compatible fluorophores to minimize bleed-through

    • Design time-lapse experiments capturing:

      • Starvation-induced autophagy induction

      • Autolysosome formation

      • Autolysosome reformation during prolonged starvation

  • Acquisition parameters optimization:

    • Minimize phototoxicity: Reduce light intensity, increase camera sensitivity

    • Balance temporal resolution with photobleaching (typically 1-5 minute intervals)

    • Employ spinning disk confocal for improved signal-to-noise ratio with less phototoxicity

    • Use environmental chamber to maintain 37°C, 5% CO₂, and humidity

  • Advanced analytical approaches:

    • Particle tracking algorithms to follow vesicle dynamics

    • Intensity-based colocalization analysis over time

    • FRAP (Fluorescence Recovery After Photobleaching) to assess protein mobility

    • FRET-based assays to examine TNFAIP8L2-RAC1 interactions in real-time

Research has shown that TNFAIP8L2 impairs autolysosome reformation during prolonged starvation, suggesting its dynamic involvement in different phases of the autophagy process .

How can TNFAIP8L2 antibodies be integrated into multi-parameter flow cytometry panels for immune phenotyping?

TNFAIP8L2's role as an immune regulator makes it valuable in comprehensive immune phenotyping panels:

Panel design considerations:

  • Cell surface marker combinations:

    • Combine FITC-TNFAIP8L2 with markers for myeloid populations (CD11b, CD14, CD16)

    • Include T-cell markers (CD3, CD4, CD8) and activation markers (CD69, CD25)

    • Add lineage markers for dendritic cells, NK cells, and B cells as needed

  • Intracellular staining protocol optimization:

    • Fix cells with 4% paraformaldehyde for 15 minutes

    • Permeabilize with 0.1% saponin buffer or commercial permeabilization reagents

    • Block with 2% normal serum

    • Stain with FITC-TNFAIP8L2 antibody (0.40 μg per 10^6 cells)

    • Include cytokine staining (IL-6, TNF, IL-10) for functional assessment

  • Spectral considerations:

    • FITC spectrum (excitation ~495nm, emission ~520nm) may overlap with PE

    • Consider alternative conjugates (AF488) if panel design requires PE

    • Use compensation controls for each fluorochrome

    • When possible, place TNFAIP8L2 on a separate laser line from potentially overlapping fluorophores

  • Analysis strategies:

    • Gate on specific immune populations first, then assess TNFAIP8L2 expression

    • Use biaxial plots, histogram overlays, and dimensionality reduction (tSNE, UMAP)

    • Correlate TNFAIP8L2 expression with activation markers and cytokine production

    • Compare expression across disease states or treatment conditions

Example applications:

  • Monitor TNFAIP8L2 expression changes in inflammatory diseases

  • Assess correlation between TNFAIP8L2 levels and MTOR pathway activation markers

  • Evaluate TNFAIP8L2 expression in tumor-infiltrating immune cells

What approaches can integrate TNFAIP8L2 antibody-based studies with omics data for systems biology analysis?

Integrating antibody-based protein detection with multi-omics datasets enables systems-level understanding of TNFAIP8L2 function:

Integration methodologies:

  • Proteogenomic correlation:

    • Measure TNFAIP8L2 protein levels via IF/WB in multiple cell states

    • Perform parallel RNA-seq to correlate transcript and protein expression

    • Identify post-transcriptional regulation mechanisms

    • Use genome-wide gene expression profiling to identify enriched pathways

  • Protein interactome mapping:

    • Use TNFAIP8L2 antibodies for co-immunoprecipitation followed by mass spectrometry

    • Validate key interactions (e.g., RAC1, MTOR) via co-IP and IF colocalization

    • Build interaction networks incorporating known binding partners

    • Overlay with phosphoproteomic data to identify signaling nodes

  • Functional genomics integration:

    • Combine CRISPR-Cas9 genetic screens with immunophenotyping

    • Identify genetic modifiers of TNFAIP8L2 function in autophagy and immune regulation

    • Use antibodies to validate screen hits via protein expression/localization changes

  • Computational approaches:

    • Network analysis of TNFAIP8L2-centered protein-protein interactions

    • Pathway enrichment analysis of differentially expressed genes in TNFAIP8L2-deficient cells

    • Machine learning classification of cellular phenotypes based on TNFAIP8L2 expression patterns

Research applications:

  • Map TNFAIP8L2's role in leukocyte activation and lipid biosynthesis pathways

  • Identify novel regulatory mechanisms connecting metabolism and inflammation

  • Develop predictive models for inflammatory disease progression based on TNFAIP8L2 network status

How can TNFAIP8L2 antibodies contribute to preclinical evaluation of immunotherapeutic approaches?

TNFAIP8L2's role as a negative regulator of immunity positions it as a potential therapeutic target and biomarker:

Preclinical research applications:

  • Biomarker development:

    • Quantify TNFAIP8L2 expression in patient-derived samples via flow cytometry

    • Correlate expression levels with disease severity or treatment response

    • Develop standardized immunoassays for clinical sample analysis

    • Evaluate TNFAIP8L2 as a predictive biomarker for immunotherapy response

  • Therapeutic target validation:

    • Use antibodies to monitor target engagement in drug development

    • Assess TNFAIP8L2 expression changes following treatment with immune modulators

    • Evaluate effects of TNFAIP8L2 modulation on immune activation and inflammation

    • Investigate TNFAIP8L2 as a drug target for inflammatory diseases

  • In vivo efficacy studies:

    • Monitor TNFAIP8L2 expression in immune cells from treated animals

    • Correlate with inflammatory markers, disease scores, and survival outcomes

    • Evaluate tissue-specific changes in protein expression

    • Assess MTOR pathway activation status in response to TNFAIP8L2 modulation

  • Mechanism of action studies:

    • Use multiplexed IF to evaluate TNFAIP8L2 alongside key signaling nodes

    • Investigate autophagy modulation as a mechanism in anti-tumor immunity

    • Assess metabolic reprogramming in immune cells following TNFAIP8L2 manipulation

Research has demonstrated that TNFAIP8L2 deficiency exacerbates inflammatory responses and lung injury in endotoxemia models by upregulating MTOR activity, suggesting its potential as a therapeutic target for inflammatory conditions .

What are the critical differences between various FITC-conjugated TNFAIP8L2 antibody preparations?

Researchers should be aware of important variations between commercially available FITC-conjugated TNFAIP8L2 antibodies:

Key parameters to compare:

ParameterVariationsResearch Implications
ImmunogenFull recombinant protein (1-184AA) vs. peptide fragments (10-110/184) Different epitope recognition affecting detection of specific domains
Host SpeciesPredominantly rabbit polyclonal Consistency between experiments when using different lots
Purification MethodProtein A/G purification vs. affinity purification Purity affecting background and specificity
Conjugation MethodDirect FITC labeling techniquesFluorophore-to-protein ratio affecting signal intensity
FormulationBuffer composition, preservatives, stabilizersLong-term stability and compatibility with live-cell applications
Validation ExtentSpecies/applications testedConfidence in cross-reactivity claims

Critical considerations:

  • Antibodies targeting different epitopes may yield varying results in detecting TNFAIP8L2 complexes with binding partners like RAC1

  • The K15,16 region is critical for RAC1 binding; antibodies targeting this region may interfere with protein-protein interactions

  • Cross-reactivity with other TNFAIP8 family members should be assessed, particularly in systems expressing multiple family members

How can multi-color immunofluorescence be optimized for studying TNFAIP8L2 interactions with the autophagy machinery?

TNFAIP8L2's complex role in autophagy regulation involves interactions with multiple proteins that can be visualized through optimized multi-color immunofluorescence:

Protocol optimization:

  • Antibody panel design:

    • FITC-TNFAIP8L2 antibody (primary or directly conjugated)

    • Markers for autophagosomes: LC3B (different fluorophore)

    • Lysosomal markers: LAMP1

    • MTOR pathway components: mTOR, phospho-S6

    • RAC1 (binding partner of TNFAIP8L2)

  • Sequential staining approach:

    • Begin with the lowest concentration antibody

    • Use sequential rather than cocktail staining to minimize cross-reactivity

    • Include blocking steps between antibody applications

    • Consider tyramide signal amplification for low abundance targets

  • Advanced fixation techniques:

    • For membrane protein preservation: Use mild fixation (1-2% PFA) or glyoxal-based fixatives

    • For detecting transient interactions: Consider proximity ligation assay (PLA)

    • For ultrastructural localization: Correlative light and electron microscopy

  • Imaging considerations:

    • Use confocal microscopy with appropriate channel settings

    • Consider spectral unmixing for closely overlapping fluorophores

    • Apply deconvolution algorithms to improve resolution

    • For co-localization analysis, acquire images at Nyquist sampling rate

Research applications:

  • Visualize RAC1-MTOR-TNFAIP8L2 interactions on lysosomal membranes

  • Track autolysosome reformation during prolonged starvation

  • Assess TNFAIP8L2 competition with MTOR for RAC1 binding

What methodological advances can improve detection sensitivity for low abundance TNFAIP8L2 in tissue samples?

Detecting low-abundance TNFAIP8L2 in tissues with variable expression levels requires specialized approaches:

Advanced detection strategies:

  • Signal amplification methods:

    • Tyramide signal amplification (TSA): Enhances FITC signal up to 100-fold

    • Implementation protocol:

      • Use HRP-conjugated secondary antibody

      • Apply FITC-tyramide substrate

      • HRP converts tyramide to reactive intermediate that covalently binds nearby proteins

      • Results in localized signal amplification

  • Optimized antigen retrieval:

    • Pressure cooker-based retrieval in TE buffer (pH 9.0)

    • Extended retrieval times (20-40 minutes)

    • Use of commercial epitope retrieval solutions with proprietary buffers

    • Alternative retrieval methods for difficult tissues:

      • Protease-induced epitope retrieval

      • Heat-induced epitope retrieval in different buffers

  • Detection system improvements:

    • Quantum dot-conjugated systems for enhanced photostability

    • Use of high-sensitivity sCMOS or EMCCDs for imaging

    • Computational approaches:

      • Deconvolution algorithms

      • Machine learning-based signal enhancement

      • Background subtraction techniques

  • Pre-analytical considerations:

    • Minimize time from tissue collection to fixation

    • Optimize fixation duration (12-24h recommended)

    • Use consistent sectioning thickness (4-6μm optimal)

    • Apply tissue-specific blocking reagents to reduce background

Research applications include detecting TNFAIP8L2 in tissues with naturally low expression (colon, lung, skin) and tracking expression changes in disease states such as inflammatory conditions or cancer .

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.