TNFAIP8L2 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to TNFAIP8L2 Antibody

TNFAIP8L2 antibodies are immunological reagents designed to detect and quantify the TNFAIP8L2 protein in research and diagnostic settings. These antibodies enable investigations into TNFAIP8L2’s functions, including its role as a negative regulator of Toll-like receptor (TLR) and T-cell receptor (TCR) signaling . TNFAIP8L2 is preferentially expressed in myeloid cells and lymphoid tissues, making these antibodies vital for studying immune cell behavior and inflammatory diseases .

Immune Regulation and Inflammation

  • TNFAIP8L2 deficiency in macrophages increases lipid biosynthesis and mitochondrial respiration, enhancing inflammatory responses .

  • In dendritic cells (DCs), TNFAIP8L2 suppresses autophagy by inhibiting the TAK1/JNK pathway, which modulates immune dysfunction during sepsis .

Cancer Research

  • High TNFAIP8L2 expression in glioma correlates with poor prognosis and altered immune cell infiltration (e.g., increased macrophages, neutrophils) .

  • The antibody has been used to identify TNFAIP8L2’s tumor-suppressive role in hepatocellular carcinoma and non-small cell lung cancer .

Infectious Diseases

  • TNFAIP8L2 knockdown in macrophages reduces Staphylococcus aureus susceptibility by altering IL-1β and GM-CSF levels .

Clinical Significance and Therapeutic Potential

TNFAIP8L2 antibodies are pivotal for exploring therapeutic strategies:

  • Autoimmune Diseases: TNFAIP8L2 downregulation is linked to systemic inflammation, making it a target for restoring immune balance .

  • Cancer Immunotherapy: Antibody-based detection aids in profiling TNFAIP8L2’s dual role as a tumor suppressor and immune checkpoint .

  • Metabolic Disorders: TNFAIP8L2 regulates lipid metabolism in macrophages, implicating it in atherosclerosis and obesity .

Table 1: TNFAIP8L2 Antibody Applications in Disease Models

Disease ModelFindingsMethod UsedSource
GliomaHigh TNFAIP8L2 linked to immune cell dysregulationIHC, Microarray
SepsisTNFAIP8L2 modulates DC autophagyWB, Flow Cytometry
AtherosclerosisTNFAIP8L2 KO increases macrophage lipid synthesisSeahorse Analysis

Table 2: Immune Cell Correlations with TNFAIP8L2 in Glioma

Immune Cell TypeCorrelation with TNFAIP8L2
MacrophagesPositive
NeutrophilsPositive
Regulatory T cells (TReg)Negative
Dendritic cells (DCs)Positive

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Our standard lead time for product dispatch is 1-3 working days following receipt of your order. Delivery times may vary depending on the purchase method or location. Please consult your local distributor for specific delivery timeframe.
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
TNFαIP8L2, also known as TIPE2, functions as a negative regulator of both innate and adaptive immunity, contributing to the maintenance of immune homeostasis. It exerts its regulatory effects by modulating Toll-like receptor and T-cell receptor signaling pathways. By suppressing hyperresponsiveness of the immune system, TIPE2 maintains immune equilibrium. Furthermore, it inhibits the activation of JUN/AP1 and NF-κB signaling pathways, and promotes Fas-induced apoptosis.
Gene References Into Functions
  1. Our research demonstrated that TIPE2 is a novel tumor suppressor gene that inhibits gastric cancer growth, potentially mediated through AKT and IκBα phosphorylation activation. PMID: 30157801
  2. Low TIPE2 expression is associated with Gastric cancer. PMID: 30015980
  3. The findings of our study indicate that TIPE2 acts as an inhibitor of hepatocellular carcinoma cell growth and aggressiveness, promoting apoptosis. This suggests that TIPE2 may inhibit the metastasis-associated PI3K/AKT signaling cascade and potentially arrest the tumor cell cycle. PMID: 29568863
  4. Poly I:C can upregulate the expression of TIPE2 in THP1 cells. PMID: 28849057
  5. In conclusion, our investigation revealed for the first time that the Th2 milieu can upregulate TIPE2 expression in macrophages. This upregulation facilitates changes in macrophage phenotype and function, potentially exacerbating eosinophilic inflammation and disease progression in chronic rhinosinusitis with nasal polyps. PMID: 28665518
  6. TIPE2 may play a role in Type 2 Diabetes Mellitus (T2DM) by regulating TNF-α production. PMID: 28626770
  7. Our study elucidates the molecular mechanisms underlying the interplay between TNF-α, TIPE2, and apoptosis during allograft rejection. These findings suggest that both TNF-α and TIPE2 might be potential therapeutic targets for successful corneal endothelium grafting. PMID: 29480366
  8. TIPE2 inhibited the expression of asthma-related inflammatory factors in hyperstretched BEAS-2B cells by suppressing the Wnt/β-catenin signaling pathway. PMID: 28188409
  9. TIPE2 might be associated with immune clearance in patients with chronic hepatitis B. PMID: 28390195
  10. TIPE2 may play crucial roles in maintaining maternal-fetal tolerance, and decreased TIPE2 expression in the decidua may be linked to 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 molecular dynamics simulations confirmed the dynamic stability of these lipids in the TH domain. This computational analysis provides insights 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 potentially be a predictor of better prognosis for Diffuse Large B-Cell Lymphoma (DLBCL). PMID: 27578327
  14. Insufficient expression of TIPE2 might contribute to the hyperreactivity of monocytes to Toll-like receptor ligands in primary biliary cirrhosis. PMID: 26644386
  15. Data provided the first evidence that TIPE2 inhibits gastric cancer cell migration, invasion, and metastasis, very likely via reversal of epithelial-mesenchymal transition (EMT). This reveals that TIPE2 may be a novel therapeutic target for human gastric cancer EMT and metastasis. PMID: 28186089
  16. Researchers 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. These data suggest that TIPE2 overexpression inhibited hypoxia-induced Wnt/β-catenin pathway activation and EMT in glioma cells. PMID: 27656836
  19. This study demonstrates 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 suggest a previously unappreciated role of TIPE2 in the crosstalk between skin squamous cell carcinoma (SCC) and tumor-associated macrophages (TAMs), highlighting TIPE2 as a promising novel target for skin SCC treatment. PMID: 26577853
  23. 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 GSK3β activity followed by the degradation and decreased translocation to the nucleus of β-catenin. PMID: 26530498
  25. TIPE2 promoted lung cancer cell apoptosis by affecting apoptosis-related molecules caspase-3, caspase-9, Bcl-2, and Bax, possibly via 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 results strongly suggest that TIPE2 plays an important role in shifting L-arginase metabolism from production of nitric oxide (NO) to urea, during host inflammatory response. PMID: 24806446
  29. Both TNF-α and TIPE2 might 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 effect and inhibit further amplification of 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), by which it attenuates 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 results indicate that TIPE2 plays a significant role in mitigating 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 and what are its primary functions in the immune system?

TNFAIP8L2 (also known as TIPE2) acts as a negative regulator of innate and adaptive immunity by maintaining immune homeostasis. This protein serves several critical functions:

  • Negatively regulates Toll-like receptor and T-cell receptor function

  • Prevents hyperresponsiveness of the immune system

  • Inhibits JUN/AP1 and NF-kappa-B activation pathways

  • Promotes Fas-induced apoptosis

TNFAIP8L2 is primarily expressed in immune tissues and cells, with a calculated molecular weight of approximately 21 kDa, though it is typically observed at 18-20 kDa in Western blot applications .

What applications are recommended for TNFAIP8L2 antibodies in research?

TNFAIP8L2 antibodies have been validated for multiple applications with specific dilution recommendations:

ApplicationRecommended DilutionPositive Samples
Western Blot (WB)1:500-1:6000THP-1 cells, mouse spleen tissue, RAW 264.7 cells
Immunohistochemistry (IHC)1:50-1:500Human lymphoma tissue, human ovary tumor tissue, mouse spleen tissue
Immunofluorescence (IF/ICC)1:50-1:500RAW 264.7 cells, HepG2 cells
Immunoprecipitation (IP)0.5-4.0 μg for 1.0-3.0 mg proteinMouse spleen tissue
Flow Cytometry (FC)0.40 μg per 10^6 cellsTHP-1 cells
ELISA1:40000Various cell lysates

All applications should be optimized for specific experimental conditions as performance may vary between antibody lots and experimental systems .

How should TNFAIP8L2 antibodies be stored and handled for optimal performance?

For maximum antibody stability and performance, follow these guidelines:

  • Store concentrated antibody at -20°C for long-term storage (typically stable for 12 months from receipt)

  • For frequent use, aliquot and store at 4°C for up to one month

  • Avoid repeated freeze-thaw cycles as this significantly reduces antibody activity

  • Most formulations contain 50% glycerol, 0.5% BSA, and 0.02% sodium azide for stability

  • When working with the antibody, keep it on ice and minimize exposure to room temperature

Proper storage is critical for maintaining antibody performance. According to manufacturer recommendations, some TNFAIP8L2 antibodies can maintain stability at 4°C for up to 6 months after reconstitution, but this varies by product .

What is the optimal sample preparation protocol for TNFAIP8L2 Western blotting?

For successful detection of TNFAIP8L2 in Western blot applications:

  • Sample Preparation:

    • Prepare cell or tissue lysates using standard lysis buffer containing protease inhibitors

    • Load 25-30 μg of total protein per lane

  • Electrophoresis and Transfer:

    • Use 12-15% SDS-PAGE gels for optimal resolution of the 18-20 kDa TNFAIP8L2 protein

    • Transfer to PVDF or nitrocellulose membranes (PVDF often provides better results)

  • Blocking and Antibody Incubation:

    • Block with 3% non-fat dry milk in TBST for 1-2 hours at room temperature

    • Incubate with primary TNFAIP8L2 antibody at 1:1000-1:2000 dilution overnight at 4°C

    • Wash 3-5 times with TBST

    • Incubate with HRP-conjugated secondary antibody at 1:10000 dilution for 1 hour

  • Detection:

    • Develop using ECL detection system with exposure times ranging from 10-30 seconds

    • TNFAIP8L2 typically appears as a band at 18-20 kDa

Positive controls should include THP-1 cells, mouse thymus tissue, mouse spleen tissue, or rat spleen tissue, where TNFAIP8L2 expression has been well-documented .

What controls are essential when validating TNFAIP8L2 antibody specificity?

Proper validation of TNFAIP8L2 antibody specificity requires several controls:

Positive Controls:

  • Cell lines: THP-1 cells, RAW 264.7 cells

  • Tissues: Mouse spleen, mouse thymus, rat spleen, human lymphoma tissue

Negative Controls:

  • TNFAIP8L2 knockout cell lines or tissues

  • TNFAIP8L2 siRNA-treated samples

  • Secondary antibody-only controls to assess non-specific binding

Blocking Peptide Validation:

  • Pre-incubating the antibody with a specific blocking peptide containing the epitope recognized by the antibody

  • Compare staining patterns with and without blocking peptide

  • Significant reduction in signal indicates antibody specificity

Recent knockout (KO) validation approaches have emerged as the gold standard for antibody validation, as demonstrated in several recent TNFAIP8L2 studies .

How does TNFAIP8L2 regulate autophagy through the RAC1-MTORC1 signaling axis?

TNFAIP8L2 plays a sophisticated role in regulating autophagy through its interaction with the RAC1-MTORC1 signaling pathway:

Mechanism of Action:

  • TNFAIP8L2 directly binds to and blocks RAC1 GTPase activity

  • TNFAIP8L2 competes with MTOR for binding to the GTP-bound state of RAC1

  • This competition negatively regulates MTORC1 activity

  • Despite suppressing MTOR activity under glutamine and serum starvation, TNFAIP8L2 overexpression fails to induce autophagy flux

  • TNFAIP8L2 appears to impair autophagic lysosome reformation (ALR) during prolonged starvation

  • TNFAIP8L2 overexpression leads to defects in MTOR reactivation, disrupting autophagy flux and potentially leading to cell death

Experimental approaches for studying this interaction include:

  • Co-Immunoprecipitation to detect TNFAIP8L2-RAC1 and RAC1-MTOR interactions

  • Western blot analysis of LC3-II conversion and p62/SQSTM1 degradation

  • Lysosomal morphology assessment using LAMP1 staining

TNFAIP8L2 deficiency has been shown to exacerbate inflammatory responses and lung injury by controlling MTOR activity in LPS-induced mouse models, highlighting the physiological relevance of this regulatory mechanism .

What methodological considerations are important when studying TNFAIP8L2's role in dendritic cell immune function?

When investigating TNFAIP8L2's role in dendritic cell (DC) immune function, consider these methodological approaches:

Experimental Design:

  • Cell Models:

    • Primary bone marrow-derived dendritic cells (BMDCs)

    • DC cell lines (validate with primary cells)

  • Genetic Manipulation:

    • TNFAIP8L2 knockout (KO) models: Show enhanced autophagy and improved immune response

    • TNFAIP8L2 overexpression (KI) models: Demonstrate inhibited autophagy and suppressed DC function

  • Signaling Pathway Analysis:

    • Focus on the TAK1/JNK pathway, which is inhibited by TNFAIP8L2 in DCs

    • This inhibition results in downregulated autophagic activity

    • Western blot analysis of phosphorylated TAK1 and JNK provides insights into pathway activation

  • Functional Assays:

    • DC maturation markers (CD80, CD86, MHC-II) by flow cytometry

    • Cytokine production profiles (IL-12, TNF-α, IL-6)

    • T cell stimulatory capacity through mixed lymphocyte reactions

  • In Vivo Models:

    • LPS-induced endotoxemia reveals TNFAIP8L2's role in inflammation

    • Sepsis models provide physiologically relevant contexts

Research has demonstrated that TIPE2 can suppress the autophagic activity of DCs by inhibiting the TAK1/JNK signaling pathway, which negatively regulates the immune function of DCs during septic complications .

What are the technical challenges in optimizing immunohistochemistry protocols with TNFAIP8L2 antibodies?

Optimizing immunohistochemistry protocols for TNFAIP8L2 requires attention to several technical challenges:

  • Antigen Retrieval Optimization:

    • Heat-induced epitope retrieval using TE buffer at pH 9.0 is recommended

    • Alternative: citrate buffer at pH 6.0

    • Insufficient retrieval is a common cause of weak staining

  • Antibody Titration:

    • Start with the manufacturer's recommended range (typically 1:50-1:500)

    • Perform systematic titration experiments to determine optimal concentration

    • Different lots may require re-optimization

  • Detection System Selection:

    • Polymer-based detection systems generally provide superior sensitivity

    • DAB (3,3'-diaminobenzidine) as chromogen for brightfield microscopy

    • For fluorescence detection, Alexa Fluor-conjugated secondary antibodies

  • Tissue-Specific Considerations:

    • Recommended positive controls: human lymphoma tissue, human ovary tumor tissue, mouse spleen tissue

    • Fixation time affects epitope accessibility

    • Background staining can vary between tissue types

  • Troubleshooting Common Issues:

    • High background: Increase blocking time/concentration or antibody dilution

    • Weak signal: Decrease antibody dilution, extend incubation time, or enhance antigen retrieval

    • Non-specific binding: Increase washing steps or add 0.1% Triton X-100 to wash buffer

Validation data indicates that mouse spleen tissue sections show robust and specific staining when processed with appropriate retrieval methods and antibody concentrations .

How can researchers design effective TNFAIP8L2 knockout/knockdown models for studying inflammatory responses?

Creating effective TNFAIP8L2 knockout/knockdown models requires systematic approaches:

  • Knockout Strategy Options:

    • CRISPR/Cas9: Target early exons (exon 1 or 2) for complete disruption

    • Traditional homologous recombination: Replace critical exons with selection markers

    • Conditional knockout: Use Cre-loxP system for tissue-specific deletion

  • Knockdown Approaches:

    • siRNA: Design multiple siRNAs targeting different regions of TNFAIP8L2 mRNA

    • shRNA: For stable knockdown, use lentiviral vectors with selection markers

    • Consider knockdown efficiency and potential for off-target effects

  • Validation Methods:

    • Genomic verification: PCR and sequencing to confirm modifications

    • Transcript analysis: RT-qPCR to quantify TNFAIP8L2 mRNA levels

    • Protein verification: Western blot using validated antibodies

    • Functional validation: Assess known TNFAIP8L2-dependent pathways

  • Inflammatory Response Assessment:

    • Compare baseline vs. stimulated conditions (LPS, TNF-α, IL-1β)

    • Measure cytokine production (ELISA, multiplex assays)

    • Analyze signaling pathway activation (phospho-specific Western blots)

    • Assess cell-specific responses in different immune cell populations

Studies have demonstrated that TNFAIP8L2 knockout in dendritic cells significantly enhances autophagy and improves immune responses in sepsis models, providing valuable insights into the protein's role in regulating inflammatory responses .

What are the most effective approaches for investigating protein-protein interactions involving TNFAIP8L2?

To effectively study TNFAIP8L2 protein interactions:

  • Co-Immunoprecipitation (Co-IP):

    • Use antibodies specifically validated for immunoprecipitation (IP)

    • Optimize lysis conditions to preserve interactions (mild, non-denaturing buffers)

    • Include crucial controls:

      • Input controls (5-10% pre-IP lysate)

      • Negative controls (IgG isotype, TNFAIP8L2 knockout samples)

      • Reciprocal IP to confirm interactions

  • Proximity Ligation Assay (PLA):

    • Allows visualization of protein interactions in situ

    • Particularly useful for examining TNFAIP8L2 interactions with signaling molecules

    • Provides spatial information about interaction locations within cells

  • Competitive Binding Assays:

    • For studying TNFAIP8L2's competition with MTOR for RAC1 binding

    • Express increasing amounts of TNFAIP8L2 to assess displacement of MTOR

    • Quantify relative amounts of binding partners

  • Pull-down Assays:

    • Use purified recombinant TNFAIP8L2 as bait

    • GST-tagged or His-tagged constructs allow for efficient purification

    • Can identify direct vs. indirect interactions

  • Nucleotide State Manipulation:

    • For GTPase interactions (e.g., RAC1), control nucleotide binding state

    • Use GTPγS (non-hydrolyzable GTP analog) for GTP-bound state

    • Use GDP for GDP-bound state

    • Include magnesium in buffers to stabilize nucleotide binding

Research has demonstrated that TNFAIP8L2 can directly bind to and block RAC1 GTPase activity, and it can compete with MTOR for binding to the GTP-bound state of RAC1, providing critical insights into its regulatory functions .

How can researchers troubleshoot inconsistent results when using TNFAIP8L2 antibodies?

When encountering inconsistent results with TNFAIP8L2 antibodies, implement this systematic troubleshooting approach:

  • Antibody Validation:

    • Verify antibody specificity using TNFAIP8L2 knockout/knockdown controls

    • Test multiple antibodies targeting different epitopes

    • Check lot-to-lot consistency if using different batches

  • Sample Preparation Optimization:

    • Ensure complete protein denaturation for Western blot

    • Verify sample integrity (minimal protein degradation)

    • Consider post-translational modifications that might affect epitope recognition

    • Standardize lysate preparation protocols

  • Protocol Adjustments:

    • Western Blot:

      • Adjust protein loading (25-30 μg typically optimal)

      • Optimize gel percentage (12-15% recommended for 18-20 kDa TNFAIP8L2)

      • Try different transfer conditions and membrane types

    • Immunohistochemistry/Immunofluorescence:

      • Test different antigen retrieval methods

      • Adjust antibody concentration and incubation time

      • Modify blocking conditions to reduce background

  • Technical Variables to Control:

    • Buffer composition (pH, salt concentration, detergents)

    • Incubation temperature and duration

    • Secondary antibody selection and dilution

    • Detection system sensitivity

  • Tissue/Cell-Specific Considerations:

    • Expression levels vary across tissues (highest in immune tissues)

    • Cell activation status affects expression

    • Species differences in epitope sequences may affect cross-reactivity

Research data indicates that TNFAIP8L2 is consistently detected at 18-20 kDa in Western blot applications, despite its calculated molecular weight of 21 kDa, which could be due to post-translational modifications or cleavage .

What are the key methodological approaches for studying TNFAIP8L2's role in autophagy regulation?

To effectively investigate TNFAIP8L2's role in autophagy regulation:

  • Autophagy Flux Assessment:

    • LC3-II conversion: Western blot analysis of LC3-I to LC3-II conversion

    • p62/SQSTM1 degradation: Monitor levels as marker of autophagy completion

    • Tandem-tagged LC3 (mRFP-GFP-LC3): Distinguish autophagosomes from autolysosomes

    • Include bafilomycin A1 treatment to block autophagosome-lysosome fusion

  • Genetic Manipulation Approaches:

    • TNFAIP8L2 overexpression: Assess effects on autophagy markers

    • TNFAIP8L2 knockdown/knockout: Determine if autophagy is enhanced

    • Rescue experiments: Re-express TNFAIP8L2 in knockout models

  • Signaling Pathway Analysis:

    • MTORC1 activity: Monitor phosphorylation of S6K and 4E-BP1

    • RAC1 activity: GTP-bound RAC1 pull-down assays

    • TAK1/JNK pathway: Assess phosphorylation status by Western blot

    • Competition assays: Evaluate TNFAIP8L2 vs. MTOR for RAC1 binding

  • Autophagic Lysosome Reformation (ALR) Assessment:

    • LAMP1 staining: Monitor lysosomal morphology and distribution

    • Lysosomal pH measurements: LysoTracker or LysoSensor dyes

    • Live-cell imaging: Track dynamics of autophagosome-lysosome fusion

  • Physiological Context:

    • Nutrient starvation responses: Serum and glutamine deprivation

    • Inflammatory stimuli: LPS, TNF-α treatment

    • In vivo models: LPS-induced endotoxemia, sepsis models

Research has demonstrated that TNFAIP8L2 overexpression fails to induce autophagy flux despite suppressing MTOR activity under starvation conditions. Instead, it appears to impair autophagic lysosome reformation during prolonged starvation, highlighting its complex regulatory role in autophagy .

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.