PPP1R13L 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 the products within 1-3 business days after receiving your order. Delivery time may vary depending on the shipping method and destination. Please consult your local distributors for specific delivery timelines.
Synonyms
PPP1R13L antibody; IASPP antibody; NKIP1 antibody; PPP1R13BL antibody; RAI antibody; RelA-associated inhibitor antibody; Inhibitor of ASPP protein antibody; Protein iASPP antibody; NFkB-interacting protein 1 antibody; PPP1R13B-like protein antibody
Target Names
PPP1R13L
Uniprot No.

Target Background

Function
PPP1R13L Antibody, FITC conjugated, is a regulator that plays a crucial role in regulating apoptosis and transcription. This antibody interacts with NF-kappa-B and p53/TP53 proteins, influencing their functions. It inhibits transcription of HIV-1 virus by blocking the activity of both NF-kappa-B and SP1. Additionally, it inhibits p53/TP53 function, potentially by preventing the interaction between p53/TP53 and ASPP1 or ASPP2, thereby suppressing the subsequent activation of apoptosis.
Gene References Into Functions
  1. Haplotypes comprising PPP1R13L rs1970764 and ATM rs11212592 may be associated with lung cancer. Haplotypes containing PPP1R13L, CD3EAP, and GLTSCR1 SNPs on Chr19q13.3 may be associated with lung cancer risk in the Chinese population. PMID: 30128886
  2. The expression of CD3EAP exon 1 was shown to be significantly associated with PPP1R13L exon 1, while CD3EAP exon 3 was significantly associated with ERCC1 exon 11 in normal and non-small cell lung cancer (NSCLC) tissues. Observations indicate that short transcripts of ERCC1, CD3EAP, and PPP1R13L are co-expressed in the A549 NSCLC cell line. PMID: 29620255
  3. Increased expression of miR-150 suppressed viability, proliferation, migration, and invasion of SW480 cells. Furthermore, iASPP was a direct target of miR-150 and played a key role in its anti-colorectal cancer (CRC) function. miR-150 may serve as a promising predictor of prognosis in CRC patients. PMID: 29750311
  4. IASPP knockdown suppressed cell viability and DNA synthesis capacity; the effect of miR-340 inhibition was partially attenuated by iASPP inhibition. PMID: 29982095
  5. The expression of iASPP was found to be higher in high-grade astrocytic gliomas compared to low-grade astrocytic gliomas. PMID: 29257240
  6. Data indicate that iASPP can promote tumor growth by increasing autophagic flux, and iASPP could serve as a poor prognostic factor and a potential therapeutic target in lung cancer. PMID: 29072696
  7. Sertad1 could antagonize iASPP function by hindering its entry into the nucleus to interact with P53 in leukemic cells when iASPP is overproduced. PMID: 29179704
  8. The interactive modulation among miR-124 and iASPP in p53-mutant or -deleted cells may serve as a crucial pathway mediating therapy resistance when p53 is mutated or deleted, in the process of photodynamic therapy treatment of Colorectal cancer. PMID: 29022915
  9. These findings suggest that XIST may regulate tumor growth and metastasis via miR-140-dependent iASPP regulation. Taken together, the data indicate that XIST may be an oncogenic lncRNA that promotes the proliferation and metastasis of lung cancer through the regulation of miR-140 and could be considered a therapeutic target in human lung cancer. PMID: 28656261
  10. FHL2 and iASPP interacted with each other and co-localized in both the nucleus and cytoplasm. Silencing either FHL2 or iASPP could reduce cell proliferation, induce cell cycle arrest at the G0/G1 phase, and increase cell apoptosis. PMID: 28402264
  11. The restoration of miR-124 reduces iASPP expression and leads to p53-dependent tumor suppression, suggesting a therapeutic strategy to treat iASPP-associated cervical cancer. PMID: 27765948
  12. miR-124 regulates p63 via iASPP, while p63 targets miR-155 via the modulation of STAT1 expression in colorectal cancer. PMID: 28418858
  13. TP73-AS1 inhibited brain glioma growth and metastasis as a competing endogenous RNA (ceRNA) through miR-124-dependent iASPP regulation. PMID: 29412778
  14. Findings revealed the detailed role of the miR-184/iASPP axis in Central nervous system lymphoma (CNSL), and this axis might modulate the proliferation and invasion of CNSL via regulating the PI3K/Akt signaling pathway. PMID: 28012196
  15. Data suggest that Keap1, rather than Nrf2, is critical for the recruitment of iASPP into the Keap1-Nrf2 complex. PMID: 29033244
  16. Three htSNPs (haplotype-tagging single nucleotide polymorphism) (rs7354, rs14384, and rs3783501) covering 95% of the common haplotype diversity in 19p13.3-GADD45B and interaction of 19p13.3-GADD45B and 19q13.3-PPP1R13L and 19q13.3-CD3EAP variants and smoking-duration were explored in lung cancer risk Chinese. PMID: 28870783
  17. This report describes the identification of a maternally inherited frameshift mutation in RAI1, causative for SMS. This is the first report about the transmission of SMS from an affected parent to an offspring. PMID: 27683195
  18. These results identified PPP1R13L as the gene underlying a novel autosomal-recessive cardio-cutaneous syndrome in humans and strongly suggest that the fatal dilated cardiomyopathy during infancy is a consequence of failure to regulate transcriptional pathways necessary for tuning cardiac threshold response to common inflammatory stressors. PMID: 28069640
  19. UCA1 might promote proliferation and migration of glioma, to regulate tumor growth and metastasis via miR-182 dependent iASPP regulation. PMID: 28137422
  20. Results demonstrate that iASPP is overexpressed in bladder cancer and promotes the malignancy of bladder cancer. PMID: 28489738
  21. lncRNA H19 interacts with miR-140 to modulate glioma growth by targeting iASPP. PMID: 27693036
  22. Increased expression of p53 and ASPP1 and downregulation of iASPP. PMID: 27177208
  23. Our study provides the first evidence that high iASPP-SV expression may be a novel prognostic factor and therapeutic target for glioma. PMID: 26628298
  24. We were able to reproduce previously found associations between PPP1R13L and CD3EAP polymorphisms and lung cancer risk in an increased study group, and we found interactions between NFKB1 rs28362491-PPP1R13L rs1970764 and smoking duration and between CD3EAP rs735482 and smoking duration. PMID: 26563375
  25. The inhibitor of apoptosis-stimulating protein of p53 (iASPP) was identified to be a direct target of miR-140 in pancreatic duct adenocarcinoma specimens and cell lines. PMID: 26787707
  26. We demonstrate that iASPP is a novel substrate of caspases in response to apoptotic stimuli. PMID: 26646590
  27. One novel region within PPP1R13L is hypomethylated in all transient neonatal diabetes type 1 patients included in this study. PMID: 27075368
  28. Destabilization of p300/CBP by downregulation of iASPP expression levels appears to represent a molecular mechanism that contributes to chemoresistance in melanoma cells. PMID: 25675294
  29. One of the proteins identified, iASPP, showed reduced levels in the presence of GSK-3. Further, blocking iASPP activity increased cell death, particularly in p53 wild-type BC3 PEL cells. PMID: 26109723
  30. The present study showed that iASPP is overexpressed in oral cavity squamous cell carcinomas (OSCC) tissues, and increased cytoplasmic iASPP is correlated with recurrence and poor survival outcomes in OSCC patients. PMID: 25149434
  31. Results suggest that NFKB1 common variants and smoking duration and smoking duration-PPP1R13L rs1970764 interaction could be associated with lung cancer development in a Chinese population. PMID: 25917613
  32. iASPP expression may act as a predictive marker of prostate cancer progression. PMID: 25341046
  33. Data highlight the importance of 14-3-3 proteins in antiviral responses and identify RelA-associated inhibitor and sirtuin 1 as novel regulators of antiviral innate immune responses. PMID: 24997996
  34. These data demonstrate that by interacting with desmoplakin and desmin, iASPP is an important regulator of desmosomal function both in vitro and in vivo. PMID: 25691752
  35. Authors have identified a novel mechanism modulating autophagy in keratinocytes that relies upon iASPP expression specifically reducing the interaction of Atg5-Atg12 with Atg16L1. PMID: 24777476
  36. MIRN124 binds to the 3'UTR of iASPP and suppresses mRNA expression and the proliferation of prostate tumor cells. PMID: 24966937
  37. Hematopoietic cells can be protected against apoptosis by iASPP. PMID: 24668753
  38. rs6966 (3' UTR of PPP1R13L, chr 19q13.32, P = 4.55 x 10(-9)) and rs414580 (intron 2 of MSR1, chr 8p22, P = 6.09 x 10(-8)) were significantly associated with ALL. PMID: 24604828
  39. Haplotype PPP1R13L rs4803817 polymorphism is associated with lung cancer risk. PMID: 24140460
  40. Overexpression of iASPP and the low expression of caspase-9 in esophageal cancer are closely correlated with tumor invasion and metastasis. PMID: 24405603
  41. A higher rate of Helicobacter pylori infection, an increased expression of inhibitor of apoptosis stimulating protein of p53 (iASPP), and decreased expression of apoptosis-stimulating of p53 protein 2(ASPP2) were present in gastric cancer. PMID: 23528480
  42. This study thus demonstrates that iASPP is highly elevated in human cervical cancer, and that overexpression of nuclear iASPP is correlated with poor prognosis and chemoresistance/radioresistance. PMID: 23420450
  43. Downregulation of miR-124 promotes the growth and invasiveness of glioblastoma cells involving upregulation of PPP1R13L. PMID: 23624869
  44. The miR-124/iASPP axis can regulate the proliferation of colorectal cancer cells. PMID: 23691514
  45. PPP1R13L and CD3EAP variants may be associated with lung cancer risk in nonsmoking Chinese women. PMID: 23624123
  46. Results suggest that iASPP may contribute to the malignant progression of head and neck squamous cell carcinoma. PMID: 22815155
  47. The PPP1R13L rs1970764 variant is a possible prognostic marker for patients with rectal cancer. PMID: 23180017
  48. When the Px(T)PxR motif is deleted or mutated via insertion of a phosphorylation site mimic (T311D), PP-1c fails to bind to all three ASPP proteins, ASPP1, ASPP2, and iASPP. PMID: 23088536
  49. These findings showed that iAPSS/iASPPsv reduced the growth inhibition and apoptosis induced by Dex or VP-16, with DNA damage accumulating which might promote the pathogenesis and/or progression of cancer. PMID: 22766503
  50. iASPP inhibited apoptosis independently of p53 in tumor cells, mainly by inhibiting the transcriptional activity of p63/p73 on the promoters of proapoptotic genes. PMID: 22538442

Show More

Hide All

Database Links

HGNC: 18838

OMIM: 607463

KEGG: hsa:10848

STRING: 9606.ENSP00000354218

UniGene: Hs.466937

Protein Families
ASPP family
Subcellular Location
Cytoplasm. Nucleus.
Tissue Specificity
Highly expressed in heart, placenta and prostate. Weakly expressed in brain, liver, skeletal muscle, testis and peripheral blood leukocyte.

Q&A

What is the biological role of PPP1R13L in cellular pathways and why is it a significant target for antibody detection?

PPP1R13L (also known as iASPP) functions as a critical regulator in apoptosis and transcription pathways through its interactions with two key proteins:

  • It binds to NF-kappa-B and inhibits its transcriptional activity

  • It interacts with p53/TP53, potentially preventing associations between p53/TP53 and ASPP1 or ASPP2, thereby suppressing apoptosis activation

Additional biological functions include:

  • Blocking HIV-1 virus transcription by inhibiting both NF-kappa-B and SP1 action

  • Potentially acting as an oncoprotein, with overexpression accelerating tumor formation in RAS/E1A transformed cells

  • Serving as a novel autophagy inhibitor in keratinocytes

This multifunctional nature makes PPP1R13L an important target for research in oncology, virology, and cell death pathways.

What are the optimal protocols for using FITC-conjugated PPP1R13L antibodies in immunofluorescence applications?

For optimal results with FITC-conjugated PPP1R13L antibodies in immunofluorescence applications:

Sample Preparation Protocol:

  • Fix cells in Solution A fixative for 15 minutes at room temperature

  • Wash in PBS buffer

  • Permeabilize with 0.25% Triton X-100 for 15 minutes

  • Wash in PBS buffer

  • Add FITC-conjugated anti-PPP1R13L antibody at 1:50-200 dilution

  • Incubate for 30 minutes at room temperature

  • Wash in PBS buffer

  • Counter-stain nuclei with DAPI (200 ng/ml)

  • Mount and visualize using appropriate fluorescence microscopy settings for FITC detection

Optimization Notes:

  • Signal intensity can be confirmed by flow cytometry using median fluorescence intensity (MFI) comparison to isotype controls

  • When analyzing subcellular localization, co-staining with organelle markers (such as KDEL for ER, GOLGA2/GM130 for Golgi) may be helpful

  • For tissue sections, antigen retrieval with TE buffer pH 9.0 is recommended

How can researchers troubleshoot non-specific binding or weak signals when using PPP1R13L antibodies in Western blotting?

When encountering issues with PPP1R13L antibody performance in Western blotting:

Troubleshooting Weak Signals:

  • Adjust concentration: Use dilution ranges between 1:500-1:2000 for most applications

  • Consider protein abundance: PPP1R13L is expressed predominantly in epithelial cells, skin, testis, heart, and stomach tissues

  • Verify expected molecular weight: Look for bands approximately 100 kDa (isoform 1) and 50 kDa (isoform 2)

  • Observed molecular weight may appear as 110 kDa in some cell types

Reducing Non-specific Binding:

  • Use affinity-purified antibodies that have been validated against the specific epitope

  • Include appropriate blocking agents (BSA or non-fat milk)

  • Increase washing steps and duration

  • Consider positive controls: HEK-293 cells and human heart tissue show reliable detection

For accurate controls, use proteasome inhibitor MG132 with PPP1R13L-overexpressing cells, which characteristically shows p53 accumulation .

What are the comparative advantages of using FITC-conjugated versus unconjugated PPP1R13L antibodies in different experimental contexts?

FITC-Conjugated PPP1R13L Antibodies:

Advantages:

  • Direct detection without secondary antibody requirements

  • Reduced protocol time and fewer washing steps

  • Elimination of potential cross-reactivity from secondary antibodies

  • Suitable for multicolor immunofluorescence when combined with other differently-conjugated primary antibodies

  • Effective for flow cytometry applications with minimal background

Limitations:

  • Fixed fluorophore with specific excitation/emission properties

  • Cannot leverage signal amplification via secondary antibodies

  • May have reduced sensitivity compared to enzyme-based detection systems

Unconjugated PPP1R13L Antibodies:

Advantages:

  • Versatility across multiple detection methods (WB, ELISA, IHC, IF)

  • Signal amplification possible through optimized secondary antibody systems

  • Compatible with various detection methods (chemiluminescence, colorimetric)

  • Generally longer shelf-life without photobleaching concerns

Limitations:

  • Requires additional incubation steps and reagents

  • Potential for cross-reactivity with secondary antibodies

Application-Specific Selection Guide:

  • Choose FITC-conjugated for direct immunofluorescence, flow cytometry, or multi-color staining

  • Select unconjugated for western blotting, where signal amplification is beneficial, or when greater flexibility in detection methods is needed

How can researchers effectively validate the specificity of PPP1R13L antibodies in their experimental systems?

Comprehensive Antibody Validation Strategy:

  • Genetic Approaches:

    • Compare antibody reactivity in wild-type versus PPP1R13L knockdown/knockout cells

    • Documented cases show characteristic changes in autophagy markers (LC3B) when PPP1R13L/iASPP is knocked down in HaCaT or N-TERT cells

  • Epitope Mapping Verification:

    • Verify antibody binding to the specific epitope region

    • For example, confirm whether the antibody targets amino acids 775-800 of human iASPP (isoform 1)

  • Cross-Reactivity Assessment:

    • Test across multiple species (human, mouse, rat) if cross-reactivity is claimed

    • Perform peptide competition assays with the immunizing peptide

  • Functional Validation:

    • In PPP1R13L-overexpressing cells, verify functional impact on p53 levels and NF-κB activity

    • Characteristic finding: PPP1R13L overexpression causes faster p53 degradation through proteasome pathway

  • Application-Specific Controls:

    • For FITC-conjugated antibodies, include isotype control with matched FITC:protein ratio

    • For immunoprecipitation, perform reverse IP and validate with mass spectrometry

Key Expected Observations for Valid Antibodies:

  • In Western blot: Bands at approximately 100 kDa and 50 kDa corresponding to isoforms 1 and 2

  • In co-immunoprecipitation: Detection of PPP1R13L interactions with Atg5–Atg12 and Atg16L1 complexes

  • In overexpression studies: Inhibition of p53-dependent apoptosis pathways

What are the key considerations for designing experiments to study PPP1R13L's role in apoptosis using antibody-based detection methods?

When designing experiments to investigate PPP1R13L's role in apoptosis:

Experimental Design Framework:

  • Cell Model Selection:

    • Primary mouse embryonic fibroblasts (MEFs) transformed with HRAS and E1A provide well-characterized models with defined p53 status

    • Epithelial cell lines (HaCaT, N-TERT) are appropriate given PPP1R13L's predominant expression in epithelial tissues

  • Manipulation Approaches:

    • Overexpression using retroviral vectors expressing PPP1R13L

    • Knockdown using specific shRNA constructs targeting PPP1R13L/iASPP

    • Caspase cleavage studies using anti-Fas antibody to trigger apoptosis

  • Key Markers to Monitor:

    • p53 levels and activation status

    • NF-κB p65/RelA activity

    • Apoptotic markers (cleaved PARP, pro-caspase 8, pro-caspase 9)

    • Bcl-2 family proteins (Puma, Noxa, Mcl-1, Bim)

  • Control Conditions:

    • Proteasome inhibitor MG132 treatment to assess protein degradation pathways

    • p53-null versus p53-wildtype conditions to distinguish p53-dependent and -independent effects

    • BrdU incorporation to monitor proliferation in parallel with apoptosis assessment

Specific Analytical Methods:

  • Co-immunoprecipitation to detect PPP1R13L interactions with p53 and NF-κB

  • Immunofluorescence to track subcellular localization during apoptosis induction

  • Western blotting to monitor protein expression levels and cleavage products

  • Flow cytometry with FITC-conjugated anti-PPP1R13L to quantify expression levels in apoptotic versus non-apoptotic populations

How does PPP1R13L expression correlate with tumor development, and what antibody-based approaches are most effective for studying this relationship?

PPP1R13L appears to have significant oncogenic properties based on several lines of research:

Expression Patterns in Cancer:

  • Overexpression detected in eight human breast carcinomas expressing wild-type p53

  • Elevated expression observed in certain leukemias

  • Expression frequently upregulated in multiple human cancer types

Functional Impact on Tumorigenesis:

  • Overexpression of PPP1R13L strongly accelerates tumor formation by RAS/E1A transformed cells

  • Creates phenotype with multiple tumor nodes consistent with increased metastasis

  • Modulates both p53-dependent and -independent apoptosis pathways

Optimal Antibody-Based Approaches:

  • Tissue Microarray Analysis:

    • Use Anti-PPP1R13L antibodies at 1:50-1:500 dilution for IHC

    • Compare expression across tumor grades and correlate with p53 status

    • Recommend antigen retrieval with TE buffer pH 9.0 for optimal results

  • In vivo Tumor Models:

    • Immunohistochemical staining of tumor xenografts derived from PPP1R13L-overexpressing cells

    • Correlate PPP1R13L expression with metastatic potential and tumor growth rate

  • Mechanistic Studies:

    • Co-immunoprecipitation to evaluate PPP1R13L interaction with p53 in tumor samples

    • Western blotting to assess PPP1R13L-induced effects on p53 degradation

    • Analyze PPP1R13L's impact on autophagy inhibition in cancer cells using LC3B labeling

Research Applications Table:

TechniqueAntibody DilutionKey Endpoint MeasurementsAdvantage in Cancer Research
IHC1:50-1:500Expression patterns in tumor tissueCorrelate with clinical outcomes
WB1:500-1:2000Protein level quantificationCompare across cell lines and patient samples
IF (FITC)1:50-200Subcellular localizationVisualize in tumor sections
IPVariableProtein-protein interactionsIdentify cancer-specific binding partners

What are the most effective protocols for using PPP1R13L antibodies in co-immunoprecipitation experiments to study protein-protein interactions?

For successful co-immunoprecipitation of PPP1R13L and its binding partners:

Optimized Co-IP Protocol:

  • Cell Lysis:

    • Use lysis buffer containing: 1 M Tris, 2.5 M NaCl, 10% glycerol, 0.5 M glycerophosphate, 1% Tween-20, 0.5% Nonidet P-40, and EDTA-free complete protease inhibitor

    • Incubate for 15 minutes on ice

    • Clear lysates by centrifugation

  • Immunoprecipitation:

    • Incubate 1 mg of cleared cell lysate with 1 μg of antibody against target protein (e.g., Atg5–Atg12 or Atg16L1)

    • Alternatively, use anti-PPP1R13L antibody for reverse co-IP

    • Incubate at 4°C overnight

  • Precipitation and Washing:

    • Add 50 μl Protein-G–Sepharose beads

    • Incubate for 2 hours at 4°C

    • Precipitate and wash three times with NP-40 lysis buffer

  • Detection:

    • Subject immunoprecipitates to SDS-PAGE

    • Perform Western blot analysis for PPP1R13L and interacting partners

Quantification Method:
For quantitative analysis of binding interactions, calculate the percentage of proteins in complex using densitometry values:

  • Percentage of Atg5 bound to PPP1R13L/iASPP = [(Atg5 IP/Atg5 input)/(iASPP IP/iASPP input)×100]

  • Percentage of Atg5 bound to Atg16L1 = [(Atg5 IP/Atg5 input)/(Atg16L1 IP/Atg16L1 input)×100]

Key Interacting Partners to Study:

  • p53/TP53 (tumor suppressor)

  • NF-κB p65/RelA (transcription factor)

  • Atg5–Atg12 complex (autophagy machinery)

  • Atg16L1 (autophagy machinery)

  • ASPP1 or ASPP2 (apoptosis-stimulating proteins)

How do different fixation and antigen retrieval methods affect the performance of PPP1R13L antibodies in immunohistochemistry?

Fixation and antigen retrieval significantly impact PPP1R13L antibody performance in immunohistochemistry:

Fixation Methods Comparison:

Fixation MethodImpact on PPP1R13L DetectionRecommended Antibody Dilution
Formalin fixation (FFPE tissue)Standard method, preserves tissue morphology but may mask epitopes1:50-1:500
Solution A fixativeEffective for cell suspensions, preserves FITC signalAs recommended by manufacturer
Fresh frozen tissueMaintains better antigenicity but poorer morphologyTypically requires less dilute antibody

Antigen Retrieval Optimization:

  • Heat-Induced Epitope Retrieval (HIER):

    • TE buffer pH 9.0 (recommended primary method)

    • Citrate buffer pH 6.0 (alternative method)

    • Heat source options: microwave, pressure cooker, water bath

  • Enzymatic Retrieval:

    • Generally less effective for PPP1R13L detection

    • May be tested if HIER methods fail

Critical Factors Affecting Performance:

  • Epitope Accessibility:
    PPP1R13L antibodies targeting different regions (e.g., AA 775-800 vs. AA 83-102) may require different retrieval methods

  • Tissue Type Considerations:

    • Human breast cancer tissue shows reliable PPP1R13L detection

    • Epithelial tissues generally show stronger signals due to higher endogenous expression

  • Detection Systems:

    • DAB-based systems for brightfield microscopy

    • Fluorescent secondary antibodies for immunofluorescence

    • For FITC-conjugated antibodies, ensure fixation methods don't compromise fluorophore activity

Validation Approach:
Include a gradient of antibody dilutions (1:50, 1:100, 1:250, 1:500) and multiple antigen retrieval methods on serial sections of the same tissue to identify optimal conditions for each specific application.

What considerations should researchers take into account when studying the role of PPP1R13L in autophagy inhibition using antibody-based techniques?

When investigating PPP1R13L's role as an autophagy inhibitor:

Experimental Design Considerations:

  • Cell Model Selection:

    • Keratinocytes (HaCaT, N-TERT) are well-validated models for studying PPP1R13L's impact on autophagy

    • Compare wildtype versus PPP1R13L/iASPP knockdown cells

  • Autophagy Induction Methods:

    • Nutrient starvation (serum-free medium)

    • Chemical inducers (rapamycin, torin)

    • Differentiation-induced autophagy

  • Key Markers to Monitor:

    • LC3B conversion (LC3-I to LC3-II) by Western blot

    • LC3B puncta formation by immunofluorescence

    • Autophagic flux by p62 degradation

    • Atg5–Atg12 complex formation

    • Atg16L1 interaction with PPP1R13L

Antibody-Based Methodologies:

  • Immunofluorescence Protocol for LC3B Labeling:

    • Fix cells following treatment conditions

    • Permeabilize with 0.25% Triton X-100

    • Incubate with anti-LC3B antibody (0.25 μg)

    • Visualize with Alexa-Fluor-647-conjugated secondary antibody

    • Analyze median fluorescence intensity and puncta formation

  • Co-immunoprecipitation for PPP1R13L-Autophagy Protein Interactions:

    • Immunoprecipitate endogenous Atg5–Atg12 and Atg16L1

    • Detect PPP1R13L co-precipitation by Western blot

    • Quantify interaction using densitometry

  • Molecular Mechanisms to Investigate:

    • PPP1R13L competition with Atg16L1 for binding to Atg5–Atg12 complex

    • Impact on autophagosome formation

    • Effect on autophagic flux

Controls and Validation:

  • Include rapamycin treatment as positive control for autophagy induction

  • Monitor mTOR pathway activity (phospho-S6, phospho-4E-BP1, phospho-mTOR)

  • Compare autophagy in differentiated versus undifferentiated cells

  • Include Beclin-1 detection as additional autophagy marker

This detailed approach will enable researchers to comprehensively characterize PPP1R13L's role in autophagy regulation using antibody-based techniques.

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.