PPP1R13L Antibody

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 orders. Delivery times may vary depending on the purchasing method or location. Please consult your local distributor for specific delivery times.
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
This regulator plays a pivotal role in the regulation of apoptosis and transcription through its interaction with NF-kappa-B and p53/TP53 proteins. It effectively blocks the transcription of HIV-1 virus by inhibiting the activity of both NF-kappa-B and SP1. Additionally, it inhibits p53/TP53 function, potentially by preventing the association between p53/TP53 and ASPP1 or ASPP2, thereby suppressing the subsequent activation of apoptosis.
Gene References Into Functions
  1. Haplotypes composed of PPP1R13L rs1970764 and ATM rs11212592 may be associated with lung cancer. Furthermore, haplotypes comprising PPP1R13L, CD3EAP, and GLTSCR1 SNPs on Chr19q13.3 may be linked to lung cancer risk within the Chinese population. PMID: 30128886
  2. The expression of CD3EAP exon 1 has been demonstrated to be significantly associated with PPP1R13L exon 1, while CD3EAP exon 3 exhibits a significant association with ERCC1 exon 11 in both normal and non-small cell lung cancer (NSCLC) tissues. Notably, short transcripts of ERCC1, CD3EAP, and PPP1R13L are co-expressed in the A549 NSCLC cell line. PMID: 29620255
  3. Elevated expression of miR-150 suppressed the viability, proliferation, migration, and invasion of SW480 cells. Moreover, iASPP was identified as a direct target of miR-150 and plays a key role in its anti-colorectal cancer (CRC) function. miR-150 has the potential to serve as a promising predictor of prognosis in CRC patients. PMID: 29750311
  4. Knockdown of iASPP 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 suggest that iASPP can promote tumor growth by increasing autophagic flux. Furthermore, iASPP may serve as a poor prognostic factor and a potential therapeutic target in lung cancer. PMID: 29072696
  7. Sertad1 can antagonize iASPP function by hindering its entry into the nucleus to interact with P53 in leukemic cells, especially when iASPP is overproduced. PMID: 29179704
  8. The interactive modulation between miR-124 and iASPP in p53-mutant or -deleted cells may represent a crucial pathway mediating therapy resistance during photodynamic therapy treatment of colorectal cancer when p53 is mutated or deleted. PMID: 29022915
  9. These findings indicate that XIST may regulate tumor growth and metastasis through miR-140-dependent iASPP regulation. In summary, our data suggest that XIST may be an oncogenic lncRNA that promotes the proliferation and metastasis of lung cancer via the regulation of miR-140 and could be considered a therapeutic target in human lung cancer. PMID: 28656261
  10. FHL2 and iASPP interact with each other and co-localize in both the nucleus and cytoplasm. Silencing either FHL2 or iASPP can 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 potential therapeutic strategy for treating iASPP-associated cervical cancer. PMID: 27765948
  12. miR-124 regulates p63 through iASPP, while p63 targets miR-155 via the modulation of STAT1 expression in colorectal cancer. PMID: 28418858
  13. TP73-AS1 inhibits brain glioma growth and metastasis as a competing endogenous RNA (ceRNA) through miR-124-dependent iASPP regulation. PMID: 29412778
  14. Findings reveal 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 by regulating the PI3K/Akt signaling pathway. PMID: 28012196
  15. Data suggest that Keap1, rather than Nrf2, is crucial 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 the interaction of 19p13.3-GADD45B and 19q13.3-PPP1R13L and 19q13.3-CD3EAP variants and smoking duration were explored in relation to lung cancer risk in the Chinese population. PMID: 28870783
  17. We report the identification of a maternally inherited frameshift mutation in RAI1, which is causative for Smith-Magenis syndrome (SMS). This represents the first report of transmission of SMS from an affected parent to an offspring. PMID: 27683195
  18. These results identify 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 a failure to regulate transcriptional pathways essential for tuning cardiac threshold responses to common inflammatory stressors. PMID: 28069640
  19. UCA1 might promote proliferation and migration of glioma, regulating tumor growth and metastasis through 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 were observed. 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 replicate previously observed associations between PPP1R13L and CD3EAP polymorphisms and lung cancer risk in an expanded 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 as 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. The 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. Furthermore, blocking iASPP activity increased cell death, particularly in p53 wild-type BC3 PEL cells. PMID: 26109723
  30. The current study revealed 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 the interaction between smoking duration and PPP1R13L rs1970764, 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 a significant 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, suppressing 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 acute lymphoblastic leukemia (ALL). PMID: 24604828
  39. The haplotype PPP1R13L rs4803817 polymorphism is associated with lung cancer risk. PMID: 24140460
  40. Overexpression of iASPP and 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, increased expression of the 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 demonstrates that iASPP is highly elevated in human cervical cancer, and 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 through the 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 demonstrate 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 inhibits apoptosis independently of p53 in tumor cells, primarily 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 PPP1R13L and why is it important in research?

PPP1R13L (Protein Phosphatase 1, Regulatory Subunit 13 Like), also known as iASPP, is a highly conserved inhibitor of p53 that selectively regulates a subset of p53 target genes. It was initially identified as a protein that binds to the NF-κB subunit p65/RelA and inhibits its transcriptional activity . PPP1R13L has emerged as an important research target due to its involvement in tumor progression, particularly in cervical cancer, acute myeloid leukemia, and other malignancies . It plays critical roles in cellular processes including apoptosis regulation, cell proliferation, epithelial-mesenchymal transition, and glycolysis, making it a significant target for cancer research .

What are the different isoforms of PPP1R13L and how do they affect antibody selection?

PPP1R13L exists in multiple isoforms, with isoform 1 (~100 kDa) and isoform 2 (~50 kDa) being the most commonly studied . When selecting an antibody, researchers should consider which isoform they intend to detect. Some antibodies are specifically designed to target epitopes present in isoform 1, such as those binding to AA 775-800 or AA 780-797 . Additionally, PPP1R13L can be cleaved by Caspase-3, producing a stable PPP1R13L (295-828aa) fragment that relocates from the cytoplasm to the nucleus, which is critical for its inhibition of p53 transcription . For studies focusing on the nuclear functions of PPP1R13L, antibodies recognizing the C-terminal region containing the Ank-SH3 domain would be most appropriate.

What are the synonyms for PPP1R13L that researchers should be aware of?

When searching literature or antibody databases, researchers should be aware of multiple synonyms for PPP1R13L, including:

  • iASPP (inhibitor of apoptosis stimulating protein of p53)

  • Inhibitor of ASPP protein

  • RelA-associated inhibitor (RAI)

  • NFkB interacting protein 1 (NKIP1)

  • PPP1R13B-like protein (PPP1R13BL)

This awareness is crucial for comprehensive literature searches and avoiding confusion when comparing research findings that use different nomenclature.

What are the validated applications for PPP1R13L antibodies in cancer research?

PPP1R13L antibodies have been validated for multiple applications in cancer research:

ApplicationValidated in tumor typesKey findings
Western BlottingCervical cancer, AML, breast cancerDetection of altered PPP1R13L expression levels correlating with cancer progression
ImmunohistochemistryBreast cancer, cervical cancerTissue localization and expression pattern analysis
ImmunofluorescenceCervical cancerSubcellular localization studies
ImmunoprecipitationVarious cancer cell linesProtein-protein interaction studies with p53, p63, and NF-κB

Research indicates that PPP1R13L promotes cervical cancer progression by suppressing p63-mediated PTEN transcription, leading to activation of the AKT/mTOR pathway . In acute myeloid leukemia, high iASPP expression correlates with poor clinical outcomes, making antibody-based detection methods valuable prognostic tools .

How can PPP1R13L antibodies be used to study its role in apoptosis regulation?

PPP1R13L antibodies can be employed to investigate its dual role in apoptosis regulation through several methodological approaches:

  • Co-immunoprecipitation experiments: Use PPP1R13L antibodies to pull down protein complexes and analyze interactions with p53, p63, p73, and NF-κB subunits to understand how PPP1R13L mediates apoptotic signaling .

  • Chromatin immunoprecipitation (ChIP): Employ PPP1R13L antibodies in ChIP assays to examine how PPP1R13L affects p53 family member binding to promoters of pro-apoptotic genes like PTEN .

  • Immunofluorescence: Detect subcellular localization changes of PPP1R13L following apoptotic stimuli, particularly its translocation from cytoplasm to nucleus after Caspase-3 cleavage .

  • Western blotting: Measure PPP1R13L expression levels in cells undergoing apoptosis, potentially revealing correlation between PPP1R13L levels and apoptotic resistance in cancer cells .

Research has shown that PPP1R13L can have both pro-apoptotic and anti-apoptotic effects depending on the cellular context and experimental conditions , making careful antibody selection and experimental design crucial.

What are the recommended dilutions and conditions for using PPP1R13L antibodies in different applications?

ApplicationRecommended DilutionBuffer ConditionsSpecial Considerations
Western Blotting1:500-1:2000Standard transfer bufferExpect bands at ~100 kDa (isoform 1) and ~50 kDa (isoform 2)
Immunohistochemistry1:50-1:500Antigen retrieval with TE buffer pH 9.0 or citrate buffer pH 6.0Sample-dependent optimization required
ELISA1:40,000-1:160,000Standard ELISA buffersHigh dilution due to antibody sensitivity
Immunofluorescence1:100-1:500PBS-based buffersSubcellular localization varies by cell type

For optimal results, researchers should always perform titration experiments to determine the ideal antibody concentration for their specific experimental system and sample type .

What cross-reactivity should be considered when selecting a PPP1R13L antibody?

When selecting a PPP1R13L antibody, consider these cross-reactivity factors:

  • Species reactivity: Different antibodies show varying reactivity profiles. For example:

    • ABIN129682: Reactive with human samples only

    • ABIN401339: Reactive with human, mouse, and dog samples, with partial reactivity to bovine specimens

    • 51141-1-AP: Validated for human and mouse samples

  • Isoform specificity: Ensure the antibody recognizes the specific isoform of interest. Some antibodies are designed to target specific amino acid regions like AA 775-800 or AA 780-797 in isoform 1 .

  • Domain recognition: For functional studies, select antibodies that recognize specific domains such as the Ank-SH3 domain, which mediates interactions with p53 family members .

  • Related protein discrimination: Verify the antibody can discriminate between PPP1R13L and its family members ASPP1 and ASPP2, which share structural similarity in their C-terminal regions .

Cross-reactivity information should be experimentally validated in your system, especially when working with less commonly studied species or tissue types.

How can antibodies help elucidate the selective regulation of p53 family target genes by PPP1R13L?

Understanding the sequence-specific regulation of p53 family target genes by PPP1R13L requires sophisticated antibody-based approaches:

  • ChIP-seq analysis: Use PPP1R13L antibodies in ChIP-seq experiments to identify genome-wide binding sites. Research has identified sequence signatures of PPP1R13L-regulated p53 response elements (REs), which feature C9 and/or G12 in addition to the typical p53 RE characteristics .

  • Re-ChIP experiments: Sequential ChIP using antibodies against PPP1R13L followed by p53, p63, or p73 antibodies to identify regions where these proteins co-localize on DNA.

  • Proximity ligation assays: Visualize and quantify specific PPP1R13L-p53 family protein interactions at different p53 target gene promoters using paired antibodies.

  • CRISPR-engineered mutant studies: Create specific mutations in the response elements of PPP1R13L-regulated genes (like the identified site1 in PTEN with pattern of C4, G7, C9, A12, C14, and G17), then use antibodies to assess altered binding patterns .

Research has shown that PPP1R13L specifically inhibits the transcriptional activity of TAp63 on PTEN through interaction with a specific response element, with C9 being a key element in this selective regulation . This discovery provides a sequence basis for understanding how PPP1R13L selectively regulates only certain p53 family target genes.

How does p53 mutation status affect PPP1R13L function and antibody-based detection methods?

The interaction between PPP1R13L and mutant p53 presents unique challenges for antibody-based research:

  • Opposing functional effects: In C33A cells (HPV-negative cervical cancer cells with p53 R273C mutation), PPP1R13L exhibits effects opposite to those observed with wild-type p53. Specifically, in these cells, PPP1R13L paradoxically promotes rather than inhibits the transcriptional activity of certain promoters .

  • Tetramer formation considerations: Since p53 functions as a tetramer, even overexpression of wild-type p53 cannot reverse the effects of endogenous mutant p53. When designing co-immunoprecipitation experiments with PPP1R13L antibodies, researchers must account for mixed tetramers of wild-type and mutant p53 .

  • p53-independent p63 inhibition: In p53-mutant cells, PPP1R13L can still inhibit p63 independently of p53. This requires careful interpretation of antibody-based detection results to distinguish direct effects on p63 from those mediated through p53 .

  • Modified validation protocols: For cells with p53 mutations, additional validation steps are needed when using PPP1R13L antibodies, including:

    • Parallel immunoprecipitation with both p53 and p63 antibodies

    • Controls with p53-null cells

    • Domain-specific antibodies to map interaction regions

These considerations are essential when studying cancers with high rates of p53 mutations, as PPP1R13L may exhibit context-dependent functions that vary based on the specific p53 mutation present .

What controls should be included when using PPP1R13L antibodies for research?

To ensure reliable results when using PPP1R13L antibodies, incorporate these essential controls:

  • Positive tissue/cell controls:

    • Human heart tissue, HEK-293 cells, and mouse testis tissue have been validated as positive controls for Western blot

    • Human breast cancer tissue serves as a positive control for immunohistochemistry

  • Knockdown/knockout validation:

    • Include PPP1R13L knockdown samples to confirm antibody specificity, as demonstrated in studies where PPP1R13L mRNA level was lower in patient-derived fibroblasts compared to controls

    • Western blot analysis confirming absence of the ~100 kDa band in PPP1R13L-deficient cells

  • Peptide competition assays:

    • Pre-incubate antibody with the immunizing peptide (e.g., synthetic peptide corresponding to amino acids 780-797 of human iASPP isoform 1)

    • Signal should be specifically blocked by the competing peptide

  • Isotype controls:

    • Include the appropriate isotype control (e.g., rabbit IgG for polyclonal antibodies or mouse IgG2b for monoclonal antibodies)

    • Use at the same concentration as the primary antibody

  • Cross-validation with multiple antibodies:

    • Whenever possible, confirm results using antibodies targeting different epitopes of PPP1R13L to rule out non-specific binding

How can researchers address discrepancies in PPP1R13L detection between RNA and protein levels?

Discrepancies between PPP1R13L RNA and protein levels are common and can be addressed through several methodological approaches:

  • Integrated RNA-protein correlation analysis:

    • RNA-protein correlations vary across genes and tissues, limiting the utility of RNA-based assays to predict protein expression

    • Perform parallel qRT-PCR and Western blot analyses to establish correlation patterns specific to your experimental system

  • Post-translational modification considerations:

    • PPP1R13L is subject to post-translational modifications and proteolytic processing (e.g., Caspase-3 cleavage creating a stable 295-828aa fragment)

    • Use antibodies targeting different regions of the protein to detect full-length vs. processed forms

  • Protein stability assessment:

    • PPP1R13L can affect p53 stability through enhanced proteasomal degradation

    • Incorporate proteasome inhibitors (e.g., MG132) in experiments to determine if protein stability, rather than transcription, explains the discrepancy

  • Subcellular localization analysis:

    • The protein may relocalize between cellular compartments while total levels remain constant

    • Perform fractionation experiments or immunofluorescence with PPP1R13L antibodies to detect compartment-specific changes

  • Paired RNA-protein assays:

    • When possible, derive information from paired RNA and proteomic assays from the same samples

    • Single-cell approaches combining RNA and protein detection may provide insights into cell-specific discrepancies

Research has shown that PPP1R13L can influence protein levels of targets like p53 through enhanced degradation rather than transcriptional changes , highlighting the importance of integrated RNA-protein analyses.

How can PPP1R13L antibodies be used to study its role in cardiac pathologies?

PPP1R13L antibodies are valuable tools for investigating the protein's role in cardiac disorders:

  • Genetic cardiomyopathy studies: In Arab Christian infants diagnosed with dilated cardiomyopathy (DCM) associated with skin, teeth, and hair abnormalities, homozygous sequence variations creating premature stop codons in PPP1R13L were identified. Antibody-based methods confirmed the complete absence of iASPP protein in patient-derived fibroblasts .

  • Inflammatory response analysis: PPP1R13L deficiency leads to hypersensitivity to lipopolysaccharide in an NF-κB-dependent manner. Antibodies can be used to study:

    • NF-κB binding activity to pro-inflammatory cytokine gene promoters

    • Correlation between PPP1R13L levels and cardiac inflammation

  • Animal model validation: PPP1R13L antibodies can be employed in immunohistochemistry and Western blot analyses of heart tissues from Ppp1r13l-deficient mice to track DCM development stages .

  • Therapeutic development: By establishing the crucial role of iASPP in dampening cardiac inflammatory responses, antibody-based screening could identify compounds that modulate PPP1R13L function as potential therapeutic targets for cardiac conditions .

What methodologies can be employed to study PPP1R13L's role in cancer progression and metastasis?

To investigate PPP1R13L's contribution to cancer progression, researchers can employ multiple antibody-based methodologies:

  • In vivo tumor models: PPP1R13L overexpression strongly accelerates tumor formation by RAS/E1A transformed cells and produces a phenotype with multiple tumor nodes, consistent with increased metastasis. Use PPP1R13L antibodies to:

    • Track protein expression in primary vs. metastatic lesions

    • Monitor changes in PPP1R13L subcellular localization during metastatic progression

  • Pathway analysis: PPP1R13L promotes cervical cancer progression through the PTEN/AKT/mTOR pathway. Antibody-based methods can assess:

    • Correlation between PPP1R13L levels and PTEN expression

    • Activation status of downstream AKT/mTOR signaling components

  • Clinical correlation studies: In acute myeloid leukemia, high iASPP expression predicts poor outcomes independently of established risk classifications. Antibody-based tissue microarray analysis can help:

    • Stratify patients based on PPP1R13L expression levels

    • Identify correlations with treatment response and survival

  • Mechanistic dissection: Using specific domain antibodies (e.g., against Ank-SH3 domain) can help elucidate how PPP1R13L interacts with p53, p63, and other partners to promote cancer progression .

  • Therapeutic response prediction: PPP1R13L antibody staining intensity in pre-treatment biopsies might serve as a biomarker for predicting response to specific therapies, particularly those targeting apoptotic pathways or p53 family function .

What emerging technologies might enhance the utility of PPP1R13L antibodies in research?

Several cutting-edge technologies promise to expand the applications of PPP1R13L antibodies:

  • Single-cell antibody screening approaches: New methods for simultaneously validating RNA-based predictions of multiple markers can be applied to PPP1R13L research. These approaches combine unique cell surface barcoding with experimental markers of interest and pooled analysis by methods like CyTOF .

  • Proximity-dependent biotinylation (BioID/TurboID): Fusion of PPP1R13L with biotin ligases, followed by detection with specific antibodies, can map the proximal interactome of PPP1R13L in different cellular compartments and under various conditions.

  • Antibody-based PROTAC development: Utilizing the specificity of PPP1R13L antibodies to design proteolysis-targeting chimeras (PROTACs) could enable selective degradation of PPP1R13L in cancer cells, potentially reversing oncogenic phenotypes identified in cervical cancer and AML studies .

  • Multiplexed imaging technologies: Methods like CO-Detection by indEXing (CODEX) or Multiplexed Ion Beam Imaging (MIBI) would allow simultaneous detection of PPP1R13L alongside dozens of other proteins in tissue sections, providing spatial context to PPP1R13L interactions.

  • Nanobody development: Generating single-domain antibodies (nanobodies) against PPP1R13L could enhance intracellular tracking and potentially interfere with specific protein-protein interactions involving distinct domains like the Ank-SH3 region .

What considerations should researchers keep in mind when designing antibody panels that include PPP1R13L?

When incorporating PPP1R13L antibodies into multiplex panels, researchers should consider:

  • Epitope compatibility: Ensure that antibodies in the panel recognize distinct epitopes and won't sterically hinder each other's binding, particularly important when studying PPP1R13L interactions with p53 family members .

  • Expression correlation validation: The utility of RNA-based assays to derive antibody panels is limited by RNA-protein correlations that vary across genes and tissues. Future approaches should incorporate explicit information on these correlations derived from paired RNA and proteomic assays .

  • Signal separation considerations:

    • For flow cytometry or mass cytometry panels, select fluorophores or metal isotopes with minimal spillover

    • For immunofluorescence multiplexing, consider the subcellular localization pattern of PPP1R13L (nuclear vs. cytoplasmic) when pairing with other markers

  • Functional marker integration: Include markers of pathways known to be regulated by PPP1R13L, such as:

    • p53 pathway members

    • NF-κB signaling components

    • PTEN/AKT/mTOR pathway elements

    • EMT markers

  • Tissue context optimization: PPP1R13L is highly expressed in tissues such as the skin, esophagus, myocardium, vagina, and cervix. Panel design should account for tissue-specific expression patterns and potential background issues .

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.