Phospho-CTNNB1 (S33/S37) Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Overview

The Phospho-CTNNB1 (S33/S37) Recombinant Monoclonal Antibody is a highly specific reagent designed to target phosphorylated β-catenin at serine residues 33 and 37. This antibody is engineered using recombinant DNA technology, ensuring high consistency and reproducibility. Its primary function is to detect and analyze the phosphorylation state of β-catenin, a critical regulator in the canonical Wnt signaling pathway .

Antibody Design

  • Clonality: Monoclonal, derived from recombinant DNA (e.g., Clone 4C11) .

  • Immunogen: A synthetic phosphorylated peptide corresponding to human β-catenin residues S33/S37 .

  • Host Species: Engineered in mammalian cells (e.g., CHO or HEK293 systems) .

  • Purification: Affinity chromatography for high specificity .

Mechanistic Role of Phosphorylated β-Catenin

Phosphorylation at S33/S37 is a hallmark of β-catenin degradation in the absence of Wnt signaling. In the canonical Wnt pathway:

  1. Wnt-off State: β-Catenin is phosphorylated by a destruction complex (APC, GSK3β, AXIN), leading to ubiquitination and proteasomal degradation .

  2. Wnt-on State: Phosphorylation is inhibited, stabilizing β-catenin, which translocates to the nucleus to activate TCF/LEF transcription factors .

The antibody enables precise detection of this phosphorylation-dependent regulation, critical for studying oncogenesis (e.g., colorectal, ovarian cancers) and developmental processes .

Immunofluorescence (IF)

  • Example: Detection of cytoplasmic β-catenin in SW480 cells treated with proteasome inhibitor MG-132 (25 µM, 4 hours). Signal loss upon peptide competition confirms specificity .

  • Protocol:

    • Fixation: 4% formaldehyde.

    • Permeabilization: 0.2% Triton X-100.

    • Blocking: 10% normal goat serum.

    • Antibody incubation: Overnight at 4°C .

ELISA

  • Use Case: Quantification of phosphorylated β-catenin in lysates or purified proteins.

  • Sensitivity: Detection thresholds depend on sample preparation and epitope accessibility .

Cancer Pathogenesis

  • CTNNB1 Mutations: Frequent in colorectal, ovarian, and hepatocellular cancers, leading to constitutive Wnt activation .

  • Nuclear Accumulation: A surrogate marker for β-catenin activation, detectable via IF .

Wnt Signaling Dynamics

  • Regulation: AKT-mediated phosphorylation at S552 (distinct from S33/S37) enhances nuclear localization, as shown in 293T cells .

  • Cross-talk: Interactions with pathways like PML-NB disruption and insulin internalization .

Comparative Analysis of Antibodies

FeaturePolyclonal (e.g., CABP0524) Recombinant Monoclonal (CSB-RA006169A33phHU)
SpecificityBroad (human, mouse, rat)High (human-specific)
ProductionRabbit serumRecombinant mammalian cells
ApplicationsWestern blot, IHC-PIF, ELISA
Dilution1:50–1:200 (IHC-P)1:20–1:200 (IF)

Product Specs

Buffer
Rabbit IgG in phosphate buffered saline, pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Description

The vectors expressing anti-CTNNB1 antibody were constructed as follows: an animal was immunized with a synthesized peptide derived from human Phospho-CTNNB1 (S33/S37). Positive splenocytes were isolated and RNA was extracted. DNA was obtained by reverse transcription and the CTNNB1 antibody gene was sequenced and screened. The heavy and light chain sequences were amplified by PCR and cloned into plasma vectors. The vector clones were then transfected into mammalian cells for production. The final product is the recombinant CTNNB1 antibody. Recombinant CTNNB1 antibody in the culture medium was purified using affinity-chromatography. It can react with CTNNB1 protein from Human and is used in the ELISA and IF assays.

CTNNB1 (Catenin Beta 1) is a protein-coding gene. Diseases associated with CTNNB1 include Pilomatrixoma and Neurodevelopmental Disorder with Spastic Diplegia and Visual Defects. Associated pathways include the NF-kappaB pathway and apoptotic cleavage of cellular proteins. According to some studies, CTNNB1 may have the following characteristics:

  • CTNNB1 mutations are highly prevalent in sporadic castrated tumors.
  • Mutations in the β-catenin gene (CTNNB1) have recently been implicated in the pathogenesis of some colon cancers and melanomas.
  • CTNNB1 signaling plays a key role in the development of a considerable proportion of prostate cancer.
  • The nuclear expression of β-catenin is an accurate immunohistochemical surrogate for CTNNB1 exon 3 mutations and thus may be considered in risk stratification for endometrial cancer.
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery times.
Synonyms
b-catenin antibody; Beta catenin antibody; Beta-catenin antibody; Cadherin associated protein antibody; Catenin (cadherin associated protein); beta 1; 88kDa antibody; Catenin beta 1 antibody; Catenin beta-1 antibody; CATNB antibody; CHBCAT antibody; CTNB1_HUMAN antibody; CTNNB antibody; CTNNB1 antibody; DKFZp686D02253 antibody; FLJ25606 antibody; FLJ37923 antibody; OTTHUMP00000162082 antibody; OTTHUMP00000165222 antibody; OTTHUMP00000165223 antibody; OTTHUMP00000209288 antibody; OTTHUMP00000209289 antibody
Target Names
Uniprot No.

Target Background

Function

CTNNB1 is a key downstream component of the canonical Wnt signaling pathway. In the absence of Wnt, it forms a complex with AXIN1, AXIN2, APC, CSNK1A1 and GSK3B, which promotes phosphorylation on N-terminal Ser and Thr residues and ubiquitination of CTNNB1 via BTRC, leading to its subsequent degradation by the proteasome. In the presence of Wnt ligand, CTNNB1 is not ubiquitinated and accumulates in the nucleus, where it acts as a coactivator for transcription factors of the TCF/LEF family, activating Wnt responsive genes. CTNNB1 is involved in the regulation of cell adhesion, as a component of an E-cadherin:catenin adhesion complex. It acts as a negative regulator of centrosome cohesion and is involved in the CDK2/PTPN6/CTNNB1/CEACAM1 pathway of insulin internalization. CTNNB1 blocks anoikis of malignant kidney and intestinal epithelial cells and promotes their anchorage-independent growth by down-regulating DAPK2. It disrupts PML function and PML-NB formation by inhibiting RANBP2-mediated sumoylation of PML. CTNNB1 promotes neurogenesis by maintaining sympathetic neuroblasts within the cell cycle. It is involved in chondrocyte differentiation via interaction with SOX9. SOX9-binding competes with the binding sites of TCF/LEF within CTNNB1, thereby inhibiting the Wnt signaling.

Gene References Into Functions
  1. CXC chemokine ligand 9 promotes the progression of diffuse large B-cell lymphoma in a beta-catenin-dependent manner. PMID: 30130730
  2. Results suggest that epigenetic regulation of CTNNB1 may serve as a novel avenue to block colon cancer cell migration and invasion. PMID: 29923144
  3. The results demonstrated that 2HF could inhibit EMT, and cell migration and invasion through the Wnt/bcatenin signaling pathway by suppressing GSK3b phosphorylation, betacatenin expression and transactivation. PMID: 30226607
  4. Collectively, these studies suggested the cellular transcription factor beta-catenin stimulates productive herpes simplex virus 1infection, in part because VP16 enhances beta-catenin dependent transcription. PMID: 30077727
  5. CTNNB1 mutations may be more related to tumorigenesis ( aldosterone-producing adenoma) rather than excessive aldosterone production PMID: 28102204
  6. CTNNB1 mutations were found in 60% of Basal cell adenoma but not in basal cell adenocarcinoma. None of the tested cases had PIK3CA mutations. CTNNB1 mutation trended to be more common in those cases having a predominant tubular or tubulotrabecular patterns. PMID: 29224720
  7. Data reveal that post-translational modifications of beta-catenin in the ubiquitin-proteasome pathway yield a truncated beta-catenin molecule containing a serine 552-phosphorylated core region without N and C termini. This proteolytic processing of beta-catenin is required for binding with TCF4 and subsequent transcriptional activation. PMID: 29330435
  8. Results identify CTNNB1 as a Girdin-interacting protein. Girdin-depleted skin cancer cells displayed scattering and impaired E-cadherin-specific cell-cell adhesion. PMID: 30194792
  9. the dysregulation of TET2/E-cadherin/beta-catenin regulatory loop is a critical oncogenic event in HCC progression PMID: 29331390
  10. High CTNNB1 expression is associated with bladder cancer progression. PMID: 30015971
  11. It has been found that miR-27a-3p modulated the Wnt/beta-catenin signaling pathway to promote epithelial-mesenchymal transition in oral squamous carcinoma stem cells by down-regulating SFRP1. PMID: 28425477
  12. Beta-catenin pathway is activated by CBX8 in in hepatocellular carcinoma. PMID: 29066512
  13. our data provide a novel evidence for the biological and clinical significance of SPAG5 as a potential biomarker, and we demonstrate that SPAG5-b-catenin-SCARA5 might be a novel pathway involved in hepatocellular carcinoma progression. PMID: 30249289
  14. Results show that hypoxia enhanced nuclear accumulation and transcriptional activity of beta-catenin which promotes expression of EMT-related genes and eventually contributes to the metastatic process in lung cancer cells. PMID: 30396950
  15. This study demonstrates that FOXC1 induces cancer stem cells (CSCs)-like properties in non-small cell lung cancer (NSCLC) by promoting beta-catenin expression. The findings indicate that FOXC1 is a potential molecular target for anti-CSC-based therapies in NSCLC PMID: 30189871
  16. High TBL1XR1 expression indicates poor disease-free survival of stage I-III colorectal cancer patients; beta-catenin signaling is critical for TBL1XR1-mediated colorectal cancer cells oncogenicity. PMID: 28295012
  17. Taking together, these results suggest that Wnt/beta-catenin signal pathway activation-dependent up-regulation of syncytin-1 contributes to the pro-inflammatory factor TNF-alpha-enhanced fusion between oral squamous cell carcinoma cells and endothelial cells. PMID: 28112190
  18. The disassociation of the beta-catenin/E-cadherin complex in the osteoblast membrane under stretch loading and the subsequent translocation of beta-catenin into the nucleus may be an intrinsic mechanical signal transduction mechanism. PMID: 29901167
  19. Aberrant CTNNB1 expression was seen in a substantial proportion of our hepatocellular carcinoma (HCC) cases. CTNNB1-positive HCC was associated with normal AFP levels, unicentric tumors, well-differentiated histology, and an unfavorable outcome. PMID: 30082549
  20. Long noncoding RNA AFAP1-AS1 enhances cell proliferation and invasion in osteosarcoma through regulating miR-4695-5p/TCF4-beta-catenin signaling. PMID: 29901121
  21. High CTNNB1 expression is associated with the recurrece of Adamantinomatous Craniopharyngiomas. PMID: 29625497
  22. High CTNNB1 expression is associated with uterine fibroids. PMID: 29066531
  23. The nucleus and/or cytoplasm expression of beta-catenin was associated with tumor progression and correlated overall survival of patients with ovarian cancer (OC). beta-catenin may be a possible potential prognostic biomarker for the patients with OC. [review] PMID: 30103006
  24. In the two wild type (WT) cases, two novel alterations were detected: a complex deletion of APC and a pathogenic mutation of LAMTOR2. Focusing on WT DT subtype, deep sequencing of CTNNB1, APC and LAMTOR2 was conducted on a retrospective series of 11 WT DT using a targeted approach PMID: 29901254
  25. DLX1 interacted with beta-catenin and enhanced the interaction between beta-catenin and TCF4 T-cell factor PMID: 29317218
  26. Nuclear beta-catenin immunoreactivity with appropriate criteria may be helpful to distinguish basal cell adenocarcinoma (BCAC) from histologically similar tumors. However, a minor subset of adenoid cystic carcinoma (ACC) with nuclear beta-catenin expression require careful diagnosis. PMID: 29496310
  27. High CTNNB1 expression is associated with metastasis in cholangiocarcinoma. PMID: 30193944
  28. beta;-catenin directly interacts with the Cx43 carboxyl-terminal domain. PMID: 29882937
  29. This study showed that beta-catenin expression was the most evident in the nucleus rather than in cytoplasm. PMID: 29297710
  30. Nuclear beta-catenin accumulation in non-mitotic glioblastoma cells is due to a feed forward mechanism between DOCK4 and beta-catenin. PMID: 28925399
  31. Study found that HIF1alpha overexpression led to an enhanced betacatenin nuclear translocation, while betacatenin silencing inhibited betacatenin nuclear translocation. The enhanced betacatenin nuclear translocation induced resulted in an enhanced cell proliferation and cell invasion, an altered cell cycle distribution, decreased apoptosis, and improved nonhomologous end joining repair under normal and irradiation cond... PMID: 29658569
  32. our results demonstrated that miR-188 inhibits glioma cell proliferation by targeting beta-catenin PMID: 29268818
  33. Marked upregulation of beta-catenin and its downstream targets effectively enhanced hepatosphere formation, with an associated induction of CD133, OCT4 and Sox2 expression and also caused an significant enhancement of HCC proliferation PMID: 29792038
  34. Wnt/beta-catenin signaling pathway may play a significant role in the pathogenesis of preeclampsia by regulating the invasion and proliferation of trophoblast. PMID: 29603045
  35. Associations between environmental variants together with single nucleotide polymorphisms (SNPs) of beta-catenin (ctnnb1) and lung cancer risk were analyzed using a logistic regression model. PMID: 29562493
  36. that CTNNB1 is overexpressed and confers a poor prognosis in acute myeloid leukemia PMID: 29496308
  37. High CTNNB1 expression is associated with cisplatin-resistance in non-small cell lung cancer. PMID: 30009824
  38. beta-catenin immunopositivity is seen in majority of cases of sinonasal sarcoma PMID: 29566950
  39. For the first time, we demonstrated that rather than excluding lymphocytes infiltration as reported in mela-noma, high levels of TILs were associated with beta-catenin overexpression in BC. PMID: 29286921
  40. Study shows that apigenin-induced lysosomal degradation of beta-catenin in Wnt/beta-catenin signaling. PMID: 28337019
  41. Used CRISPR-Cas9 technology to study effect of knockout of catenin beta 1 (CTNNB1) on cell behavior and signal pathways in HEK293 cells. Results showed knockout of CTNNB1 effected Wnt/beta-catenin signaling pathway and suppressed adhesion and proliferation of HEK 293T cells. PMID: 29249062
  42. our results also revealed that lncRNA SNHG20 knockdown inhibited Wnt/b catenin signaling activity by suppressing beta-catenin expression and reversing the downstream target gene expression. Taken together, lncRNA SNHG20 plays an pivotal role in ovarian cancer progression by regulating Wnt/b-catenin signaling PMID: 29101241
  43. Wnt3A regulates the expression of 1,136 genes, of which 662 are upregulated and 474 are downregulated in CCD-18Co cells. A set of genes encoding inhibitors of the Wnt/beta-catenin pathway stand out among those induced by Wnt3A, which suggests that there is a feedback inhibitory mechanism. PMID: 29044515
  44. The aim of our study was to analyze the immunohistochemical expression of beta-catenin, E-cadherin and Snail, depending on clinico-morphological aspects of the laryngeal squamous cell carcinomas. Results revealed variable E-cadherin, beta-catenin and Snail expression, depending on differentiation degree and tumor stage. PMID: 29250652
  45. In this study we showed that the activation of Wnt/beta-catenin pathway culminates in the upregulation of MGAT1 enzyme both at transcriptional and post-transcriptional levels. We also showed that overexpression of the beta-catenin gene (CTNNB1) increased the promoter activity of MGAT1. PMID: 29310626
  46. CTNNB1 mutation is associated with acquired resistance to KIT inhibitor in metastatic melanoma. PMID: 28421416
  47. three CTNNB1 SNPs were suggested to have the potential to be novel biomarkers for risk prediction of cancer in overall population or some specific subgroups. [Review] PMID: 28963373
  48. A CTNNB1 exon 3 mutation restricted to the areas exhibiting both positive glutamine synthetase (GS) and C-reactive protein (CRP) expression, whereas wild-type CTNNB1 was found in areas showing only CRP staining. These two cases illustrate focal beta-catenin activation that can occur within Inflammatory hepatocellular adenoma (IHCAs). PMID: 28618047
  49. Results show that E-cadherin/beta-catenin complex is disrupted by ICAT promoting epithelial-mesenchymal transition of cervical cancer cells. PMID: 29048651
  50. Toosendanin administration inhibited growth and liver metastasis of orthotopically implanted SGC7901 tumors in vivo through miR200amediated beta-catenin pathway. Our data suggest that Toosendanin may suppress oncogenic phenotypes of human GC cells partly via miR200a/beta-catenin axis. Hence, Toosendanin may have a promising chemotherapeutic activity for GC therapy. PMID: 29048657

Show More

Hide All

Database Links

HGNC: 2514

OMIM: 114500

KEGG: hsa:1499

STRING: 9606.ENSP00000344456

UniGene: Hs.476018

Involvement In Disease
Colorectal cancer (CRC); Pilomatrixoma (PTR); Medulloblastoma (MDB); Ovarian cancer (OC); Mesothelioma, malignant (MESOM); Mental retardation, autosomal dominant 19 (MRD19); Vitreoretinopathy, exudative 7 (EVR7)
Protein Families
Beta-catenin family
Subcellular Location
Cytoplasm. Nucleus. Cytoplasm, cytoskeleton. Cell junction, adherens junction. Cell junction. Cell membrane. Cytoplasm, cytoskeleton, microtubule organizing center, centrosome. Cytoplasm, cytoskeleton, spindle pole. Cell junction, synapse. Cytoplasm, cytoskeleton, cilium basal body.
Tissue Specificity
Expressed in several hair follicle cell types: basal and peripheral matrix cells, and cells of the outer and inner root sheaths. Expressed in colon. Present in cortical neurons (at protein level). Expressed in breast cancer tissues (at protein level).

Q&A

What is the biological significance of CTNNB1 phosphorylation at S33/S37 sites?

CTNNB1 (beta-catenin) phosphorylation at S33/S37 residues represents a critical regulatory mechanism within the canonical Wnt signaling pathway. In the absence of Wnt ligands, CTNNB1 forms a complex with AXIN1, AXIN2, APC, CSNK1A1 (casein kinase 1α), and GSK3B (glycogen synthase kinase 3β) . Within this destruction complex, CSNK1A1 first phosphorylates CTNNB1 at serine 45 (S45), which serves as a priming event for subsequent sequential phosphorylation at T41, S37, and S33 by GSK3A/GSK3B . The phosphorylation at S33 and S37 specifically creates a recognition site for the SCF-β-TrCP ubiquitin ligase complex, which targets CTNNB1 for proteasomal degradation . This phosphorylation-dependent degradation mechanism prevents nuclear accumulation of CTNNB1 and thereby inhibits transcription of Wnt target genes. Consequently, mutations affecting these phosphorylation sites or dysregulation of this process are implicated in various cancers, including colon cancer, melanoma, and prostate cancer .

How do Phospho-CTNNB1 (S33/S37) antibodies differ from other beta-catenin antibodies?

Phospho-CTNNB1 (S33/S37) antibodies are specifically designed to recognize beta-catenin only when phosphorylated at serine residues 33 and 37, making them fundamentally different from pan-CTNNB1 antibodies that recognize total beta-catenin regardless of phosphorylation status . This specificity allows researchers to distinguish between the phosphorylated form destined for degradation and the non-phosphorylated, transcriptionally active form. Some antibodies may target only S33 or S37 individually, while others (like those described in the search results) recognize both phosphorylated residues simultaneously . Additionally, there are antibodies that recognize the extended phosphodegron including S33/S37/T41, which provides information about the complete GSK3-mediated phosphorylation pattern . The production method also differs between antibodies - monoclonal antibodies like the BC-22 clone offer high specificity and reproducibility, while polyclonal antibodies may provide broader epitope recognition but potentially with more batch-to-batch variation .

What are the primary applications of Phospho-CTNNB1 (S33/S37) antibodies in research?

Phospho-CTNNB1 (S33/S37) antibodies serve multiple critical applications in research investigating Wnt signaling dynamics. The primary applications include:

  • Western blotting (WB): For quantifying phosphorylated CTNNB1 levels in cell or tissue lysates, especially when studying the effects of Wnt pathway activators or inhibitors .

  • Immunofluorescence (IF): For visualizing the subcellular localization of phosphorylated CTNNB1, with recommended dilutions typically ranging from 1:20 to 1:200 .

  • ELISA: For sensitive quantification of phosphorylated CTNNB1 in complex biological samples .

  • Analyzing Wnt pathway activation: For distinguishing between active and inactive states of the pathway by measuring the ratio of phosphorylated to non-phosphorylated CTNNB1 .

  • Studying disease mechanisms: For investigating aberrant Wnt signaling in various pathologies, including neurodevelopmental disorders (with spastic diplegia and visual defects) and cancers (particularly colon, prostate, and endometrial cancers) .

These antibodies are particularly valuable for monitoring CTNNB1 degradation dynamics in response to various experimental manipulations, including genetic knockouts of pathway components or pharmacological interventions .

How are recombinant monoclonal antibodies against Phospho-CTNNB1 (S33/S37) produced?

The production of recombinant monoclonal antibodies against Phospho-CTNNB1 (S33/S37) involves a sophisticated multi-step process combining immunological techniques with molecular biology methods. Initially, an animal is immunized with a synthesized peptide derived specifically from human Phospho-CTNNB1 (S33/S37) . Following immune response development, B cells (splenocytes) producing the desired antibody are isolated and their RNA extracted . This RNA undergoes reverse transcription to obtain cDNA encoding the antibody heavy and light chains. The antibody gene sequences are then identified through screening and sequencing procedures .

The selected heavy and light chain sequences are amplified via PCR and cloned into appropriate plasma expression vectors. These vector constructs are subsequently transfected into mammalian cells for recombinant protein production . The secreted antibody in the culture medium undergoes purification through affinity chromatography to obtain the final product - a highly specific recombinant monoclonal antibody against Phospho-CTNNB1 (S33/S37) . This recombinant production approach offers advantages over traditional hybridoma methods, including better reproducibility, elimination of animal maintenance for antibody production, and the potential for engineering modifications to enhance specificity or functionality.

How can Phospho-CTNNB1 (S33/S37) antibodies be used to study the kinetics of Wnt pathway activation?

The study of Wnt pathway activation kinetics using Phospho-CTNNB1 (S33/S37) antibodies requires a sophisticated experimental approach that capitalizes on the temporal dynamics of beta-catenin phosphorylation and degradation. To effectively investigate these kinetics, researchers should implement a time-course analysis following Wnt ligand stimulation or pathway inhibitor treatment, with samples collected at precisely timed intervals (typically 0, 15, 30, 60, 120, and 240 minutes) . This approach allows for the visualization of the rapid changes in phosphorylation status.

An alternative approach involves quantifying non-phospho-CTNNB1 (the species NOT phosphorylated at S33/S37/T41) as an inverse readout of phosphorylation status . This method has demonstrated higher sensitivity to condition-dependent changes compared to measuring total CTNNB1 alone. For rigorous mechanistic studies, combining these antibody-based approaches with small molecule inhibitors (such as CHIR-99021 for GSK3A/GSK3B inhibition) enables precise dissection of the phosphorylation cascade and its regulation .

What are the technical challenges in detecting phosphorylated CTNNB1 and how can they be overcome?

Detecting phosphorylated CTNNB1 presents several significant technical challenges that require careful methodological consideration. The primary challenges include:

  • Rapid degradation: Phosphorylated CTNNB1 (at S33/S37) is quickly targeted for proteasomal degradation, resulting in extremely low steady-state levels that may fall below detection thresholds . To address this challenge, researchers can:

    • Employ proteasome inhibitors (e.g., MG132 or bortezomib) prior to cell lysis, though this approach requires careful control experiments as it may affect other pathway components .

    • Use highly sensitive detection methods such as enhanced chemiluminescence or fluorescently-tagged secondary antibodies.

    • Focus on measuring non-phosphorylated CTNNB1 as an inverse indicator of phosphorylation status .

  • Phosphatase activity: Endogenous phosphatases in cell lysates can dephosphorylate CTNNB1 during sample preparation, leading to false-negative results. This can be mitigated by:

    • Including robust phosphatase inhibitor cocktails in all lysis buffers.

    • Maintaining samples at cold temperatures throughout processing.

    • Using denaturing lysis conditions when compatible with downstream applications.

  • Cross-reactivity concerns: Some phospho-specific antibodies may recognize similar phosphorylation motifs on other proteins. Researchers should:

    • Validate antibody specificity using appropriate controls (phosphatase-treated samples, CTNNB1 knockdown cells).

    • Consider using multiple antibodies targeting different epitopes for confirmation.

    • Implement proper blocking procedures to minimize non-specific binding.

  • Subcellular localization complexity: Phosphorylated CTNNB1 may be distributed across different cellular compartments with varying extraction efficiencies. This challenge can be addressed by:

    • Performing subcellular fractionation before analysis.

    • Using immunofluorescence techniques to visualize spatial distribution.

    • Employing specialized extraction buffers optimized for different cellular compartments.

How do mutations in the phosphodegron region affect antibody recognition and Wnt signaling outcomes?

Mutations in the phosphodegron region of CTNNB1, particularly those affecting residues S33, S37, T41, and S45, have profound implications for both antibody recognition and Wnt signaling outcomes. These mutations frequently occur in various cancers and can significantly alter pathway regulation . From an antibody recognition perspective, point mutations at S33 or S37 (the epitopes recognized by Phospho-CTNNB1 (S33/S37) antibodies) directly prevent phosphorylation at these sites and consequently abolish antibody binding . This creates a technically challenging scenario where the absence of signal could indicate either pathway activation (normal biology) or the presence of mutations (pathology).

The functional consequences of these mutations are equally significant. When S33 or S37 residues are mutated, GSK3B cannot phosphorylate these sites even when the priming phosphorylation at S45 by CSNK1A1 occurs normally . This prevents recognition by the SCF-β-TrCP ubiquitin ligase complex, resulting in stabilization of CTNNB1 and constitutive pathway activation independent of Wnt ligand stimulation . Importantly, different mutations within the phosphodegron can have varying effects on signal intensity. For example, mutations at S33 may allow partial phosphorylation at S37, resulting in reduced but not eliminated degradation.

To effectively study systems with potential phosphodegron mutations, researchers should implement a comprehensive analysis approach that includes:

  • Genotyping the CTNNB1 phosphodegron region by sequencing

  • Using antibodies targeting multiple phosphorylation sites separately

  • Comparing phospho-specific antibody signals with total CTNNB1 levels

  • Employing functional readouts of Wnt pathway activation (e.g., TCF/LEF reporters)

  • Utilizing non-phospho-CTNNB1 antibodies to detect stabilized forms resulting from mutations

What is the relationship between HUWE1 and CTNNB1 phosphorylation, and how can it be investigated using phospho-specific antibodies?

The relationship between HUWE1 (HECT, UBA and WWE domain containing E3 ubiquitin protein ligase 1) and CTNNB1 phosphorylation represents a sophisticated regulatory mechanism in Wnt signaling that can be effectively investigated using phospho-specific antibodies. Research indicates that HUWE1 enhances WNT signaling by influencing the phosphorylation status of CTNNB1 at the critical S33/S37/T41 residues .

In CSNK1A1 knockout cells, where the priming phosphorylation at S45 is absent, HUWE1 appears to inhibit the GSK3A/GSK3B-mediated phosphorylation of the S33/S37/T41 residues . Experimental evidence shows that HUWE1 loss in CSNK1A1 knockout cells reduces non-phospho-CTNNB1 abundance by approximately 37%, which correlates closely with a similar reduction in soluble CTNNB1 . This suggests that HUWE1 normally prevents excessive phosphorylation of CTNNB1 at these residues.

To investigate this relationship using phospho-specific antibodies, researchers can implement several methodological approaches:

  • Comparative analysis using both phospho-specific (S33/S37) and non-phospho-CTNNB1 antibodies in wildtype, HUWE1 knockout, CSNK1A1 knockout, and double knockout cellular models .

  • Complementation experiments with GSK3A/GSK3B inhibitors (such as CHIR-99021) to verify the mechanism. Treatment of CSNK1A1 knockout; HUWE1 knockout cells with CHIR-99021 increased non-phospho-CTNNB1 abundance by 3.2-fold, suggesting GSK3A/GSK3B indeed mediates the increased phosphorylation observed after HUWE1 loss .

  • Functional validation using WNT reporter assays to correlate changes in phosphorylation status with transcriptional outcomes. GSK3A/GSK3B inhibition in CSNK1A1 knockout; HUWE1 knockout cells increased WNT reporter activity by 10.9-fold, restoring signaling to levels comparable to CSNK1A1 knockout cells with intact HUWE1 .

  • Time-course analysis following proteasome inhibition to capture the dynamic relationship between HUWE1 activity and phosphorylation-dependent CTNNB1 degradation.

This experimental framework utilizing phospho-specific antibodies enables precise dissection of how HUWE1 interfaces with the canonical CTNNB1 phosphorylation cascade to modulate Wnt signaling.

What are the optimal sample preparation methods for detecting phosphorylated CTNNB1?

Detecting phosphorylated CTNNB1 requires meticulous sample preparation to preserve phosphorylation status and ensure reliable results. The following methodological workflow is recommended:

  • Cell harvest and lysis considerations:

    • Perform rapid harvesting to minimize post-harvest signaling changes

    • Use ice-cold phosphate-buffered saline for all washing steps

    • Add phosphatase inhibitor cocktails (containing sodium fluoride, sodium orthovanadate, sodium pyrophosphate, and β-glycerophosphate) to all buffers

    • Include protease inhibitors to prevent degradation of CTNNB1

    • Consider flash-freezing samples in liquid nitrogen if immediate processing is not possible

  • Lysis buffer composition:

    • RIPA buffer (50 mM Tris-HCl pH 7.4, 150 mM NaCl, 1% NP-40, 0.5% sodium deoxycholate, 0.1% SDS) with phosphatase and protease inhibitors works well for most applications

    • For challenging samples, consider using specialized buffers containing 8M urea or 6M guanidine hydrochloride to ensure complete protein denaturation and phosphoepitope exposure

  • Sample processing:

    • Maintain constant cold temperature (4°C) during all processing steps

    • Limit freeze-thaw cycles to prevent phosphoepitope degradation

    • Sonicate samples briefly to shear DNA and ensure complete lysis

    • Centrifuge at high speed (≥14,000 × g) to remove insoluble material

    • Determine protein concentration using methods compatible with phosphatase inhibitors (Bradford or BCA assay)

  • Special considerations for specific applications:

    • For Western blotting: Include phosphorylated protein standards as positive controls

    • For immunoprecipitation: Use antibody-conjugated beads with gentle agitation overnight at 4°C

    • For immunofluorescence: Fix samples rapidly with paraformaldehyde (avoid methanol fixation which can disrupt phosphoepitopes)

  • Alternative approaches for challenging scenarios:

    • If direct detection of phosphorylated CTNNB1 is difficult due to rapid degradation, quantify non-phospho-CTNNB1 as an inverse measure

    • Consider phosphatase treatment of control samples to validate antibody specificity

    • When comparing conditions, standardize harvest timing carefully as phosphorylation states can change rapidly

How can researchers validate the specificity of Phospho-CTNNB1 (S33/S37) antibodies?

Validating the specificity of Phospho-CTNNB1 (S33/S37) antibodies is essential for ensuring experimental rigor and data reliability. Researchers should implement a multi-faceted validation strategy:

  • Phosphatase treatment controls:

    • Divide samples into treated and untreated groups

    • Treat one set with lambda phosphatase to remove phosphate groups

    • The phospho-specific antibody signal should disappear in treated samples while total CTNNB1 signal remains unchanged

    • This confirms that the antibody specifically recognizes the phosphorylated form

  • Genetic validation approaches:

    • Use CTNNB1 knockout or knockdown cells as negative controls

    • Generate point mutations at S33 and S37 (S33A/S37A) to create non-phosphorylatable CTNNB1

    • Compare antibody reactivity between wild-type and mutant proteins

    • In valid antibodies, signal should be absent in the mutant constructs

  • Pharmacological manipulation:

    • Treat cells with GSK3 inhibitors like CHIR-99021 to reduce S33/S37 phosphorylation

    • Treat with Wnt pathway activators to reduce phosphorylation

    • Treat with Wnt pathway inhibitors to increase phosphorylation

    • A specific antibody will show corresponding signal changes aligned with the expected biology

  • Peptide competition assays:

    • Pre-incubate the antibody with excess phosphorylated peptide containing the S33/S37 epitope

    • Pre-incubate with non-phosphorylated peptide as control

    • The phospho-peptide should block specific binding while the non-phospho-peptide should not

    • This confirms epitope-specific recognition

  • Cross-validation with multiple antibodies:

    • Compare results using different phospho-CTNNB1 antibodies from diverse suppliers

    • Use antibodies with different epitope recognition (e.g., monoclonal vs polyclonal)

    • Consistent results across different antibodies increase confidence in specificity

  • Validation across applications:

    • Test antibody performance in multiple techniques (WB, IF, IP, ELISA)

    • Confirm that results are consistent across methodologies

    • Document optimal working conditions for each application

What experimental controls are critical when studying CTNNB1 phosphorylation in the Wnt signaling pathway?

Studying CTNNB1 phosphorylation in the Wnt signaling pathway requires a comprehensive set of experimental controls to ensure data validity and interpretability. Critical controls include:

  • Pathway manipulation controls:

    • Positive control: WNT ligand stimulation (e.g., WNT3A) to reduce CTNNB1 phosphorylation

    • Negative control: WNT pathway inhibitors to increase phosphorylation

    • Dose-response relationships should be established for all pathway modulators

    • Time-course experiments to capture dynamic changes in phosphorylation status

  • Kinase and phosphatase controls:

    • GSK3 inhibition (e.g., CHIR-99021) to block phosphorylation at S33/S37/T41

    • CSNK1A1 inhibition to prevent priming phosphorylation at S45

    • Phosphatase inhibitors to preserve phosphorylation during sample preparation

    • Phosphatase treatment of control samples to validate antibody specificity

  • Genetic controls:

    • CTNNB1 knockout/knockdown cells as negative controls for antibody specificity

    • CSNK1A1 knockout cells to study priming-independent phosphorylation

    • GSK3A/GSK3B knockout or knockdown to confirm kinase specificity

    • Rescue experiments with wild-type vs. phospho-mutant CTNNB1

  • Detection controls:

    • Measurement of total CTNNB1 alongside phosphorylated forms

    • Quantification of non-phospho-CTNNB1 as a complementary readout

    • Parallel assessment of downstream transcriptional targets via reporter assays

    • Subcellular fractionation to distinguish cytoplasmic from nuclear CTNNB1 pools

  • Technical controls:

    • Loading controls (e.g., GAPDH, β-actin) to normalize protein amounts

    • Recombinant phosphorylated and non-phosphorylated CTNNB1 as standards

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

    • Multiple biological and technical replicates to ensure reproducibility

  • Contextual controls:

    • Cell density standardization (Wnt signaling can be affected by cell-cell contacts)

    • Serum starvation before treatment to reduce baseline pathway activity

    • Vehicle controls for all treatments

    • Cell type comparisons to assess context-dependency of observations

How can researchers effectively interpret contradictory results in CTNNB1 phosphorylation studies?

Interpreting contradictory results in CTNNB1 phosphorylation studies requires systematic analysis of potential methodological and biological factors that may influence experimental outcomes. When faced with conflicting data, researchers should employ the following analytical framework:

  • Methodological discrepancy analysis:

    • Antibody factors: Different antibodies may recognize distinct epitopes or have varying specificities. Compare antibody clones, host species, and manufacturing methods .

    • Detection sensitivity variations: Western blotting, immunofluorescence, and ELISA have different detection thresholds. Quantify lower limits of detection for each method used .

    • Sample preparation differences: Variations in lysis buffers, phosphatase inhibitors, and protein extraction protocols can significantly impact phosphoepitope preservation .

    • Timing considerations: CTNNB1 phosphorylation is highly dynamic. Standardize time points for cell harvesting after treatments.

  • Biological context assessment:

    • Cell line differences: Various cell types may have distinct baseline Wnt pathway activity or CTNNB1 regulation mechanisms.

    • Genetic background variations: Mutations in Wnt pathway components may affect CTNNB1 phosphorylation dynamics .

    • Microenvironment factors: Cell density, extracellular matrix, and paracrine signaling can influence Wnt pathway activity.

    • Temporal dynamics: Contradictory results may reflect different points in a complex temporal response rather than true contradictions .

  • Integrative analytical approaches:

    • Multi-antibody strategy: Use multiple phospho-specific antibodies targeting different epitopes or from different suppliers.

    • Complementary readouts: Combine direct phospho-CTNNB1 detection with measurements of non-phospho-CTNNB1 and total CTNNB1 .

    • Functional correlation: Assess whether phosphorylation changes correlate with expected functional outcomes (e.g., transcriptional reporter activity) .

    • Quantitative analysis: Apply statistical methods to determine if apparent contradictions are statistically significant or within normal experimental variation.

  • Resolution strategies for specific contradiction types:

    Contradiction TypeProbable CausesResolution Strategy
    Phospho-signal present despite pathway activationIncomplete pathway activation; Subpopulation effectsSingle-cell analysis; Subcellular fractionation
    No phospho-signal despite pathway inhibitionRapid degradation; Antibody sensitivity issuesProteasome inhibition; Alternative detection methods
    Discrepancy between phospho-status and functional readoutsParallel pathways; Post-translational modificationsPathway component knockdowns; Mass spectrometry analysis
    Inconsistent results between replicatesTechnical variability; Unstable reagentsStandardized protocols; Fresh reagent preparation
  • Validation in multiple experimental systems:

    • Cross-validate findings in different cell lines

    • Confirm in vitro observations in appropriate in vivo models when possible

    • Use both gain-of-function and loss-of-function approaches

How can Phospho-CTNNB1 (S33/S37) antibodies be used in cancer research and diagnostics?

Phospho-CTNNB1 (S33/S37) antibodies offer powerful tools for cancer research and potentially diagnostics, given the critical role of aberrant Wnt signaling in tumorigenesis. In research applications, these antibodies enable:

  • Identification of dysregulated Wnt signaling: By comparing phospho-CTNNB1 (S33/S37) levels between normal and cancer tissues, researchers can identify tumors with altered Wnt pathway activity. Reduced phosphorylation suggests pathway hyperactivation, which occurs in many cancer types including colon, prostate, and endometrial cancers .

  • Mutation screening: CTNNB1 mutations affecting the phosphodegron region (particularly S33/S37) are common in certain cancers. Phospho-specific antibodies can serve as an immunohistochemical surrogate to identify samples likely harboring these mutations before confirmation by sequencing . The absence of phospho-signal in the presence of high total CTNNB1 suggests mutations in the phosphorylation sites.

  • Therapeutic response monitoring: As pharmacological Wnt pathway inhibitors enter clinical trials, phospho-CTNNB1 antibodies provide valuable biomarkers for assessing target engagement and efficacy. Restoration of CTNNB1 phosphorylation following treatment would indicate successful pathway inhibition.

  • Stratification of cancer subtypes: Different patterns of CTNNB1 phosphorylation may correlate with specific cancer subtypes or prognosis. For example, research suggests that nuclear β-catenin expression (which inversely correlates with phosphorylation) may be considered in risk stratification for endometrial cancer .

  • Mechanism-of-action studies: When investigating novel compounds affecting the Wnt pathway, phospho-CTNNB1 antibodies help determine whether the mechanism involves regulation of the destruction complex or operates through alternative means.

For potential diagnostic applications, standardized protocols using phospho-CTNNB1 antibodies could assist in:

  • Distinguishing between Wnt-driven and Wnt-independent tumors to guide treatment selection

  • Identifying patients likely to respond to emerging Wnt pathway inhibitors

  • Monitoring for acquired resistance mechanisms during treatment

  • Complementing genetic testing for CTNNB1 mutations with functional protein assessment

Future development of multiplexed immunoassays combining phospho-CTNNB1 with other Wnt pathway components could enhance diagnostic precision and therapeutic decision-making in oncology.

What emerging techniques might enhance the utility of Phospho-CTNNB1 (S33/S37) antibodies in research?

Several emerging techniques hold promise for significantly enhancing the utility of Phospho-CTNNB1 (S33/S37) antibodies in research by improving sensitivity, specificity, spatial resolution, and throughput:

  • Proximity ligation assays (PLA): This technique can detect phosphorylated CTNNB1 with significantly higher sensitivity than conventional immunoassays by generating an amplifiable DNA signal only when two antibodies bind in close proximity. Implementing PLA with antibodies targeting total CTNNB1 and phospho-S33/S37 would enable detection of low-abundance phosphorylated species and visualization of their precise subcellular localization.

  • Mass cytometry (CyTOF): By combining phospho-CTNNB1 antibodies with metal-isotope labeling, researchers can simultaneously measure multiple phosphorylation sites and pathway components at the single-cell level. This approach would reveal heterogeneity in CTNNB1 phosphorylation within cell populations and correlate these patterns with other signaling events.

  • Super-resolution microscopy: Techniques such as STORM, PALM, or STED microscopy paired with phospho-CTNNB1 antibodies could resolve the spatial organization of destruction complexes and phosphorylated CTNNB1 at nanometer resolution, providing unprecedented insights into the physical context of CTNNB1 phosphorylation events.

  • Intracellular phospho-protein reporters: Development of genetically encoded biosensors that undergo conformational changes upon binding by phospho-CTNNB1 antibody fragments would enable real-time monitoring of phosphorylation dynamics in living cells.

  • Automated high-content imaging platforms: Integration of phospho-CTNNB1 antibodies into high-throughput imaging systems would facilitate large-scale screens for compounds or genetic factors affecting CTNNB1 phosphorylation patterns.

  • Phospho-proteomic integration: Combining targeted phospho-CTNNB1 antibody-based assays with global phospho-proteomic profiling would contextualize CTNNB1 phosphorylation within broader signaling networks.

  • Spatial transcriptomics correlation: Coupling phospho-CTNNB1 immunostaining with spatial transcriptomics techniques would allow researchers to directly link phosphorylation status to Wnt target gene expression patterns at the tissue level.

  • Nanobody-based detection systems: Development of nanobodies (single-domain antibody fragments) against phospho-CTNNB1 epitopes would enable superior tissue penetration, reduced background, and potentially intracellular expression for live-cell imaging.

These advanced methodologies, when optimized for phospho-CTNNB1 detection, promise to provide deeper mechanistic insights into the spatial, temporal, and contextual aspects of CTNNB1 phosphorylation in both normal and pathological settings.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.