CTNNB1 Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Target Profile

CTNNB1 Recombinant Monoclonal Antibody is an engineered immunoglobulin produced via recombinant DNA technology to ensure batch-to-batch consistency and epitope specificity. It recognizes the CTNNB1 protein (UniProt ID: P35222), which regulates cell-cell adhesion and nuclear transcription as part of the Wnt pathway .

ParameterDetails
Target AliasesBeta-catenin, Armadillo, Catenin beta-1
Gene ID1499
Biological RoleCell adhesion, Wnt signaling, transcriptional co-activation

Epitope and Specificity

  • Immunogen: Full-length human CTNNB1 protein .

  • Cross-reactivity: Tested against >19,000 human proteins; no reactivity with CTNNG or ARMC4 homologs .

  • Specificity Metrics:

    • S-score: ≥2.5 (difference between target Z-score and next-highest signal) .

    • Western Blot: Detects ~80–85 kDa bands in HeLa lysates .

Immunocytochemistry (ICC)

  • Staining Pattern: Plasma membrane localization in HeLa cells .

  • Protocol: 1–2 µg/ml for 1 hour at RT, visualized via CF488 (green) and RedDot (nuclear counterstain) .

Immunohistochemistry (IHC)

  • Tissue Specificity:

    • Human tonsil, cervical carcinoma (AE00114) .

    • Breast cancer (AE00113) .

  • Protocol: Epitope retrieval at pH6 (AE00114) or pH8 (AE00113), DAB staining post-HRP polymer .

Western Blot (WB)

  • Sensitivity: Detects 30 µg of HeLa lysate at 0.5–1 µg/ml (AE00114) .

  • Band Integrity: Single ~80 kDa band confirms specificity .

Oncogenic Studies

  • Hepatocellular Carcinoma (HCC): CTNNB1 gain-of-function mutations upregulate MMP9, driving immune evasion. The antibody aids in identifying CTNNB1-driven tumor microenvironments .

  • Breast Cancer: Distinguishes ductal vs. lobular carcinoma via membranous vs. cytoplasmic beta-catenin staining patterns .

Mechanism of Action Studies

  • Wnt Pathway Analysis: Used to quantify CTNNB1 dynamics in live-cell imaging, revealing nuclear-cytoplasmic shuttling modulated by Wnt signaling .

Comparative Advantages

FeatureCTNNB1 Recombinant AntibodyConventional Polyclonal Antibodies
SpecificityMono-specific (S-score ≥2.5) Risk of cross-reactivity
Batch ConsistencyHigh (recombinant production) Variable
Multiplex CompatibilityCompatible with IF, FACS, and IHC Limited by host species

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 of receiving your order. Delivery times may vary depending on the purchase method or location. Please consult your local distributors for specific delivery timelines.
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, also known as β-catenin, 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, promoting phosphorylation on N-terminal Ser and Thr residues and ubiquitination of CTNNB1 via BTRC. This leads to the subsequent degradation of CTNNB1 by the proteasome. In the presence of Wnt ligand, CTNNB1 is not ubiquitinated and accumulates in the nucleus, acting as a coactivator for transcription factors of the TCF/LEF family, leading to the activation of Wnt-responsive genes. CTNNB1 is involved in regulating cell adhesion as a component of an E-cadherin:catenin adhesion complex. It functions as a negative regulator of centrosome cohesion and participates in the CDK2/PTPN6/CTNNB1/CEACAM1 pathway of insulin internalization. CTNNB1 blocks anoikis of malignant kidney and intestinal epithelial cells, promoting their anchorage-independent growth by downregulating DAPK2. It disrupts PML function and PML-NB formation by inhibiting RANBP2-mediated sumoylation of PML. Additionally, CTNNB1 promotes neurogenesis by maintaining sympathetic neuroblasts within the cell cycle and is involved in chondrocyte differentiation via interaction with SOX9. SOX9 binding competes with the binding sites of TCF/LEF within CTNNB1, inhibiting Wnt signaling.
Gene References Into Functions
  1. CXC chemokine ligand 9 promotes the progression of diffuse large B-cell lymphoma in a β-catenin-dependent manner. PMID: 30130730
  2. Research suggests that epigenetic regulation of CTNNB1 may serve as a novel avenue to block colon cancer cell migration and invasion. PMID: 29923144
  3. Results demonstrate that 2HF could inhibit EMT, and cell migration and invasion through the Wnt/βcatenin signaling pathway by suppressing GSK3b phosphorylation, βcatenin expression, and transactivation. PMID: 30226607
  4. Collectively, these studies suggest that the cellular transcription factor β-catenin stimulates productive herpes simplex virus 1 infection, in part because VP16 enhances β-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 pattern. PMID: 29224720
  7. Data reveal that post-translational modifications of β-catenin in the ubiquitin-proteasome pathway yield a truncated β-catenin molecule containing a serine 552-phosphorylated core region without N and C termini. This proteolytic processing of β-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/β-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/β-catenin signaling pathway to promote epithelial-mesenchymal transition in oral squamous carcinoma stem cells by down-regulating SFRP1. PMID: 28425477
  12. The β-catenin pathway is activated by CBX8 in hepatocellular carcinoma. PMID: 29066512
  13. Our data provide novel evidence for the biological and clinical significance of SPAG5 as a potential biomarker, and we demonstrate that SPAG5-β-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 β-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 β-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; β-catenin signaling is critical for TBL1XR1-mediated colorectal cancer cells oncogenicity. PMID: 28295012
  17. Taken together, these results suggest that Wnt/β-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 β-catenin/E-cadherin complex in the osteoblast membrane under stretch loading and the subsequent translocation of β-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-β-catenin signaling. PMID: 29901121
  21. High CTNNB1 expression is associated with the recurrence of Adamantinomatous Craniopharyngiomas. PMID: 29625497
  22. High CTNNB1 expression is associated with uterine fibroids. PMID: 29066531
  23. The nucleus and/or cytoplasm expression of β-catenin was associated with tumor progression and correlated overall survival of patients with ovarian cancer (OC). β-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 β-catenin and enhanced the interaction between β-catenin and TCF4 T-cell factor. PMID: 29317218
  26. Nuclear β-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 β-catenin expression require careful diagnosis. PMID: 29496310
  27. High CTNNB1 expression is associated with metastasis in cholangiocarcinoma. PMID: 30193944
  28. β;-catenin directly interacts with the Cx43 carboxyl-terminal domain. PMID: 29882937
  29. This study showed that β-catenin expression was most evident in the nucleus rather than in the cytoplasm. PMID: 29297710
  30. Nuclear β-catenin accumulation in non-mitotic glioblastoma cells is due to a feed-forward mechanism between DOCK4 and β-catenin. PMID: 28925399
  31. Study found that HIF1alpha overexpression led to enhanced βcatenin nuclear translocation, while βcatenin silencing inhibited βcatenin nuclear translocation. The enhanced βcatenin nuclear translocation induced resulted in enhanced cell proliferation and cell invasion, altered cell cycle distribution, decreased apoptosis, and improved nonhomologous end joining repair under normal and irradiation conditions... PMID: 29658569
  32. Our results demonstrated that miR-188 inhibits glioma cell proliferation by targeting β-catenin. PMID: 29268818
  33. Marked upregulation of β-catenin and its downstream targets effectively enhanced hepatosphere formation, with an associated induction of CD133, OCT4, and Sox2 expression and also caused a significant enhancement of HCC proliferation. PMID: 29792038
  34. The Wnt/β-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 β-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. β-catenin immunopositivity is seen in the majority of cases of sinonasal sarcoma. PMID: 29566950
  39. For the first time, we demonstrated that rather than excluding lymphocytes infiltration as reported in melanoma, high levels of TILs were associated with β-catenin overexpression in BC. PMID: 29286921
  40. Study shows that apigenin-induced lysosomal degradation of β-catenin in Wnt/β-catenin signaling. PMID: 28337019
  41. Used CRISPR-Cas9 technology to study the effect of knockout of catenin beta 1 (CTNNB1) on cell behavior and signal pathways in HEK293 cells. Results showed knockout of CTNNB1 affected the Wnt/β-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/β catenin signaling activity by suppressing β-catenin expression and reversing the downstream target gene expression. Taken together, lncRNA SNHG20 plays a pivotal role in ovarian cancer progression by regulating Wnt/β-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/β-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 β-catenin, E-cadherin, and Snail, depending on clinico-morphological aspects of the laryngeal squamous cell carcinomas. Results revealed variable E-cadherin, β-catenin, and Snail expression, depending on differentiation degree and tumor stage. PMID: 29250652
  45. In this study, we showed that the activation of the Wnt/β-catenin pathway culminates in the upregulation of the MGAT1 enzyme at both transcriptional and post-transcriptional levels. We also showed that overexpression of the β-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 the 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 β-catenin activation that can occur within Inflammatory hepatocellular adenoma (IHCAs). PMID: 28618047
  49. Results show that the E-cadherin/β-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 miR200a-mediated β-catenin pathway. Our data suggest that Toosendanin may suppress oncogenic phenotypes of human GC cells partly via the miR200a/β-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 CTNNB1 and why is it important in research?

CTNNB1 (catenin beta 1) encodes beta-catenin, a 92 kDa protein that functions as a key downstream component of the canonical Wnt signaling pathway . The protein plays dual roles in cellular function: it serves as a structural component in cell adhesion complexes and as a critical signaling molecule in the Wnt pathway, making it essential for various developmental and homeostatic processes . In the absence of Wnt ligands, beta-catenin forms a complex with AXIN1, AXIN2, APC, CSNK1A1, and GSK3B, which promotes its phosphorylation, ubiquitination, and subsequent proteasomal degradation . Conversely, when Wnt ligands are present, beta-catenin accumulates in the nucleus and acts as a coactivator for TCF/LEF family transcription factors, activating Wnt-responsive genes . Beta-catenin's involvement in numerous cellular processes, including cell adhesion, centrosome cohesion, insulin internalization, and anoikis resistance, has made it a crucial target for research on development, cancer, and other diseases .

What is the difference between recombinant monoclonal antibodies and traditional monoclonal antibodies for CTNNB1 detection?

Recombinant monoclonal antibodies for CTNNB1 detection, such as the rabbit recombinant monoclonal antibody CTNNB1/2030R, are produced using recombinant DNA technology to ensure batch-to-batch consistency and specificity . Unlike traditional hybridoma-derived monoclonal antibodies, recombinant antibodies are generated by cloning antibody genes into expression vectors, allowing for precise control over production and reducing variability . This technology eliminates the need for animal immunization for each production batch, ensuring more standardized reagents for research applications .

Traditional monoclonal antibodies like the mouse monoclonal clone 5H10 are typically produced using hybridoma technology after immunizing animals with the target antigen . While these antibodies have been widely used, they may exhibit greater batch-to-batch variation compared to recombinant antibodies . For CTNNB1 detection specifically, recombinant antibodies typically offer improved reproducibility in applications such as immunohistochemistry, western blotting, and immunofluorescence, which is critical for longitudinal studies requiring consistent reagents .

What cellular compartments does beta-catenin localize to, and how does this affect antibody selection?

Beta-catenin demonstrates complex subcellular localization patterns that reflect its multifunctional roles in cellular processes . As documented in technical specifications for CTNNB1 antibodies, beta-catenin can be found in multiple cellular compartments including: adherens junctions, cell junctions, cell membrane, centrosome, cilium basal body, cytoplasm, cytoskeleton, microtubule organizing center, nucleus, spindle pole, and synapse . This diverse localization profile corresponds to beta-catenin's involvement in both structural roles (at cell junctions) and signaling functions (in the nucleus) .

When selecting antibodies for beta-catenin detection, researchers must consider the specific cellular compartment they wish to analyze . For instance, detecting nuclear beta-catenin accumulation (indicative of active Wnt signaling) might require antibodies that perform well under the fixation and permeabilization conditions needed for nuclear protein preservation . Some antibodies may preferentially recognize certain conformational states or post-translationally modified forms of beta-catenin that predominate in specific compartments . Researchers should carefully review validation data showing the antibody's performance in detecting beta-catenin in their compartment of interest, as demonstrated in immunofluorescence images showing membrane, cytoplasmic, and nuclear staining patterns .

How should I optimize immunohistochemistry protocols for CTNNB1 detection in formalin-fixed tissues?

Optimizing immunohistochemistry (IHC) protocols for CTNNB1 detection in formalin-fixed, paraffin-embedded (FFPE) tissues requires careful attention to several critical parameters . Based on technical specifications for recombinant CTNNB1 antibodies, the following methodological approach is recommended:

  • Antigen retrieval: Heat-induced epitope retrieval is essential for optimal CTNNB1 detection. Specifically, tissues should be heated in 10mM Tris with 1mM EDTA, pH 9.0, for 45 minutes at 95°C, followed by cooling at room temperature for 20 minutes . This step is crucial for breaking protein cross-links formed during formalin fixation, which can mask the beta-catenin epitope.

  • Antibody concentration: For rabbit recombinant monoclonal antibodies like CTNNB1/2030R, use a concentration of 1-2 μg/ml for optimal staining . This concentration typically provides the best signal-to-noise ratio for beta-catenin detection.

  • Incubation conditions: Incubate the primary antibody for 30 minutes at room temperature . Longer incubation times may increase background staining without improving specific signal.

  • Positive controls: Include known positive controls such as human brain tissue, breast carcinoma samples, or cell lines like HeLa or MCF-7 that express detectable levels of beta-catenin . These controls help validate the staining procedure and provide a reference for expected staining patterns.

  • Detection system: Use a detection system appropriate for rabbit IgG antibodies, typically based on polymer-HRP technology, followed by DAB (3,3'-diaminobenzidine) visualization .

When evaluating staining results, remember that beta-catenin localization is biologically significant—membrane staining represents adhesion functions, while nuclear accumulation indicates active Wnt signaling . In colorectal cancer tissues, for example, abnormal nuclear accumulation of beta-catenin is frequently observed and can be compared with adjacent normal tissues showing predominantly membranous staining .

What are the optimal conditions for using CTNNB1 antibodies in western blotting applications?

For optimal western blotting results with CTNNB1 antibodies, researchers should follow these methodological guidelines based on validated protocols :

  • Sample preparation:

    • Extract total protein using RIPA buffer supplemented with protease inhibitors

    • For complete protein extraction, consider including phosphatase inhibitors to preserve phosphorylated forms of beta-catenin

    • Use positive control samples such as HeLa cell lysates, which express detectable levels of beta-catenin

  • Protein loading and separation:

    • Load 20-30 μg of total protein per lane

    • Use 8-10% polyacrylamide gels to achieve optimal separation around the 92 kDa range (beta-catenin molecular weight)

    • Include a molecular weight marker to confirm the expected size

  • Transfer conditions:

    • Transfer to PVDF membrane (preferred over nitrocellulose for beta-catenin detection)

    • Use semi-dry or wet transfer systems at 100V for 60-90 minutes with cold transfer buffer

  • Antibody incubation:

    • Block membrane with 5% non-fat dry milk in TBST for 1 hour at room temperature

    • For rabbit recombinant monoclonal antibodies, dilute to 2-4 μg/ml in blocking solution

    • For mouse monoclonal antibodies like clone 5H10, dilute to 0.5-1 μg/ml

    • Incubate with primary antibody overnight at 4°C with gentle agitation

    • Wash 3-5 times with TBST, 5 minutes each

    • Incubate with appropriate HRP-conjugated secondary antibody (1:5000-1:10000) for 1 hour at room temperature

  • Detection and analysis:

    • Develop using ECL substrate and image using a digital imaging system

    • Expected band size for full-length beta-catenin is approximately 92 kDa

    • In some cases, smaller bands may represent degradation products or splice variants

Antibody TypeRecommended DilutionIncubation ConditionsExpected Band Size
Rabbit recombinant monoclonal (CTNNB1/2030R)2-4 μg/mlOvernight at 4°C92 kDa
Mouse monoclonal (5H10)0.5-1 μg/mlOvernight at 4°C92 kDa

For quantitative analysis, researchers should normalize beta-catenin expression to loading controls such as GAPDH or β-actin, and consider analyzing subcellular fractions separately to distinguish between cytoplasmic, nuclear, and membrane-associated pools of beta-catenin .

How can I use CTNNB1 antibodies for flow cytometry applications?

Using CTNNB1 antibodies for flow cytometry requires specific methodology to achieve accurate detection of both surface-associated and intracellular beta-catenin . Based on validated protocols, follow these procedural guidelines:

  • Sample preparation:

    • For cell lines: Harvest adherent cells using non-enzymatic cell dissociation solution rather than trypsin to preserve cell surface proteins

    • For primary tissues: Generate single-cell suspensions using gentle mechanical dissociation combined with appropriate tissue-specific digestion

    • Wash cells twice with cold PBS containing 2% FBS (FACS buffer)

  • Fixation and permeabilization:

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

    • Permeabilize with 0.1% Triton X-100 or commercial permeabilization buffer for 10 minutes

    • This step is crucial for accessing intracellular and nuclear beta-catenin pools

  • Antibody staining:

    • Block with 5% normal serum from the same species as the secondary antibody

    • For rabbit recombinant monoclonal antibodies: Use 1-2 μg per 10^6 cells

    • For mouse monoclonal antibodies: Use 0.5-1 μg per 10^6 cells

    • Incubate for 30-45 minutes on ice

    • Wash twice with FACS buffer

    • Incubate with fluorophore-conjugated secondary antibody at manufacturer's recommended dilution

    • Include appropriate isotype controls and single-color controls for compensation

  • Analysis considerations:

    • Analyze beta-catenin expression in conjunction with cell surface markers for population identification

    • Interpret beta-catenin signal intensity in the context of subcellular localization

    • Consider co-staining with markers of Wnt pathway activation

Flow cytometric analysis of beta-catenin has been validated in multiple cell types, including HeLa cells, where PFA-fixed, permeabilized cells show distinct staining patterns when labeled with recombinant rabbit monoclonal antibody (CTNNB1/2030R) followed by goat anti-rabbit IgG-CF488 . This approach enables quantitative assessment of beta-catenin levels at the single-cell level, allowing researchers to correlate expression with other cellular parameters or identify distinct cell populations based on beta-catenin expression levels .

How can I troubleshoot non-specific background staining in immunohistochemistry using CTNNB1 antibodies?

Non-specific background staining is a common challenge when using CTNNB1 antibodies for immunohistochemistry . Based on technical expertise and validated protocols, here are methodological approaches to systematically identify and resolve this issue:

  • Optimize antibody concentration:

    • Excessive antibody concentration often causes high background

    • Perform a titration experiment using serial dilutions (0.5-4 μg/ml) of the CTNNB1 antibody on known positive controls

    • For recombinant monoclonal antibodies like CTNNB1/2030R, start with the recommended 1-2 μg/ml concentration and adjust as needed

    • The optimal concentration provides specific staining with minimal background

  • Improve blocking procedures:

    • Insufficient blocking is a major cause of background staining

    • Extend blocking time to 1-2 hours using 5-10% normal serum from the same species as the secondary antibody

    • Add 0.1-0.3% Triton X-100 to blocking solution to reduce non-specific binding

    • Consider using commercial protein blockers that contain multiple blocking agents

  • Modify antigen retrieval protocols:

    • Overly aggressive antigen retrieval can increase background staining

    • Compare the recommended Tris-EDTA method (pH 9.0, 45 minutes at 95°C) with milder conditions

    • Consider alternative buffers (citrate buffer, pH 6.0) if background persists

    • Allow slides to cool gradually after heating to prevent tissue damage

  • Reduce endogenous enzyme activity:

    • For peroxidase-based detection systems, block endogenous peroxidase activity with 3% hydrogen peroxide for 10 minutes before antibody incubation

    • For alkaline phosphatase systems, add levamisole to block endogenous activity

  • Optimize washing steps:

    • Increase wash duration and frequency (e.g., 5 washes for 5 minutes each)

    • Ensure washing buffer covers the entire tissue section

    • Use gentle agitation during washing steps

  • Control comparison analysis:

    • Always run negative controls (primary antibody omitted or isotype control)

    • Compare staining patterns with known positive controls (e.g., human brain, breast carcinoma)

    • Include normal adjacent tissue as an internal control when analyzing tumor samples

  • Tissue-specific considerations:

    • Some tissues (like liver, kidney) have higher endogenous biotin, requiring streptavidin/biotin blocking

    • Highly vascularized tissues may require additional blocking of endogenous immunoglobulins

When analyzing results, remember that genuine beta-catenin staining shows distinct cellular localization patterns—membranous at cell junctions, cytoplasmic, and/or nuclear—depending on the tissue and Wnt signaling status . Background staining typically appears diffuse and lacks this specific localization pattern.

What could cause variability in CTNNB1 detection across different experimental replicates?

Variability in CTNNB1 detection across experimental replicates can stem from multiple methodological and biological factors . Understanding and controlling these factors is essential for obtaining reproducible results:

  • Antibody-related factors:

    • Lot-to-lot variability: Traditional monoclonal antibodies may show greater variation between production lots compared to recombinant antibodies

    • Antibody storage conditions: Repeated freeze-thaw cycles or improper storage can reduce antibody performance

    • Solution preparation: Variations in antibody dilution calculation or buffer composition

    • Recommended solution: Use recombinant monoclonal antibodies like CTNNB1/2030R that offer improved lot-to-lot consistency

  • Sample preparation variables:

    • Fixation timing: Delay between tissue collection and fixation affects protein preservation

    • Fixation duration: Over- or under-fixation impacts epitope accessibility

    • Processing variations: Inconsistent dehydration or embedding procedures

    • Storage effects: Prolonged storage of paraffin blocks or cut sections

    • Recommended solution: Standardize sample collection and processing protocols; prepare all experimental samples simultaneously when possible

  • Protocol execution inconsistencies:

    • Antigen retrieval: Variations in heating temperature, time, or buffer composition

    • Wash step execution: Inconsistent washing can affect background and signal intensity

    • Timing differences: Variations in incubation times for primary or secondary antibodies

    • Detection system preparation: Inconsistent substrate development times

    • Recommended solution: Use automated staining platforms when available; maintain detailed protocol documentation

  • Biological considerations:

    • Beta-catenin levels fluctuate with Wnt pathway activation status

    • Protein degradation proceeds rapidly after tissue collection

    • Subcellular localization changes with cellular context

    • Recommended solution: Control for experimental timing; consider using phosphatase inhibitors to preserve signaling states

  • Analysis and interpretation factors:

    • Subjective thresholding for positive/negative determination

    • Inconsistent region selection for quantification

    • Variation in imaging parameters (exposure, gain settings)

    • Recommended solution: Implement quantitative image analysis with standardized parameters; blind analysis when possible

To systematically address variability, implement a controlled comparative analysis approach:

VariableControl MeasureImplementation Strategy
Antibody factorsUse consistent lot numbersPurchase sufficient quantity for entire study
Sample preparationStandardize processingProcess all samples in parallel when possible
Protocol executionDetailed documentationCreate checklist-based protocols
Biological variationInclude reference samplesProcess known positive control in each batch
Analysis methodsAutomated quantificationUse image analysis software with fixed settings

How can I verify the specificity of my CTNNB1 antibody for my particular application?

Verifying antibody specificity is crucial for generating reliable data with CTNNB1 antibodies . Here is a comprehensive methodological approach to confirm specificity for your particular application:

  • Genetic validation approaches:

    • siRNA/shRNA knockdown: Transfect cells with CTNNB1-targeting siRNA/shRNA and control constructs, then perform your detection method (western blot, IHC, etc.) to confirm signal reduction in knockdown samples

    • CRISPR-Cas9 knockout: Generate CTNNB1 knockout cell lines as the most stringent negative control; validate complete knockout by sequencing

    • Overexpression systems: Transfect cells with CTNNB1 expression vectors to create positive controls with elevated beta-catenin levels

  • Biochemical validation strategies:

    • Peptide competition assay: Pre-incubate the antibody with recombinant CTNNB1 protein or immunizing peptide before application; specific signals should be abolished or significantly reduced

    • Immunoprecipitation-Mass Spectrometry: Perform immunoprecipitation with the CTNNB1 antibody followed by mass spectrometry to confirm target identity

    • Epitope mapping: For antibodies with known epitopes, analyze binding to synthesized peptide fragments containing the epitope sequence

  • Cross-validation with independent antibodies:

    • Orthogonal antibody testing: Compare results using multiple CTNNB1 antibodies with different epitopes (e.g., compare CTNNB1/2030R with clone 5H10)

    • Detection of expected expression patterns: Verify that staining patterns match known beta-catenin localization (membrane-associated in normal epithelia, nuclear in Wnt-activated cells)

    • Antibody cross-reactivity assessment: Test antibody on cells/tissues from different species to confirm specificity toward intended species targets

  • Application-specific validation:

    • For IHC: Confirm antibody performance by staining tissues with known beta-catenin expression patterns (e.g., colon cancer with nuclear beta-catenin)

    • For Western blotting: Verify detection of a single band at the expected molecular weight (92 kDa) without significant non-specific bands

    • For immunofluorescence: Confirm co-localization with known interaction partners (e.g., E-cadherin at cell junctions)

    • For flow cytometry: Compare staining pattern with isotype controls and validate using cells with known beta-catenin expression levels

  • Experimental controls to include routinely:

    • Technical negative controls: Omit primary antibody while maintaining all other steps

    • Biological negative controls: Use tissues/cells known to lack or have minimal CTNNB1 expression

    • Positive controls: Include samples with established beta-catenin expression (e.g., HeLa cells, MCF-7 cells, human brain tissue)

For recombinant antibodies like CTNNB1/2030R, vendor validation data often includes protein array analysis against more than 19,000 full-length human proteins, providing comprehensive specificity information . This data should be reviewed carefully before selecting an antibody for your specific application.

How can CTNNB1 antibodies be used to investigate Wnt signaling pathway activation in cancer tissues?

CTNNB1 antibodies are powerful tools for investigating Wnt signaling pathway activation in cancer tissues, offering insights into this frequently dysregulated pathway . The following methodological approaches leverage these antibodies for comprehensive analysis:

  • Subcellular localization analysis:

    • Nuclear accumulation of beta-catenin serves as a direct indicator of canonical Wnt pathway activation

    • Immunohistochemistry with recombinant monoclonal antibodies like CTNNB1/2030R (1-2 μg/ml) can visualize this translocation

    • Quantitative assessment involves calculating the percentage of tumor cells with nuclear beta-catenin staining

    • Compare staining patterns between tumor tissue and adjacent normal epithelium, where beta-catenin is typically restricted to membrane locations

    • Confocal microscopy with CTNNB1 antibodies can provide high-resolution subcellular localization data

  • Correlation with clinical parameters:

    • Stratify tumors based on beta-catenin localization patterns (membranous, cytoplasmic, nuclear)

    • Correlate staining patterns with:

      • Tumor stage and grade

      • Patient survival outcomes

      • Treatment response profiles

      • Molecular subtypes

    • Integrate with other Wnt pathway component analyses (e.g., APC, AXIN1/2, GSK3B)

  • Multi-marker analysis approaches:

    • Combine CTNNB1 antibody staining with antibodies against:

      • Wnt target genes (e.g., AXIN2, c-MYC, Cyclin D1)

      • Other pathway components (e.g., DVL, LRP5/6)

      • Post-translational modifications (e.g., phospho-beta-catenin)

    • Use multiplexed immunofluorescence or sequential immunohistochemistry techniques

    • Analyze co-expression patterns to assess pathway activation state

  • Single-cell analysis techniques:

    • Flow cytometry with CTNNB1 antibodies (1-2 μg/10^6 cells) enables quantitative assessment of beta-catenin levels at single-cell resolution

    • Combined with surface markers for cell lineage identification

    • Mass cytometry (CyTOF) with metal-conjugated CTNNB1 antibodies provides multi-parameter data

    • Single-cell sequencing approaches correlated with protein-level data can reveal heterogeneity in Wnt activation within tumors

  • Functional validation experiments:

    • Immunoprecipitation with CTNNB1 antibodies to isolate beta-catenin protein complexes

    • Chromatin immunoprecipitation (ChIP) to identify beta-catenin-bound genomic regions

    • Proximity ligation assays (PLA) to detect interactions between beta-catenin and TCF/LEF transcription factors

Recent research utilizing CTNNB1 antibodies has revealed that colorectal cancer tissues often show stronger activity of the intestinal enhancer of CTNNB1 (ieCTNNB1) compared to adjacent normal tissues, highlighting the regulatory mechanisms controlling beta-catenin expression levels . This enhancer-dependent mechanism has been shown to control the dosage of Wnt signaling and homeostasis in intestinal epithelia, with potential implications for targeted therapy development .

What are the considerations for using CTNNB1 antibodies in multiplex immunofluorescence applications?

Multiplex immunofluorescence (mIF) with CTNNB1 antibodies enables simultaneous visualization of beta-catenin and other proteins, providing comprehensive spatial information about Wnt signaling in the context of the tissue microenvironment . Successfully implementing this advanced technique requires attention to several methodological considerations:

  • Antibody selection and validation:

    • Choose recombinant monoclonal antibodies like CTNNB1/2030R that offer high specificity and low background

    • Validate each antibody individually before multiplexing

    • Select antibodies raised in different host species to avoid cross-reactivity in secondary detection

    • For same-species antibodies, consider directly conjugated primary antibodies or sequential staining with intermediate blocking steps

    • Verify epitope compatibility with fixation and antigen retrieval methods

  • Panel design optimization:

    • Combine CTNNB1 antibodies with markers that answer specific biological questions:

      • Wnt pathway components (e.g., GSK3β, APC, Axin)

      • Cell type markers (e.g., EpCAM, CD45, CD31)

      • Functional markers (e.g., Ki67, cleaved caspase-3)

    • Balance fluorophore selection based on:

      • Spectral overlap considerations

      • Signal intensity relative to target abundance

      • Tissue autofluorescence profile

    • Include single-color controls for spectral unmixing

  • Technical protocol considerations:

    • Optimal CTNNB1 antibody dilution for mIF is 1-3 μg/ml

    • Sequential staining approach:

      • Apply CTNNB1 antibody first at 1-3 μg/ml for 1 hour at room temperature

      • Wash thoroughly (3-5 times with PBS-T)

      • Apply fluorophore-conjugated secondary antibody

      • Block unbound antibody sites before next primary antibody

      • Repeat for additional markers

    • For cyclic immunofluorescence, validate antibody stripping efficiency between cycles

  • Imaging and analysis strategies:

    • Capture high-resolution images using confocal or structured illumination microscopy

    • Implement computational analysis approaches:

      • Cell segmentation to identify individual cells

      • Subcellular compartment analysis (membrane, cytoplasm, nucleus)

      • Colocalization measurements with other markers

      • Spatial relationship analysis between different cell populations

    • Quantitative metrics to consider:

      • Nuclear:cytoplasmic beta-catenin ratio

      • Membrane beta-catenin intensity

      • Colocalization coefficients with interacting partners

  • Experimental controls and validation:

    • Include tissue-matched negative controls (primary antibody omission)

    • Use positive control tissues with known beta-catenin expression patterns

    • Validate multiplex findings with orthogonal methods (e.g., single-marker IHC, western blotting)

    • Consider biological controls (e.g., Wnt pathway activators/inhibitors in cell lines)

Immunofluorescence analysis of beta-catenin localization has been successfully demonstrated in HeLa cells using the CTNNB1/2030R antibody, showing specific staining patterns that can be combined with nuclear counterstaining . This approach can be extended to multiplex applications by carefully selecting compatible antibodies and fluorophores.

How can CTNNB1 antibodies be used to study beta-catenin's role in development and stem cell biology?

CTNNB1 antibodies provide powerful tools for investigating beta-catenin's critical roles in development and stem cell biology, where Wnt signaling orchestrates cell fate decisions and tissue patterning . The following methodological approaches enable detailed analysis of these processes:

  • Lineage-specific expression analysis in development:

    • Immunohistochemical mapping: Apply CTNNB1 antibodies (1-2 μg/ml) to embryonic tissue sections at different developmental stages

    • Triple immunofluorescence: Combine beta-catenin detection with lineage markers and proliferation/differentiation indicators

    • Whole-mount immunostaining: For transparent embryos or organoids, use CTNNB1 antibodies with clearing techniques for 3D visualization

    • Quantitative assessment: Measure nuclear beta-catenin levels as development progresses to identify temporal activation patterns

  • Stem cell niche characterization:

    • Adult stem cell identification: In tissues like intestine, where Lgr5+ crypt base columnar cells depend on Wnt signaling, CTNNB1 antibodies reveal signaling gradients

    • Co-localization studies: Combine CTNNB1 antibodies with stem cell markers (e.g., Lgr5, CD133) and niche factors

    • Ex vivo tissue analysis: Apply beta-catenin antibodies to freshly isolated stem cell populations

    • Organoid culture systems: Track beta-catenin dynamics during organoid formation and differentiation

  • Cell fate transition monitoring:

    • Time-course experiments: Apply CTNNB1 antibodies to capture dynamic changes during differentiation

    • Single-cell resolution techniques: Use flow cytometry with CTNNB1 antibodies (1-2 μg/10^6 cells) to quantify protein levels as cells transition between states

    • Live cell imaging: For real-time visualization, use fluorescently-tagged antibody fragments in live cell-compatible systems

    • Correlative microscopy: Combine functional assays with subsequent immunostaining for beta-catenin

  • Genetic perturbation analysis:

    • Enhancer studies: Investigate tissue-specific enhancers like ieCtnnb1, which regulates Wnt signaling dosage in intestinal epithelia

    • Knockout/knockdown phenotyping: After CTNNB1 genetic manipulation, use antibodies to confirm protein reduction and analyze resulting developmental defects

    • Rescue experiments: After genetic disruption, restore beta-catenin expression and use antibodies to confirm localization patterns

    • Pathway modulation: Apply Wnt activators/inhibitors and track beta-catenin localization changes

  • Specialized applications in stem cell biology:

    • Chromatin immunoprecipitation (ChIP): Use CTNNB1 antibodies to identify genomic binding sites during differentiation

    • Proximity ligation assay (PLA): Detect interactions between beta-catenin and lineage-specific transcription factors

    • Mass cytometry: Combine CTNNB1 antibodies with dozens of other markers for comprehensive profiling

    • Spatial transcriptomics correlation: Integrate beta-catenin protein localization data with spatial gene expression patterns

Recent research has identified tissue-specific enhancers controlling CTNNB1 expression, such as the intestinal enhancer ieCtnnb1/ieCTNNB1, which regulates Wnt signaling dosage and epithelial homeostasis . This enhancer is transcriptionally active in both Lgr5-expressing crypt base columnar cells and other crypt cells, highlighting the importance of precise beta-catenin regulation in maintaining intestinal stem cell function . CTNNB1 antibodies have been instrumental in characterizing these regulatory mechanisms and their impact on development and tissue maintenance.

How are new enhancer mechanisms affecting our understanding of CTNNB1 regulation?

Recent discoveries regarding enhancer-mediated regulation of CTNNB1 are revolutionizing our understanding of how beta-catenin levels and Wnt signaling are controlled in a tissue-specific manner . These findings have significant implications for both basic research and therapeutic development:

  • Intestinal enhancer of CTNNB1 (ieCtnnb1/ieCTNNB1):

    • Recently identified as a critical regulatory element located 29,068 base pairs upstream of the CTNNB1 transcription start site

    • Displays typical enhancer features (open chromatin with enriched H3K27ac and H3K4me1) specifically in intestinal tissues

    • Resides within the same topologically associating domain (TAD) as CTNNB1, enabling long-range chromatin interactions

    • Deletion of ieCtnnb1 in mouse models leads to:

      • Altered intestinal epithelial composition

      • Compromised small intestinal crypt function

      • Hampered epithelial turnover under normal physiological conditions

      • Prevention of Wnt/β-catenin-driven colorectal cancer development and progression

  • Transcription factor regulation of CTNNB1 enhancers:

    • Key trans-factors binding to ieCTNNB1 include HNF4α and phosphorylated CREB1

    • These factors regulate CTNNB1 expression in a tissue-specific manner

    • The enhancer's activity is stronger in colorectal cancer tissues compared to adjacent normal tissues

    • Human ieCTNNB1 contains a single-nucleotide polymorphism associated with CTNNB1 expression levels in gastrointestinal epithelia

  • Tissue-specific enhancer network:

    • Beyond ieCtnnb1, other tissue-specific enhancers have been identified:

      • neCtnnb1: Regulates neocortical neurogenesis

      • Potentially other uncharacterized enhancers in different tissues

    • These findings reveal a complex enhancer landscape controlling beta-catenin expression

    • Multiple enhancers likely act in concert to fine-tune CTNNB1 levels in different developmental and physiological contexts

  • Methodological approaches to study enhancer-mediated regulation:

    • Reporter gene assays using enhancer regions driving expression of marker genes

    • CRISPR-Cas9-mediated deletion of enhancer regions

    • Chromosome conformation capture techniques (3C, 4C, Hi-C) to map enhancer-promoter interactions

    • Single-cell RNA sequencing to assess cell type-specific effects of enhancer activity

    • CTNNB1 antibodies to measure resulting protein expression changes after enhancer manipulation

  • Therapeutic implications:

    • Enhancer-targeting approaches may provide more tissue-specific modulation of Wnt signaling compared to direct CTNNB1 targeting

    • Polymorphisms in enhancer regions could predict susceptibility to Wnt-driven diseases

    • Epigenetic therapies might modulate enhancer activity to normalize CTNNB1 expression in disease states

These discoveries highlight the inadequacy of a one-size-fits-all approach to studying beta-catenin regulation and suggest that tissue-specific enhancers play crucial roles in fine-tuning Wnt signaling output . For researchers using CTNNB1 antibodies, these findings emphasize the importance of considering the cellular context and regulatory landscape when interpreting beta-catenin expression and localization patterns.

What emerging technologies are improving CTNNB1 antibody applications in research?

Emerging technologies are significantly enhancing the utility of CTNNB1 antibodies in research settings, enabling more precise, quantitative, and multidimensional analyses of beta-catenin biology . These technological advances include:

  • Advanced imaging innovations:

    • Super-resolution microscopy: Techniques like STORM, PALM, and STED provide nanoscale resolution of beta-catenin localization, revealing previously undetectable distribution patterns at adherens junctions and nuclear complexes

    • Lattice light-sheet microscopy: Enables long-term, low-phototoxicity imaging of beta-catenin dynamics in living systems

    • Expansion microscopy: Physical enlargement of specimens allows conventional microscopes to achieve super-resolution imaging of beta-catenin networks

    • Correlative light and electron microscopy (CLEM): Combines the specificity of CTNNB1 immunofluorescence with ultrastructural context

  • Single-cell analysis technologies:

    • Mass cytometry (CyTOF): Metal-labeled CTNNB1 antibodies enable simultaneous measurement of dozens of proteins in single cells

    • Imaging mass cytometry: Combines mass spectrometry with microscopy for spatial proteomic analysis

    • Single-cell Western blotting: Measures beta-catenin levels and modifications in individual cells

    • Microfluidic approaches: Facilitate high-throughput single-cell protein analysis with minimal sample input

  • Spatial multi-omics integration:

    • Digital spatial profiling: Combines CTNNB1 antibody staining with region-specific molecular analysis

    • Spatial transcriptomics: Correlates beta-catenin protein localization with spatially resolved gene expression

    • Multiplexed ion beam imaging (MIBI): Enables simultaneous imaging of 40+ targets including CTNNB1

    • Co-detection by indexing (CODEX): Allows highly multiplexed imaging using CTNNB1 antibodies alongside numerous other markers

  • Antibody engineering advancements:

    • Recombinant nanobodies: Smaller binding domains with superior tissue penetration for beta-catenin detection in thick specimens

    • Site-specific conjugation: Precisely positioned fluorophores or other labels that minimize interference with antibody binding

    • Bispecific antibodies: Simultaneously target beta-catenin and interacting partners to study protein complexes

    • Intrabodies: Genetically encoded antibody fragments that can track beta-catenin in living cells

  • Computational analysis pipelines:

    • Deep learning image analysis: Automated detection and quantification of beta-catenin subcellular localization

    • Trajectory inference algorithms: Track beta-catenin signaling states through developmental or disease progressions

    • Network analysis tools: Integrate beta-catenin data with other pathway components

    • Virtual tissue modeling: Incorporate beta-catenin distribution data into predictive models of tissue behavior

These technological innovations are particularly valuable for studying the complex regulatory mechanisms of beta-catenin, such as the recently discovered enhancer-dependent control of CTNNB1 expression in intestinal epithelia . For example, single-cell sequencing technologies have revealed that deletion of the intestinal enhancer ieCtnnb1 alters epithelial cell compositions and compromises intestinal crypt functions, demonstrating how advanced techniques can provide mechanistic insights into beta-catenin regulation and function .

What are the key considerations for selecting the optimal CTNNB1 antibody for specific research applications?

Selecting the optimal CTNNB1 antibody requires careful consideration of multiple factors to ensure reliable, reproducible, and meaningful results across different research applications . Based on the comprehensive evidence reviewed, researchers should prioritize the following key considerations:

  • Application compatibility and validation: Choose antibodies specifically validated for your intended application (IHC, WB, IF, flow cytometry) . For example, the rabbit recombinant monoclonal antibody CTNNB1/2030R has been validated for western blotting (2-4 μg/ml), immunohistochemistry (1-2 μg/ml), immunofluorescence (1-3 μg/ml), and flow cytometry (1-2 μg/10^6 cells) . Review validation data including images showing expected staining patterns in appropriate control samples.

  • Epitope characteristics and accessibility: Consider the location of the antibody's target epitope within the beta-catenin protein . Antibodies recognizing different regions may perform differently depending on protein conformation, post-translational modifications, or protein-protein interactions. For instance, antibodies targeting the C-terminus might be better for detecting both membrane-associated and nuclear beta-catenin pools.

  • Clonality and production technology: Recombinant monoclonal antibodies generally offer superior batch-to-batch consistency compared to traditional hybridoma-derived antibodies . For long-term studies requiring consistent reagents, recombinant antibodies like CTNNB1/2030R provide significant advantages in reproducibility .

  • Species reactivity and cross-reactivity: Verify that the antibody recognizes beta-catenin in your species of interest . While some antibodies like CTNNB1/2030R are specific to human samples, others such as clone 5H10 recognize both human and chicken beta-catenin . Cross-reactivity should be experimentally confirmed rather than assumed based on sequence homology.

  • Specificity validation evidence: Evaluate the comprehensiveness of specificity testing provided by manufacturers . The most robust antibodies have undergone extensive validation, such as protein array testing against thousands of human proteins, genetic knockout controls, and orthogonal method confirmation .

  • Research context alignment: Match the antibody to your specific research question . For studies of Wnt signaling activation, select antibodies demonstrated to effectively detect nuclear beta-catenin accumulation . For investigations of cell adhesion, choose antibodies validated for detecting membrane-localized beta-catenin at adherens junctions .

  • Technical considerations for specific applications:

    • For IHC: Select antibodies compatible with your tissue fixation method and antigen retrieval protocol

    • For multiplexed applications: Consider host species and isotype to avoid cross-reactivity issues

    • For quantitative applications: Choose antibodies with linear signal response across a range of protein concentrations

By systematically evaluating these factors, researchers can select the optimal CTNNB1 antibody for their specific research needs, ensuring that their investigations of beta-catenin biology in development, homeostasis, and disease yield reliable and interpretable results .

How has our understanding of beta-catenin's diverse cellular functions evolved through antibody-based research?

Antibody-based research has been instrumental in transforming our understanding of beta-catenin from a simple structural protein to a multifunctional regulator of cellular processes, highlighting its critical roles in both normal physiology and disease states . This evolution in understanding has proceeded through several key phases, each enabled by increasingly sophisticated antibody applications:

  • Initial identification as a structural component (1980s-early 1990s):

    • Early antibodies revealed beta-catenin as a component of adherens junctions

    • Immunofluorescence localization demonstrated co-distribution with E-cadherin at cell-cell contacts

    • Beta-catenin was primarily viewed as a structural adaptor linking cadherins to the actin cytoskeleton

  • Dual function discovery in signaling and adhesion (mid-1990s):

    • Antibody-based studies unexpectedly revealed nuclear localization of beta-catenin

    • Immunohistochemistry demonstrated beta-catenin nuclear accumulation in colorectal cancers

    • This finding provided the crucial link between the previously separate fields of cell adhesion and Wnt signaling

    • Antibodies enabled visualization of beta-catenin's dynamic redistribution between membrane, cytoplasm, and nucleus

  • Mechanistic elucidation of Wnt signaling (late 1990s-2000s):

    • Antibodies targeting different beta-catenin domains helped map interaction interfaces with other proteins

    • Phospho-specific antibodies revealed the regulatory phosphorylation events controlling beta-catenin stability

    • Chromatin immunoprecipitation with beta-catenin antibodies identified direct target genes

    • Immunoprecipitation studies uncovered the destruction complex components (AXIN1, AXIN2, APC, GSK3B) that regulate beta-catenin

  • Tissue-specific functions and regulatory mechanisms (2010s):

    • Diverse antibody applications revealed context-dependent roles in different tissues and developmental stages

    • Antibodies enabled detection of beta-catenin's involvement in:

      • Cell fate decisions during development

      • Stem cell self-renewal in adult tissues

      • Centrosome regulation and mitotic processes

      • Insulin internalization via the CDK2/PTPN6/CTNNB1/CEACAM1 pathway

  • Complex regulatory network exploration (recent advances):

    • Antibody-based research has uncovered sophisticated mechanisms controlling beta-catenin expression

    • Studies have revealed tissue-specific enhancers like ieCtnnb1/ieCTNNB1, which fine-tune beta-catenin levels and Wnt signaling output

    • Mapping of enhancer-binding transcription factors (HNF4α, phosphorylated CREB1) has identified upstream regulators of beta-catenin expression

    • Single-cell analyses using CTNNB1 antibodies have demonstrated cell type-specific responses to Wnt pathway modulation

  • Therapeutic targeting insights (ongoing):

    • Antibody-based studies have revealed the complexity of targeting beta-catenin in disease

    • Research has shown how enhancer-dependent mechanisms can modulate Wnt signaling dosage in a tissue-specific manner

    • These findings suggest potential for tissue-selective therapeutic approaches with reduced off-target effects

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.