CTAG1A Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze / thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days of receiving it. Delivery times may vary depending on the method of purchase and location. Please consult your local distributors for specific delivery information.
Synonyms
CTAG1A antibody; CTAG1B antibody; Autoimmunogenic cancer/testis antigen NY ESO 1 antibody; Autoimmunogenic cancer/testis antigen NY-ESO-1 antibody; Cancer antigen 3 antibody; Cancer/testis antigen 1 antibody; Cancer/testis antigen 1B antibody; Cancer/testis antigen 6.1 antibody; CT6.1 antibody; CTAG 1 antibody; CTAG 1B antibody; CTAG antibody; CTAG1 antibody; CTAG1B antibody; CTG1B_HUMAN antibody; ESO 1 antibody; ESO1 antibody; L antigen family member 2 antibody; LAGE 2 antibody; LAGE 2 protein antibody; LAGE 2B antibody; LAGE-2 antibody; LAGE2 antibody; LAGE2 protein antibody; LAGE2A antibody; LAGE2B antibody; New York esophageal squamous cell carcinoma 1 antibody; NY ESO 1 antibody; NYESO 1 antibody; NYESO1 antibody
Target Names
Uniprot No.

Target Background

Gene References Into Functions
  1. NY-ESO-1 protein expression was observed in 28 out of 38 cancer specimens. 2/38 showed strong universal (4+) NY-ESO-1 staining, and 9/40 cancer lesions exhibited strong (4+) staining for survivin. PMID: 29058035
  2. A significant association was found between AKAP4 gene expression and metastasis (P-value: 0.045). However, expression of the CTAG1B (NY-ESO-1) gene was not observed in our cases. PMID: 29480665
  3. Among 22 melanoma patients with stage III lymph node metastasis, overall survival was significantly higher in the XAGE-1b and NY-ESO-1 double-negative group compared to other groups. PMID: 28105694
  4. Our findings indicate a strong humoral immune response against NY-ESO-1 in natural human T-cell leukemia virus type 1 infection, regardless of the clinical status. PMID: 28716148
  5. Certain autoantibodies, such as anti-MAGEA4, anti-CTAG1 or anti-TP53, and their combinations, could potentially contribute to the development of early cancer detection tests (not necessarily limited to gastric cancer) when combined with other markers. PMID: 27140836
  6. High NY-ESO-1 expression is associated with Lung Cancer. PMID: 27793776
  7. Our results support the potential utility of NY-ESO-1, PRAME, and MAGEA4 as targets for immunotherapy and as ancillary prognostic parameters in synovial sarcomas. PMID: 27993576
  8. Data suggests that only a small fraction of HLA-A*02:01- (HA)-binding ESO peptides are immunogenic, specifically those with high peptide-binding strength and peptide/HA complex stability. This study compared in silico-predicted and observed cytotoxic T-lymphocyte recognition of tumor antigen epitopes in melanoma patients and transgenic/knockout mice. (ESO = tumor antigen NY-ESO-1) PMID: 28536262
  9. These data demonstrate that MAGE-A1-, MAGE-A3-, and NY-ESO-1-specific T cells with antigen-specific cytotoxicity can be cultured from healthy donors and patient-derived cells, making adoptive immunotherapy with these cytotoxic T lymphocyte feasible. PMID: 28677424
  10. Comparing the overall expression of CTAs, a decreased expression of all melanoma-associated antigens (MAGEs) post-treatment and a slightly increased expression of New York esophageal squamous cell carcinoma 1 (NY-ESO-1) was observed. The simultaneous cytoplasmic and nuclear expression of pan-MAGE or MAGE-A3/A4 correlated with reduced treatment-failure-free-survival (TFFS). PMID: 27466502
  11. MAGE-A is more highly expressed than NY-ESO-1 in a majority of human malignancies. PMID: 27070449
  12. These cells were used to target a human lung cancer line that expressed NY-ESO-1. PMID: 26324743
  13. The regulation of NY-ESO-1 processing by the ubiquitin receptors Rpn10 and Rpn13, as well as by the standard and immunoproteasome, is governed by non-canonical ubiquitination on non-lysine sites. PMID: 26903513
  14. CTAs (MAGE-A4, NY-ESO-1, MAGE-A10) were more likely expressed in patients with squamous cell carcinoma of the lung, and when CTAs were combined with CD133, they served as better prognostic factors. PMID: 26191258
  15. Among mesenchymal tumors, myxoid liposarcomas showed the highest positivity for NY-ESO-1 (88%), followed by synovial sarcomas (49%), myxofibrosarcomas (35%), and conventional chondrosarcomas (28%). PMID: 25412843
  16. High expression of NY-ESO-1 is associated with Triple-Negative Breast Cancer. PMID: 26413775
  17. NY-ESO-1 expression in melanoma was associated with tumor progression, including increased tumor thickness, and with reduced tumor infiltrating lymphocytes. PMID: 25954764
  18. NY-ESO-1 is expressed in esophageal adenocarcinomas, Barrett's metaplasia and normal tissues other than germ cells. PMID: 24744590
  19. NY-ESO-1 cancer antigen expression plays a role in immunotherapy in thyroid cancer. PMID: 24811699
  20. Primary autoantibodies against intracellular MM-specific tumor antigens SSX-2 and NY-ESO-1 are rare but functional in multiple myeloma patients after allogeneic stem cell transplantation. PMID: 25078248
  21. We have also demonstrated that NY-ESO-1 expression may lead to a humoral immune response in patients with meningioma. PMID: 24777967
  22. NY-ESO-1 tetramer(+) cells were detected concomitantly with high proportions of Treg but were distinct from the latter and displayed characteristics of TH1 effectors. PMID: 24777968
  23. Neck squamous cell carcinoma patients showing protein expression of MAGE-A family members or NY-ESO-1 represent a subgroup with an extraordinarily poor survival. PMID: 24482145
  24. Our observations indicate a strong link between NY-ESO-1 expression and ERG activation. PMID: 24789172
  25. NY-ESO-1 and SP17 were not significantly associated with a specific histotype, but high-level GAGE expression was more frequent in squamous cell carcinoma. GAGE expression was demonstrated to be significantly higher in stage II-IIIa than stage I NSCLC. PMID: 24103781
  26. NY-ESO-1 appears to be a sensitive and specific marker for myxoid and round cell liposarcoma among mesenchymal myxoid neoplasms. PMID: 23599152
  27. Positive results of immunohistostaining were obtained in 16 (35.6%), 7 (15.6%) and 36 (80.0%) samples using MAGE-C1, NY-ESO-1 and Sp17 antibodies, respectively. PMID: 23923079
  28. This study analyzed NY-ESO-1 expression in 222 melanoma specimens including 16 primary and 206 metastatic tumors. Results support previous findings showing higher expression of NY-ESO-1 in metastatic (58/206) versus primary (0/16) tumors. Results also show that the epithelioid subtype of melanoma has the highest incidence of NY-ESO-1 expression. PMID: 24290058
  29. In two non-epithelial cancers (glioma and mesothelioma), the epigenetic regulation of the NY-ESO-1 gene requires the sequential recruitment of the HDAC1-mSin3a-NCOR, Dnmt3b-HDAC1-Egr1 and Dnmt1-PCNA-UHRF1-G9a complexes. PMID: 23312906
  30. Melanoma patients' humoral immune systems responded to NY-ESO-1 differently in each individual. PMID: 23454162
  31. CTAG1B mRNA and protein are overexpressed with high frequency in myxoid and round cell liposarcoma. PMID: 22936067
  32. High CTAG1 expression and down-regulation of HLA class-I is associated with non-small cell lung cancer. PMID: 23645764
  33. NY-ESO-1 is strongly and diffusely expressed in a majority of synovial sarcomas, but only rarely in other mesenchymal lesions. This suggests roles in targeted therapy and differential diagnosis. PMID: 22388761
  34. The presence of circulating T cells responding to Melan-A or NY-ESO-1 had a strong independent prognostic impact on survival in advanced melanoma. PMID: 22529253
  35. Cancer/testis antigens are novel targets of immunotherapy for adult T-cell leukemia/lymphoma. PMID: 22323448
  36. Polymeric structure and TLR4 may play important roles in rendering NY-ESO-1 immunogenic and thus serve as a potent molecular adjuvant. NY-ESO-1 thus represents the first example of a cancer/testis antigen. PMID: 21900253
  37. Integrated NY-ESO-1 immune responses may have predictive value for ipilimumab treatment in patients with advanced metastatic melanoma. PMID: 21933959
  38. Primary tumors with and without lymph node metastases showed no significant differences in MAGE-A 3/4 (P=0.672) and NY-ESO-1 (P=0.444) expression. PMID: 21556122
  39. LAGE-1a and NY-ESO-1 homology cannot be easily exploited in an anti-NY-ESO-1 vaccine given the low frequency of protein expression detected by IHC or serum analysis. PMID: 21247062
  40. This report describes immunohistochemical expression of NY-ESO-1 in renal oncocytoma and chromophobe renal cell carcinoma. PMID: 20591578
  41. Most melanoma patients with spontaneous NY-ESO-1-specific responses in this study exhibit spontaneous CD4-positive T cell responses to at least one of the three immunodominant LAGE-1 epitopes. PMID: 21131422
  42. A versatile prime-boost vaccine strategy allows the generation of powerful, high-avidity tumor-associated immunodominant NY-ESO-1-transgene specific CD8-positive cytotoxic T cell responses. PMID: 20733200
  43. Tumor antigen NY-ESO-1 plays a role in the immune responses to tumor and self-antigens. PMID: 20368442
  44. ESO 9V peptide isoform is more efficient in inducing conjugate formation and cytolytic granule polarization than the ESO 9L isoform. PMID: 20053942
  45. MAGE-A3/6 and NY-ESO-1 were expressed in 50.0% (66/132) and 18.2% (24/132) of non-small-cell lung carcinomas, respectively. PMID: 19795170
  46. High NY-ESO-1 expression is associated with oral squamous cell carcinoma. PMID: 20044626
  47. Postvaccine T-cell clones are shown to recognize and lyse NY-ESO-1 expressing tumor cell lines in vitro. PMID: 19728336
  48. NY-ESO-1 119-143 is a promiscuous major histocompatibility complex class II T-helper epitope recognized by Th1- and Th2-type tumor-reactive CD4+ T cells. PMID: 11782380
  49. NY-ESO-1 is a marker that can be used to follow the early progression of testicular tumorigenesis when the tumors express a similar pattern to the cells of origin, although later tumors cease to express NY-ESO-1. PMID: 12065688
  50. This study investigated the abilities of human monocyte-derived DCs and DCs derived in vitro from CD34-positive stem cells to present NY-ESO-1 epitopes to MHC class I-restricted cytotoxic T cells. PMID: 12138174

Show More

Hide All

Database Links

HGNC: 24198

OMIM: 300156

KEGG: hsa:1485

STRING: 9606.ENSP00000332602

UniGene: Hs.534310

Protein Families
CTAG/PCC1 family
Subcellular Location
Cytoplasm.
Tissue Specificity
Expressed in testis and ovary and in a wide variety of cancers. Detected in uterine myometrium. Expressed from 18 weeks until birth in human fetal testis. In the adult testis, is strongly expressed in spermatogonia and in primary spermatocytes, but not in

Q&A

What is CTAG1A and why is it significant in cancer research?

CTAG1A (Cancer/Testis Antigen 1A) is a cancer/testis antigen encoded by the CTAG1A gene located on the X-chromosome. It is identical in sequence to CTAG1B, which is located approximately 30,000 base pairs away on the same chromosome . CTAs like CTAG1A are particularly attractive targets for immunotherapy because they have minimal or no expression in normal tissues, except for germline tissues, but are aberrantly expressed in various human cancers . CTAG1A is significant in cancer research because it can be recognized by cytotoxic T lymphocytes (CTLs), making it a potential target for adaptive immunotherapy approaches . High expression of CTAG1A/B has been observed in multiple cancer types, including lung cancer, where expression frequencies can reach up to 68.4% of cases .

How does CTAG1A expression differ between normal and cancerous tissues?

CTAG1A expression in normal conditions is tightly restricted to adult testis germ cell differentiation . In cancerous tissues, this gene becomes re-activated through various mechanisms, primarily epigenetic modifications. Research indicates that CTAG1A expression is silenced in normal tissues through promoter hypermethylation, and this restriction can be lifted in cancer cells . Specific cancer types showing high CTAG1A/B expression include synovial sarcomas, myxoid liposarcomas, lung cancers, and glioblastomas . The re-activation of this gene in cancer has been linked to its potential role in oncogenesis, making it both a biomarker for cancer and a potential therapeutic target .

What laboratory techniques are commonly used to detect CTAG1A expression?

Several complementary laboratory techniques can be employed to detect CTAG1A expression in research settings:

  • RT-PCR and Real-time PCR: These methods are used to detect CTAG1A at the mRNA level. Research protocols typically involve RNA extraction from tissue samples, followed by cDNA synthesis using reverse transcriptase. For endpoint RT-PCR, gene-specific primers for CTAG1A are used with appropriate annealing temperatures (similar to the 67°C used for CT83 in related studies) . Products can then be visualized by ethidium bromide staining and ultraviolet light exposure after electrophoresis on agarose gels .

  • Immunohistochemistry (IHC): This technique allows for the visualization of CTAG1A protein expression in tissue sections, enabling researchers to determine its cellular and subcellular localization. IHC staining of separate, independent samples has been used to confirm the expression of CTAG1A and related proteins identified through network analysis approaches .

  • Western Blotting: This method provides quantitative information about CTAG1A protein expression using specific antibodies against the target protein.

  • Microarray Analysis: Gene expression data from microarray studies can identify differential expression of CTAG1A between normal and tumor tissues, as demonstrated in various datasets available through repositories like the Gene Expression Omnibus (GEO) at NCBI .

What are the challenges in producing specific antibodies against CTAG1A?

Producing specific antibodies against CTAG1A presents several challenges:

  • Sequence similarity with CTAG1B: Since CTAG1A and CTAG1B are identical in sequence but located at different genomic positions, developing antibodies that can distinguish between these two proteins is virtually impossible . Most commercial antibodies detect both CTAG1A and CTAG1B, commonly referred to as CTAG1A/B antibodies.

  • Cross-reactivity with other CTAs: The cancer/testis antigen family shares some structural similarities, requiring extensive validation to ensure antibody specificity against CTAG1A without cross-reacting with other CTA family members.

  • Low expression levels in some samples: CTAG1A expression can vary significantly between cancer types and even within the same cancer type, making detection challenging when expression levels are low.

  • Post-translational modifications: These modifications can affect antibody binding, necessitating the development of antibodies that recognize specific forms of the protein relevant to the research question.

To overcome these challenges, researchers should validate antibodies using positive controls (tissues known to express CTAG1A, such as testis or specific cancer cell lines) and negative controls (normal tissues that do not express CTAG1A).

How do epigenetic mechanisms regulate CTAG1A expression, and how can this be manipulated experimentally?

CTAG1A expression is primarily regulated through epigenetic mechanisms, particularly DNA methylation. In normal tissues and some cancer cells, CTAG1A promoter hypermethylation restricts its expression . This epigenetic silencing can be experimentally manipulated through demethylating agents:

Experimental approach for epigenetic induction of CTAG1A:

  • DNA methyltransferase inhibitors: Decitabine (DAC) has been shown to induce sufficient expression of cancer-testis antigens, including CTAG1A/NY-ESO-1, in glioblastoma for targeting by adoptive T-cell therapy . Treatment protocols typically involve culturing cancer cells with clinically relevant concentrations of DAC over multiple days.

  • Methylation analysis: To confirm the mechanism of action, researchers can perform bisulfite sequencing or methylation-specific PCR of the CTAG1A promoter region before and after treatment with demethylating agents.

  • Single-cell resolution analysis: Advanced techniques have demonstrated that DAC treatment can render CTAG1A into inducible tumor antigens at single-cell resolution, which is critical for understanding heterogeneity in cancer cell populations .

  • Functional validation: The biological significance of CTAG1A re-expression can be assessed by examining effects on cell proliferation, migration, invasion, or susceptibility to immune targeting.

What are the current methodologies for integrating CTAG1A antibodies in immunotherapeutic approaches?

Several methodologies have been developed to integrate CTAG1A antibodies in immunotherapeutic approaches:

  • Antibody-dependent cellular cytotoxicity (ADCC): CTAG1A antibodies can be engineered to engage immune effector cells through their Fc regions, leading to targeted killing of CTAG1A-expressing cancer cells.

  • Bispecific antibodies: These antibodies simultaneously bind to CTAG1A on cancer cells and to immune cells (typically T cells), bringing them into proximity to promote immune-mediated tumor destruction.

  • Antibody-drug conjugates (ADCs): CTAG1A antibodies can be conjugated to cytotoxic agents, delivering these therapeutic payloads specifically to CTAG1A-expressing cancer cells.

  • Combination with adoptive T-cell therapy: Research has shown that NY-ESO-1 (CTAG1A) T-cell receptor–engineered effector cells can target DAC-induced antigen in primary glioma cells, promoting specific and polyfunctional T-cell cytokine profiles . The methodological approach involves:

    • Epigenetic priming of cancer cells with demethylating agents to induce CTAG1A expression

    • Engineering T cells with receptors specific for CTAG1A

    • Co-culture experiments to assess T-cell activation, cytokine production, and target cell killing

This combined approach has demonstrated enhanced T-cell functionality against glioblastoma, potentially improving targeted immune therapies in these difficult-to-treat cancers .

How can CTAG1A be mapped to functional pathways through network reconstruction?

Mapping CTAG1A to functional pathways through network reconstruction involves several sophisticated methodological steps:

  • Gene co-expression module extraction: Researchers can extract CTAG1A co-expression gene modules from cancer samples (such as synovial and myxoid sarcomas) using the following approach :

    • Establish differential expression of CTAG1A between different classes of samples (e.g., normal vs. metastatic tissue, benign vs. primary tumor)

    • Apply statistical tests (Student's t-test or Mann-Whitney's non-parametric test) to identify significant differences in expression levels

    • Derive gene modules that are co-expressed either positively or negatively with CTAG1A using gene set enrichment analysis (GSEA)

  • Interaction data mining: First and second level interactors of the CTAG1A/B protein can be extracted from comprehensive databases like PICKLE, which integrates data from multiple interaction databases . Data should be filtered for confirmed physical/functional interactions, with appropriate threshold values (e.g., E value > 0.7) to minimize false positives.

  • Network analysis: The gene lists obtained from co-expression analysis can be subjected to network analysis to identify CTAG1A network neighbors and key hubs linked with shortest paths, using algorithms like cytoHubba in Cytoscape .

  • Experimental validation: The expression of identified network neighbors can be verified in independent samples using techniques like immunohistochemical staining, as demonstrated with genes like TLE1 and RANBP2 in sarcoma samples .

This approach has successfully identified CTAG1A/B as an ortholog of the yeast/Drosophila transcription factor Pcc1p and a member of the KEOPS transcription complex, implicating it in telomere maintenance and transcriptional regulation through association with chromatin remodeling factors .

What are the technical considerations for optimizing immunohistochemistry protocols using CTAG1A antibodies?

Optimizing immunohistochemistry (IHC) protocols for CTAG1A antibodies requires attention to several technical considerations:

  • Fixation and antigen retrieval: CTAG1A epitopes can be sensitive to fixation methods. Formalin-fixed, paraffin-embedded (FFPE) tissues typically require heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) to unmask antigenic sites that may be cross-linked during fixation.

  • Antibody validation: Due to the sequence identity between CTAG1A and CTAG1B, validation is crucial . Recommended validation steps include:

    • Positive controls: Testicular tissue (known to express CTAG1A) or cancer cell lines with confirmed CTAG1A expression

    • Negative controls: Normal adult tissues (except testis) which should not express CTAG1A

    • Correlation with RT-PCR results on the same samples to confirm specificity

  • Signal amplification systems: For cases with low CTAG1A expression, polymer-based detection systems or tyramide signal amplification can enhance sensitivity without increasing background staining.

  • Multiplexing strategies: To study the relationship between CTAG1A expression and other markers (such as immune cell infiltration or MHC-I expression), researchers can employ multiplexed IHC using sequential staining protocols or spectral imaging systems.

  • Quantification methods: Digital pathology platforms can be used for objective quantification of CTAG1A expression, including:

    • H-score calculation (combining intensity and percentage of positive cells)

    • Determination of subcellular localization patterns

    • Correlation with clinical parameters or other molecular markers

Optimized IHC protocols have been instrumental in confirming the expression of CTAG1A and related proteins identified through network analysis approaches in cancer tissues .

How can researchers address data inconsistencies when CTAG1A antibody results conflict with genomic or transcriptomic data?

When facing inconsistencies between CTAG1A antibody results and genomic or transcriptomic data, researchers should implement a systematic troubleshooting approach:

  • Technical verification:

    • Antibody validation: Confirm antibody specificity using positive and negative controls, and consider using multiple antibodies targeting different epitopes.

    • Primer specificity: For transcriptomic data, verify primer specificity for CTAG1A, especially given its sequence identity with CTAG1B .

    • Sample quality: Assess RNA integrity for transcriptomic data and protein quality for antibody-based methods.

  • Biological explanations:

    • Post-transcriptional regulation: Discrepancies between mRNA and protein levels may reflect post-transcriptional regulatory mechanisms.

    • Protein stability: CTAG1A protein might have different half-lives in various cellular contexts.

    • Tumor heterogeneity: Expression may vary within different regions of the same tumor, leading to sampling bias.

  • Methodological approaches to reconcile discrepancies:

    • Single-cell analysis: Techniques like single-cell RNA-seq paired with antibody-based protein detection can reveal cell-to-cell variability and correlate mRNA with protein levels at the single-cell level.

    • Laser capture microdissection: This approach can isolate specific cell populations for more precise molecular analysis.

    • Temporal studies: Examining expression at different time points may reveal dynamic regulation patterns.

Research has shown that CTAG1A/B expression frequency can vary significantly depending on the detection method. For example, in lung cancer studies, CTAG1A/B expression frequency was found to be 68.4%, which was higher than that reported in The Cancer Genome Atlas (TCGA) database . Understanding these discrepancies is essential for accurate interpretation of research findings and their clinical implications.

How can CTAG1A antibodies be used to study the correlation between CTAG1A expression and disease progression?

CTAG1A antibodies can be utilized in several methodological approaches to investigate the correlation between CTAG1A expression and disease progression:

  • Tissue microarray (TMA) analysis: This high-throughput approach allows for the simultaneous evaluation of CTAG1A expression across multiple patient samples representing different disease stages:

    • Primary tumors vs. metastatic lesions

    • Early-stage vs. advanced-stage disease

    • Pre-treatment vs. post-treatment samples

  • Longitudinal liquid biopsy studies: Detection of circulating CTAG1A protein or antibodies against CTAG1A in patient serum at different time points can provide insights into disease dynamics without requiring repeated tissue biopsies.

  • Multiplexed imaging: Co-staining for CTAG1A alongside markers of proliferation (Ki-67), invasion (MMPs), or cancer stem cells can reveal associations between CTAG1A expression and aggressive phenotypes.

  • Correlation with clinicopathological parameters: Statistical analysis can be performed to correlate CTAG1A expression levels with:

    • Tumor grade and stage

    • Patient survival outcomes

    • Response to specific therapeutic interventions

    • Recurrence patterns

Research has demonstrated correlations between cancer/testis antigen expression and clinical features. For example, studies of KK-LC-1 (another cancer/testis antigen) have shown its expression in early stages of gastric cancer, suggesting that CTA expression occurs at the beginning of malignancy and is subsequently maintained . Similar methodological approaches can be applied to study CTAG1A in various cancer types.

What are the protocols for using CTAG1A antibodies in flow cytometry for cancer diagnostics and research?

Using CTAG1A antibodies in flow cytometry requires specific protocols to overcome technical challenges associated with this primarily intracellular antigen:

Sample preparation protocol:

  • Cell isolation: For fresh tissue samples, prepare single-cell suspensions using enzymatic digestion (collagenase/DNase cocktail) followed by mechanical dissociation and filtration through cell strainers.

  • Fixation and permeabilization: Since CTAG1A is predominantly an intracellular antigen:

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

    • Permeabilize with 0.1% Triton X-100 or commercial permeabilization buffers designed for nuclear antigens

    • Alternative protocols using methanol-based fixation/permeabilization may provide better detection for some antibody clones

  • Blocking and antibody staining:

    • Block non-specific binding with 5% normal serum from the same species as the secondary antibody

    • Incubate with validated CTAG1A primary antibody at optimized concentration

    • For direct detection, use fluorochrome-conjugated anti-CTAG1A antibodies

    • For indirect detection, follow with fluorochrome-conjugated secondary antibodies

  • Multiparameter panel design:

    • Include markers for cell identification (e.g., EpCAM for epithelial cells)

    • Add markers for cell proliferation or functional states (Ki-67, cleaved caspase-3)

    • Consider including MHC-I markers to assess potential for T-cell recognition

  • Data analysis considerations:

    • Use appropriate isotype controls and fluorescence-minus-one (FMO) controls

    • Establish positive thresholds based on known positive and negative control samples

    • Quantify both percentage of positive cells and mean fluorescence intensity (MFI)

Flow cytometry allows for the quantitative assessment of CTAG1A expression at the single-cell level, enabling researchers to identify heterogeneity within tumor cell populations and correlate CTAG1A expression with other cellular characteristics .

How can systems-level approaches integrate CTAG1A antibody data with other -omics datasets?

Systems-level integration of CTAG1A antibody data with other -omics datasets provides a comprehensive understanding of its biological context and functional significance:

  • Multi-omics data integration strategy:

    • Correlate CTAG1A protein expression (from antibody-based methods) with transcriptomic data to identify concordant or discordant regulation

    • Integrate with epigenomic data (DNA methylation, histone modifications) to elucidate regulatory mechanisms

    • Associate with genomic data to identify genetic alterations that may influence CTAG1A expression

    • Connect with proteomic data to map CTAG1A-interacting partners

  • Network reconstruction methodology:

    • Extract CTAG1A co-expression modules from relevant cancer datasets

    • Mine confirmed physical/functional interactions from comprehensive databases

    • Apply gene set enrichment analysis (GSEA) to identify pathway associations

    • Use algorithms like cytoHubba to identify key network hubs and shortest paths

  • Experimental validation of computational predictions:

    • Verify expression of identified network neighbors using independent antibody-based methods

    • Perform functional studies to validate predicted interactions or regulatory relationships

    • Assess clinical relevance by examining correlations with patient outcomes

This systems-level approach has been successfully demonstrated in studies mapping CTAG1B/A to sarcoma transcription pathways, where researchers identified and validated the expression of at least two network neighbors, RANBP2 and TLE1 . Such comprehensive analyses provide insights into the broader biological context of CTAG1A function in cancer and can guide the development of more effective targeted therapies.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.