GLYAT antibodies are available in various formats, each optimized for specific experimental needs:
| Supplier | Type | Reactivity | Applications | Dilution | Price (USD) |
|---|---|---|---|---|---|
| Proteintech | Rabbit Polyclonal | Human, Mouse | WB, ELISA | WB: 1:500–1:1000 | ~$300 |
| Novus Biologicals | Mouse Monoclonal | Human | WB, IHC, ELISA | WB: 1:500, IHC: 1:10–1:500 | ~$478 |
| Sigma-Aldrich | Rabbit Polyclonal | Human | Immunoblotting, IHC | IB: 0.04–0.4 μg/mL | ~$598 |
| Abbexa | Rabbit Polyclonal | Human | WB, ELISA | WB: 1:500–1:2000 | ~$398 |
Polyclonal antibodies (e.g., Proteintech, Sigma-Aldrich) offer broader epitope recognition, while monoclonal antibodies (e.g., Novus Biologicals) provide higher specificity .
Reactivity varies; most antibodies target human samples, with some cross-reactivity to mouse or rat .
GLYAT antibodies are employed in diverse experimental workflows:
Western Blotting (WB): Detects GLYAT protein levels in cell lysates or tissue extracts. For example, a study in breast cancer used WB to confirm GLYAT downregulation in malignant tissues .
Immunohistochemistry (IHC): Localizes GLYAT in paraffin-embedded sections, as demonstrated in kidney adenocarcinoma samples .
ELISA: Quantifies GLYAT in biological fluids, aiding metabolic studies .
GLYAT antibodies have been instrumental in linking GLYAT expression to cancer progression:
Breast Cancer: GLYAT was found to suppress tumor growth and metastasis by inhibiting the PI3K/AKT/Snail signaling pathway. Antibodies confirmed reduced GLYAT levels in aggressive subtypes .
Liver and Kidney Cancers: Studies using GLYAT antibodies revealed its tumor-suppressive role, with downregulation correlating with poor prognosis in hepatocellular carcinoma (HCC) and clear cell renal cell carcinoma (ccRCC) .
Epithelial-Mesenchymal Transition (EMT): GLYAT antibodies demonstrated its role in maintaining epithelial markers (e.g., E-cadherin) and suppressing mesenchymal markers (e.g., vimentin) in cancer cells .
GLYAT (Glycine N-Acyltransferase) is an enzyme involved in glycine metabolism that has been implicated in the progression of various malignant tumors. Recent research has demonstrated that GLYAT is significantly downregulated in hepatocellular carcinoma (HCC) tissues compared to normal liver tissues . Its clinical relevance lies in its potential as both a diagnostic and prognostic biomarker, with decreased expression correlating with poorer outcomes in HCC patients . The significance of GLYAT extends beyond its role in metabolism, as it appears to influence tumor cell proliferation, invasion, and migration capabilities, making it a promising therapeutic target .
Multiple complementary techniques provide robust GLYAT expression analysis in research settings:
RT-qPCR: Effective for quantifying GLYAT mRNA expression levels in cell lines and tissue samples with high sensitivity
Western blotting: Recommended dilution of 1/1000 for protein detection using GLYAT antibodies
Immunohistochemistry (IHC): Optimal dilutions range from 1/10 to 1/50 for paraffin-embedded sections
Immunofluorescence (IF): Useful for cellular localization studies of GLYAT
ELISA: Provides quantitative measurement of GLYAT in solution
For validation purposes, researchers should employ at least two independent methods when characterizing GLYAT expression, as demonstrated in comprehensive studies where RT-qPCR, Western blotting, and IHC collectively provided consistent evidence of GLYAT downregulation in HCC tissues .
Research indicates distinct GLYAT expression patterns across different cellular models:
Normal liver cells: Higher GLYAT expression observed in LO2 cell line (normal human hepatic cell line)
HCC cell lines: Significantly lower GLYAT expression in Huh 7, HepG2, PLC, and SK-HEP1 compared to normal liver cells
Tissue samples: Consistent downregulation in HCC tissues compared to adjacent normal liver tissues, verified through multiple detection methods including RT-qPCR, Western blotting, and immunohistochemistry
Understanding these expression patterns is crucial for experimental design, as researchers should select appropriate cell lines based on their baseline GLYAT expression levels. For instance, Huh 7 cells have been used for knockdown experiments while HepG2 cells were utilized for overexpression studies in functional characterization of GLYAT .
GLYAT expression significantly influences the immune landscape within HCC tumors through multiple mechanisms:
Immune infiltration correlation: GLYAT expression shows positive correlation with resting mast cells, monocytes, M2 macrophages, M1 macrophages, naive CD4+ T cells, and activated NK cells, while negatively correlating with follicular helper T cells, memory B cells, resting dendritic cells, regulatory T cells (Tregs), and M0 macrophages
Immune score impact: Low GLYAT expression groups demonstrate higher immune and ESTIMATE scores compared to high GLYAT expression groups, suggesting increased immune cell infiltration in tumors with reduced GLYAT expression
NK cell activity: GLYAT expression positively correlates with activated NK cells, which are crucial for early immune responses against liver cancer. This suggests that decreased GLYAT may contribute to reduced anti-tumor immunity
Regulatory T cell relation: Low GLYAT expression associates with increased Tregs, which induce immune tolerance and suppress tumor-specific T cell activity, potentially explaining the poorer prognosis in patients with low GLYAT expression
These findings collectively suggest that GLYAT downregulation may contribute to an immunosuppressive microenvironment conducive to tumor progression, highlighting potential avenues for combined therapeutic approaches targeting both GLYAT and immune checkpoints .
The functional impact of GLYAT on HCC progression operates through several cellular mechanisms:
Epithelial-mesenchymal transition (EMT): Previous research in breast cancer demonstrated that GLYAT downregulation enhances invasion, migration, and proliferation by modulating EMT, a finding potentially relevant to HCC given EMT's crucial role in intra- and extrahepatic metastasis
Cell proliferation regulation: In vitro experiments have shown that GLYAT overexpression significantly inhibits the proliferation of hepatocellular cell lines, while knockdown enhances proliferative capacity
Invasion and migration control: GLYAT overexpression markedly reduces the invasive and migratory abilities of HCC cells, suggesting a role in metastasis suppression
Clinical correlation: The inhibitory effects of GLYAT on these malignant behaviors align with clinical observations where decreased GLYAT expression associates with higher histological grade and advanced clinical stage in HCC patients
GLYAT expression appears to have significant implications for immunotherapy response prediction:
Immune checkpoint correlation: GLYAT expression exhibits negative correlations with most immune checkpoint genes (except IDO2), suggesting that patients with low GLYAT may have higher expression of immune checkpoint molecules
Immunophenoscore analysis: HCC patients with low GLYAT expression demonstrate characteristics that predict better response to immune checkpoint inhibitors (ICIs) targeting PD-1 and CTLA-4
Therapeutic implications: These findings suggest that inhibition of GLYAT expression might potentially enhance the efficacy of ICIs, offering a promising avenue for combination therapy approaches
Patient stratification potential: GLYAT expression could potentially serve as a biomarker for identifying HCC patients most likely to benefit from immunotherapy, though this requires validation in prospective clinical trials
Researchers should consider the following application-specific conditions when using GLYAT antibodies:
Western blotting: Recommended dilution of 1/1000; researchers should verify the expected molecular weight of approximately 33.9 kDa as calculated for human GLYAT
Immunohistochemistry (paraffin sections): Optimal dilutions range from 1/10 to 1/50; researchers should establish their own optimal concentration through titration experiments
Immunofluorescence: Available with specific antibodies such as the D-12 mouse monoclonal antibody recognizing epitope 150-177 of human GLYAT
Immunoprecipitation: Several antibodies are validated for this application, including the D-12 mouse monoclonal antibody
ELISA: Multiple antibodies are suitable for ELISA applications, though optimal working concentrations should be determined experimentally
For all applications, proper controls should be included, and storage recommendations (typically aliquoting and storing at -20°C while avoiding repeated freeze/thaw cycles) should be strictly followed to maintain antibody performance .
For effective GLYAT gene manipulation in functional studies, researchers have successfully utilized several approaches:
RNA interference via shRNA: Successful GLYAT knockdown has been achieved using short hairpin RNA targeting specific sequences (e.g., 5′-GGAAACAGCATTTACAGATTC-3′) delivered via lentiviral vectors with GFP reporters
Lentiviral overexpression: For gain-of-function studies, recombinant lentivirus carrying the human GLYAT overexpression plasmid with GFP markers has been employed
CRISPR/Cas9 genome editing: GLYAT CRISPR/Cas9 knockout plasmids are available for human applications, providing alternative gene editing approaches
HDR-based modification: Homology-directed repair plasmids for GLYAT are available for precise gene editing applications
Double Nickase approach: This alternative CRISPR strategy for GLYAT modification offers reduced off-target effects compared to standard CRISPR/Cas9
Selection methods typically employ puromycin (2 μg/mL) for 48 hours post-transduction to establish stable cell lines. Validation of gene manipulation effectiveness should combine both protein-level (Western blotting) and transcript-level (RT-qPCR) assessments to confirm successful modification .
Rigorous antibody validation requires comprehensive controls:
Positive tissue controls: Normal liver tissue serves as an excellent positive control for GLYAT antibody validation due to its high endogenous expression
Negative tissue controls: Tissues known to have minimal GLYAT expression or isotype-matched controls on target tissues
Specificity validation: Compare staining patterns with multiple antibodies targeting different GLYAT epitopes; the D-12 antibody targets amino acids 150-177, while other antibodies may target different regions
Peptide competition assays: Pre-incubation of the antibody with the immunizing peptide should abolish specific staining
Genetic manipulation controls: GLYAT overexpression and knockdown samples provide excellent validation tools; comparing antibody signal between Huh 7-ctrl versus Huh 7-sh cells or HepG2-ctrl versus HepG2-oe cells can confirm specificity
Cross-reactivity assessment: Particularly important when studying related family members like GLYATL1, researchers should verify that the antibody does not cross-react with GLYATL1, which has been studied using specific antibodies like the B-12 mouse monoclonal that recognizes epitope 265-279 of human GLYATL1
These validation approaches ensure experimental reproducibility and reliable interpretation of results when working with GLYAT antibodies.
For accurate quantification of GLYAT expression in clinical samples, researchers should consider:
For prognostic applications, Kaplan-Meier analyses with appropriate cutoff values for GLYAT expression levels should be employed, as these have successfully demonstrated the correlation between decreased GLYAT expression and poorer progress in HCC .
Some findings regarding GLYAT expression in HCC present apparent contradictions that researchers must address:
Age correlation paradox: Higher GLYAT expression has been observed in patients over 65 years of age despite this demographic generally having poorer prognosis in HCC
Gender correlation inconsistency: Lower GLYAT expression has been reported in women despite the lower incidence of HCC in females
Prognostic variability: Individual variability in HCC prognosis necessitates the development of genomic clinicopathological prognostic models
To address these contradictions, researchers should:
Perform multivariate analyses that account for confounding factors
Stratify patient cohorts by additional clinical parameters beyond age and gender
Develop nomograms that integrate GLYAT expression with other independent risk factors (such as T stage) to improve prognostic accuracy
Conduct larger cohort studies with diverse patient populations to validate the robustness of GLYAT as a biomarker
Investigate potential biological explanations for these paradoxical findings, such as hormone-dependent regulation of GLYAT expression
These approaches will help clarify whether these contradictions represent true biological phenomena or artifacts of study design.
To advance understanding of GLYAT's role in immunotherapy response, researchers should consider:
Co-culture systems: Develop in vitro co-culture models of HCC cells (with varied GLYAT expression) and immune cells to study direct interactions and mechanisms
Immune checkpoint blockade models: Evaluate the effect of GLYAT manipulation on response to anti-PD-1 or anti-CTLA-4 treatments in preclinical models
Single-cell RNA sequencing: Apply this technology to characterize cell-specific effects of GLYAT expression on diverse immune cell populations within the tumor microenvironment
Spatial transcriptomics: Investigate the spatial relationship between GLYAT-expressing cells and immune cell infiltrates in HCC tissue sections
Patient-derived xenograft (PDX) models: Establish PDX models from patients with varying GLYAT expression levels to test immunotherapy response in vivo
Clinical sample analysis: Retrospectively analyze GLYAT expression in samples from HCC patients who have received immunotherapy to correlate expression with treatment outcomes
These approaches would help validate the predictive value of GLYAT for immunotherapy response and potentially identify mechanisms by which GLYAT influences the tumor immune microenvironment.
GLYAT's primary function in glycine metabolism suggests potential metabolic roles in HCC that require specific experimental approaches:
Metabolomic profiling: Compare metabolite profiles between HCC cells with different GLYAT expression levels to identify altered metabolic pathways
Isotope tracing experiments: Use labeled glycine to track changes in metabolic flux dependent on GLYAT expression
Integration with genomic data: Correlate GLYAT expression with expression of other metabolic enzymes to identify potential compensatory mechanisms
Functional metabolic assays: Measure parameters such as oxygen consumption rate, extracellular acidification rate, and ATP production in cells with modified GLYAT expression
In vivo metabolic imaging: Apply techniques such as hyperpolarized MRI to assess metabolic alterations in animal models with varied GLYAT expression
Understanding GLYAT's metabolic functions may provide additional insights into its role in HCC progression and potentially identify metabolic vulnerabilities that could be therapeutically targeted. The integration of metabolomics with functional studies of GLYAT represents an important frontier in understanding its comprehensive role in liver cancer biology.