GLT8D1 Antibody

Shipped with Ice Packs
In Stock

Description

Key Applications of GLT8D1 Antibodies in Research

ApplicationMethodKey FindingsReference
Protein LocalizationImmunofluorescenceWT GLT8D1 localizes to Golgi; I290M/R92C mutants mislocalize to ER
Expression AnalysisWestern BlotGLT8D1 overexpression in glioma stem cells (GSCs) correlates with poor prognosis
Functional StudiesGlycosyltransferase AssayPathogenic mutants (e.g., R92C) show reduced enzymatic activity
Disease ModelingImmunohistochemistryElevated GLT8D1 in gastric cancer (GC) tissues linked to advanced TNM stages

Neurodegenerative Diseases

  • ALS Association: GLT8D1 mutations (e.g., R92C, I290M) were identified in familial amyotrophic lateral sclerosis (ALS) cases. Antibodies confirmed mutant proteins induce ER stress and cytotoxicity .

  • Mechanistic Insights: In HEK293T and N2A cells, anti-FLAG antibodies revealed mutant GLT8D1 increases lactate dehydrogenase (LDH) release by 40% compared to wild-type, indicating cytotoxicity .

Cancer Biology

  • Glioma Progression: GLT8D1 knockdown via shRNA reduced CD133+ glioma stem cell proliferation by 60% and tumor size in xenograft models, validated using GLT8D1-targeted antibodies .

  • Gastric Cancer (GC): GLT8D1 overexpression in GC tissues (confirmed by IHC) correlated with advanced T/N stages (p < 0.01) and poor survival (median OS: 372 vs. 452 days) .

Technical Considerations for GLT8D1 Antibody Use

  • Validation: Antibodies like PA5-112978 and HPA010588 are validated against 364 human recombinant proteins to ensure specificity .

  • Storage: Most antibodies require storage at −20°C in stabilizing buffers to retain activity .

  • Experimental Controls: Include wild-type and knockout cell lines to confirm signal specificity in Western blot or IHC .

Emerging Therapeutic Implications

GLT8D1 antibodies are pivotal in exploring therapeutic strategies:

  • ALS: Targeting ER stress pathways exacerbated by GLT8D1 mutations .

  • Cancer: Inhibiting GLT8D1-CD133 interactions in gliomas reduces Wnt/β-catenin signaling, a pathway critical for tumor stemness .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Description

The CSB-PA715010LA01HU product is a polyclonal antibody generated in rabbits using the recombinant human GLT8D1 protein (amino acids 33-332) as the immunogen. This antibody is an unconjugated IgG and exhibits high specificity for the GLT8D1 protein. GLT8D1 plays a crucial role in the nervous system by regulating the proliferation, self-renewal, and differentiation of neural stem cells (NSCs). This anti-GLT8D1 antibody has been rigorously tested and validated for use in various research applications, including ELISA, Western blotting, immunohistochemistry, and immunofluorescence. Its purity exceeds 95% through protein G purification, and it demonstrates reactivity with both human and mouse samples.

Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the chosen shipping method and destination. Please consult your local distributors for specific delivery timelines.
Synonyms
Da2 24 antibody; GALA4A antibody; GL8D1_HUMAN antibody; glt8d1 antibody; Glycosyltransferase 8 domain containing 1 antibody; Glycosyltransferase 8 domain-containing protein 1 antibody; Glycosyltransferase AD 017 antibody; MGC94018 antibody; MSTP139 antibody
Target Names
GLT8D1
Uniprot No.

Target Background

Gene References Into Functions
  1. ALMS1, GLT8D1, and CSNK2B have been identified as risk genes associated with schizophrenia. PMID: 29483533
  2. The single nucleotide polymorphism rs6976 may influence the susceptibility to hip osteoarthritis by altering the shape of the proximal femur. PMID: 25939412
  3. Research suggests an association between a polymorphism (rs2535629) in the ITIH3 gene and psychiatric disorders in Asian populations. This polymorphism is thought to influence the susceptibility to these disorders by affecting the expression levels of GLT8D1. PMID: 24373612
  4. Studies have revealed that GLT8D1 exhibits differential methylation and expression patterns in human squamous cell carcinomas. PMID: 22461910
Database Links

HGNC: 24870

KEGG: hsa:55830

STRING: 9606.ENSP00000266014

UniGene: Hs.297304

Protein Families
Glycosyltransferase 8 family
Subcellular Location
Membrane; Single-pass type II membrane protein.

Q&A

What is GLT8D1 and why is it significant in research?

GLT8D1 is a glycosyltransferase enzyme containing a conserved glycosyltransferase 8 domain. Its significance in research has grown substantially following the discovery of its association with amyotrophic lateral sclerosis (ALS) and its role in cancer progression, particularly in gliomas. Mutations in GLT8D1, especially those affecting the glycosyltransferase domain, have been linked to familial ALS, while its hypoxia-induced expression correlates with higher-grade gliomas and worse clinical outcomes . When detecting GLT8D1 via antibodies, researchers should be aware of its normal subcellular localization in the Golgi apparatus, though certain mutations can cause mislocalization to other cellular compartments such as the endoplasmic reticulum .

How should GLT8D1 antibodies be validated before experimental use?

Validation of GLT8D1 antibodies should include:

  • Specificity testing using both positive controls (tissues or cells known to express GLT8D1) and negative controls (knockout/knockdown cells)

  • Western blot analysis to confirm detection of the correct molecular weight protein (~42 kDa for human GLT8D1)

  • Immunoprecipitation followed by mass spectrometry to confirm antibody target specificity

  • Cross-validation with multiple antibodies targeting different epitopes of GLT8D1

  • Testing antibody performance in relevant applications (Western blot, immunofluorescence, immunohistochemistry)

Research reveals that GLT8D1 antibodies should detect discrete punctate perinuclear localization in wild-type cells, consistent with Golgi apparatus localization . Validation experiments should include controls to demonstrate that this characteristic staining pattern is observed in appropriate cell types.

What are the optimal fixation conditions for immunocytochemistry with GLT8D1 antibodies?

For immunocytochemistry applications with GLT8D1 antibodies, optimal fixation typically involves:

  • Fixation with 4% paraformaldehyde for 15-20 minutes at room temperature

  • Permeabilization with 0.2% Tween-20 to allow antibody access to intracellular compartments

  • Blocking with 1% BSA to reduce non-specific binding

  • Incubation with primary GLT8D1 antibody at optimal dilution (typically 1:100 to 1:500, but should be empirically determined)

  • Visualization with fluorescently-conjugated secondary antibodies

These conditions have been successfully employed in studies examining GLT8D1 localization, particularly when investigating the distinct patterns between wild-type and mutant GLT8D1 proteins . Co-staining with organelle markers (particularly Golgi markers) can provide valuable context for GLT8D1 localization.

How can GLT8D1 antibodies be employed to distinguish between wild-type and mutant protein in ALS research?

Differential detection of wild-type versus mutant GLT8D1 requires:

  • Using antibodies that can recognize both wild-type and mutant forms for initial detection

  • Employing immunofluorescence microscopy to observe subcellular localization patterns:

    • Wild-type GLT8D1 displays discrete punctate perinuclear localization (Golgi apparatus)

    • Mutant forms (e.g., I290M) show abnormal reticular pattern throughout the cytoplasm (ER mislocalization)

  • Combining with organelle markers for Golgi and ER to confirm localization differences

  • Western blot analysis to detect differences in steady-state expression levels (for example, I290M variant shows increased expression compared to wild-type and R92C)

Research has demonstrated that mutation-specific effects can be observed through these techniques, with the I290M mutation causing dramatic mislocalization and increased expression levels compared to wild-type protein .

What are the most effective approaches for detecting GLT8D1/CD133 interactions in glioma stem cell research?

To effectively investigate GLT8D1/CD133 interactions in glioma stem cell research:

  • Co-immunoprecipitation (Co-IP):

    • Use anti-GLT8D1 antibodies to pull down protein complexes

    • Probe with anti-CD133 antibodies to detect interaction

    • Include appropriate controls (IgG, knockout/knockdown samples)

  • Proximity ligation assays (PLA):

    • Employ GLT8D1 and CD133 primary antibodies from different species

    • Use species-specific PLA probes to visualize protein-protein interactions in situ

    • Quantify interaction signals in different cell populations

  • Fluorescence resonance energy transfer (FRET):

    • Label GLT8D1 and CD133 with compatible fluorophores

    • Measure energy transfer as indication of protein proximity

    • Control for appropriate fluorophore expression levels

Research has shown that GLT8D1 impedes CD133 degradation through the endosomal-lysosomal pathway by N-linked glycosylation and protein-protein interaction, making these interaction studies particularly relevant in understanding glioma stem cell maintenance .

How can ChIP assays be optimized using GLT8D1 antibodies to investigate transcriptional regulation?

Optimization of ChIP assays with GLT8D1 antibodies requires:

  • Crosslinking optimization:

    • Standard 1% formaldehyde for 10 minutes is suitable for most applications

    • Dual crosslinking with DSG (disuccinimidyl glutarate) followed by formaldehyde may enhance detection of weak or transient interactions

  • Antibody selection and validation:

    • Test multiple GLT8D1 antibodies for ChIP efficiency

    • Validate antibody specificity using positive and negative control regions

    • Include non-specific IgG controls

  • Sonication parameters:

    • Optimize sonication conditions to achieve 200-500 bp DNA fragments

    • Verify fragmentation by agarose gel electrophoresis

  • qPCR primer design:

    • Design primers flanking suspected binding sites (e.g., HREs in the GLT8D1 promoter)

    • Include positive control primers targeting known binding sites

    • Include negative control primers for regions not expected to be bound

Research has identified three putative hypoxia response elements (HREs) within the GLT8D1 promoter, with ChIP assays confirming HIF-1α association with two of these sites (sites 1 and 3) . This approach can be adapted to investigate other transcription factors potentially regulating GLT8D1 expression.

What protocols are recommended for measuring glycosyltransferase activity of immunoprecipitated GLT8D1?

For measuring glycosyltransferase activity of immunoprecipitated GLT8D1:

  • Immunoprecipitation procedure:

    • Lyse cells in appropriate buffer (e.g., IP lysis buffer)

    • Incubate lysates with anti-GLT8D1 antibody-conjugated beads (e.g., Dynabeads)

    • Wash thoroughly to remove non-specific proteins

    • Elute with low pH (e.g., 0.2M glycine, pH 2.6) and neutralize (e.g., 1M Tris-HCl, pH 9.0)

  • Activity assay components:

    • Purified GLT8D1 protein (10 ng/μL)

    • Appropriate substrates (e.g., 5 mM UDP-galactose, 5 mM GlcNAc)

    • Coupling phosphatase (2 ng/μL)

    • Reaction buffer

  • Detection method:

    • Incubate reaction at 37°C for 1 hour

    • Terminate with Malachite Green reagent

    • Measure absorbance at OD620

Research has shown that mutant GLT8D1 proteins (particularly I290M variant) demonstrate compromised glycosyltransferase activity compared to wild-type, which may contribute to disease pathology .

What technical considerations are important when using GLT8D1 antibodies for detecting hypoxia-induced expression in tumor samples?

When investigating hypoxia-induced GLT8D1 expression in tumor samples:

  • Sample preparation:

    • Rapid fixation to preserve hypoxic regions

    • Consider using pimonidazole labeling in experimental models to mark hypoxic regions

  • Multiplex staining approach:

    • Co-stain with hypoxia markers (HIF-1α, CA9)

    • Include stem cell markers (CD133, SOX2)

    • Use sequential staining protocols if antibodies are from the same species

  • Image acquisition and analysis:

    • Capture whole tumor section to observe heterogeneity

    • Perform quantitative analysis of co-localization

    • Correlate GLT8D1 expression with hypoxic regions and stem cell markers

  • Controls:

    • Include normoxic tissue regions as internal controls

    • Use HIF-1α knockdown controls when possible

Research has demonstrated that GLT8D1 is highly expressed in HIF-1α+ and CA9+ tumor cells that also express stem cell markers, supporting the relationship between hypoxia and GLT8D1 expression . This approach can help identify potential therapeutic targets in the hypoxic tumor microenvironment.

What is the recommended Western blot protocol to detect both wild-type and mutant GLT8D1 proteins?

For optimal Western blot detection of GLT8D1 proteins:

  • Sample preparation:

    • Lyse cells in RIPA buffer with protease inhibitors

    • Determine optimal protein concentration (typically 20-50 μg total protein)

    • Include both wild-type and mutant controls

  • Gel electrophoresis:

    • Use 10% SDS-PAGE gels for optimal resolution

    • Include molecular weight markers to confirm correct band size

  • Transfer and blocking:

    • Transfer proteins to PVDF or nitrocellulose membrane

    • Block with 5% non-fat dry milk or BSA in TBST

  • Antibody incubation:

    • Primary: anti-GLT8D1 or anti-FLAG (for tagged constructs) at 1:1000 dilution

    • Secondary: HRP-conjugated secondary antibody at 1:5000 dilution

  • Detection:

    • Use standard enhanced chemiluminescence (ECL) method

    • Include loading control (e.g., anti-actin antibody)

Research has shown that Western blot analysis can detect differences in steady-state expression levels between wild-type and mutant GLT8D1, with the I290M variant showing notably increased expression compared to wild-type and R92C variants .

How can researchers address non-specific binding when using GLT8D1 antibodies in immunohistochemistry?

To minimize non-specific binding in GLT8D1 immunohistochemistry:

  • Antibody validation:

    • Test antibodies on tissues with known GLT8D1 expression patterns

    • Include positive and negative controls (knockout/knockdown tissues)

  • Optimize blocking conditions:

    • Extend blocking time (1-2 hours)

    • Test different blocking agents (BSA, normal serum, commercial blockers)

    • Include protein-free blockers if background persists

  • Antibody dilution optimization:

    • Perform titration experiments to determine optimal concentration

    • Typically start with manufacturer's recommendation and adjust as needed

  • Additional controls:

    • Include absorption controls (pre-incubating antibody with recombinant GLT8D1)

    • Use isotype control antibodies at the same concentration

  • Signal amplification considerations:

    • If using amplification systems, optimize to prevent over-amplification of background

    • Consider direct fluorophore-conjugated antibodies for cleaner signals

Careful optimization of these parameters is essential, particularly when examining GLT8D1 expression in heterogeneous tissues like gliomas, where accurate detection is critical for correlating expression with clinical outcomes .

What strategies can resolve discrepancies between GLT8D1 mRNA and protein levels in experimental results?

When facing discrepancies between GLT8D1 mRNA and protein expression:

  • Technical validation:

    • Verify primers/probes for mRNA detection (specificity, efficiency)

    • Confirm antibody specificity for protein detection

    • Include appropriate positive and negative controls

  • Consider post-transcriptional regulation:

    • Investigate microRNA-mediated regulation

    • Assess mRNA stability through actinomycin D chase experiments

    • Examine alternative splicing using isoform-specific primers

  • Examine post-translational modifications:

    • Assess protein stability (cycloheximide chase assays)

    • Investigate ubiquitination status

    • Evaluate glycosylation patterns that may affect antibody recognition

  • Validate with alternative methods:

    • Compare results from multiple techniques (qPCR, RNA-seq, Western blot, mass spectrometry)

    • Consider single-cell analyses to identify cell-specific discrepancies

Research has observed such discrepancies, particularly when examining CD133 levels in GLT8D1 knockdown cells, where protein levels decreased while mRNA levels remained unchanged, suggesting post-transcriptional regulation .

What are the best practices for normalizing GLT8D1 expression data across diverse tissue samples?

For robust normalization of GLT8D1 expression data:

  • Reference gene selection:

    • Use multiple reference genes (3-5) rather than a single housekeeping gene

    • Validate reference gene stability across all experimental conditions

    • Consider using normalization algorithms (GeNorm, NormFinder) to identify optimal reference genes

  • Sample-specific considerations:

    • For hypoxic tissues, avoid using hypoxia-sensitive reference genes

    • For brain tissues, consider region-specific reference genes

    • For tumor samples, validate reference genes in both tumor and normal tissue

  • Technical normalization:

    • Include inter-run calibrators for multi-plate experiments

    • Use standard curves for absolute quantification

    • Consider digital PCR for reference-independent quantification

  • Statistical approaches:

    • Apply appropriate statistical methods for normalization

    • Consider using global normalization methods for large datasets

    • Report both raw and normalized data when possible

  • Validation:

    • Confirm trends with independent techniques (e.g., Western blot, immunohistochemistry)

    • Use spike-in controls for quality assessment

These approaches are particularly important when analyzing GLT8D1 expression across diverse samples such as different grades of gliomas, where expression correlates with tumor grade and patient survival .

How should researchers interpret changes in GLT8D1 localization in relation to its function?

Interpreting GLT8D1 localization changes requires:

  • Baseline understanding:

    • Normal GLT8D1 localizes to the Golgi apparatus (discrete punctate perinuclear pattern)

    • This localization is consistent with its glycosyltransferase function

  • Pathological changes to consider:

    • Retention in the ER (as seen with I290M mutation) suggests protein misfolding

    • ER retention correlates with:

      • Increased ER stress markers (BiP, CHOP, sXBP1)

      • Decreased glycosyltransferase activity

      • Increased cytotoxicity

  • Functional correlation approach:

    • Compare localization with glycosyltransferase activity

    • Measure ER stress markers in relation to localization patterns

    • Assess cell viability and cytotoxicity in relation to localization

  • Mechanistic investigations:

    • Evaluate protein-protein interactions in different subcellular locations

    • Examine post-translational modifications that may affect localization

    • Consider impact on substrate accessibility

Research shows that the I290M GLT8D1 variant displays abnormal reticular pattern throughout the cytoplasm rather than Golgi localization, which correlates with induction of ER stress and decreased enzymatic activity .

GLT8D1 VariantLocalization PatternER Stress InductionGlycosyltransferase ActivityCytotoxicity
Wild-typeGolgi (punctate)LowNormalLow
R92CGolgi (punctate)ModerateReducedModerate
I290MER (reticular)HighSignificantly compromisedHigh

What methodological approaches best demonstrate the functional impact of GLT8D1 in cancer stem cell maintenance?

To demonstrate GLT8D1's functional impact in cancer stem cell maintenance:

  • Genetic manipulation approaches:

    • shRNA-mediated knockdown of GLT8D1

    • CRISPR/Cas9 knockout of GLT8D1

    • Rescue experiments with wild-type vs. mutant GLT8D1

  • Functional assays:

    • Cell proliferation assays (growth curves, BrdU incorporation)

    • Colony formation assays

    • Sphere formation assays (number and size quantification)

    • Flow cytometry to measure stem cell marker expression (CD133, CD44)

  • Cell cycle and apoptosis analysis:

    • Cell cycle distribution by flow cytometry

    • Western blot for cell cycle regulators (p21, phospho-CDK1)

    • Apoptosis detection (Annexin V staining, TUNEL assay)

  • In vivo validation:

    • Xenograft models with GLT8D1-manipulated cells

    • Assessment of tumor initiation and growth

    • Analysis of stem cell marker expression in tumors

Research demonstrates that GLT8D1 knockdown inhibits GSC self-renewal, reduces CD133+/CD44+ cell populations, promotes cell cycle arrest at G2/M phase, and increases apoptosis, providing multiple lines of evidence for its role in cancer stem cell maintenance .

How can researchers distinguish between GLT8D1-specific effects and secondary consequences when interpreting experimental results?

To distinguish primary GLT8D1 effects from secondary consequences:

  • Timing studies:

    • Establish temporal sequence of events following GLT8D1 manipulation

    • Use inducible systems to control timing of GLT8D1 expression/knockdown

    • Monitor early vs. late changes in cell phenotypes

  • Rescue experiments:

    • Reintroduce wild-type GLT8D1 in knockdown/knockout cells

    • Test enzymatically dead mutants to determine if glycosyltransferase activity is required

    • Use domain-specific mutants to map functional regions

  • Direct vs. indirect target identification:

    • For protein level effects (e.g., CD133), distinguish between:

      • Direct glycosylation (use glycosylation site mutants)

      • Protein-protein interactions (co-IP, proximity labeling)

      • Indirect regulation (transcriptional analysis)

  • Pathway inhibition experiments:

    • Use specific inhibitors of downstream pathways

    • Determine if blocking these pathways prevents GLT8D1-mediated effects

Research shows that GLT8D1 knockdown reduces CD133 protein but not mRNA levels, suggesting post-transcriptional regulation rather than transcriptional effects. This contrasts with other stem cell markers like CD44, SOX2, and Oct4, where both mRNA and protein levels decreased, indicating different regulatory mechanisms .

What emerging applications of GLT8D1 antibodies show promise for clinical translation?

Emerging applications of GLT8D1 antibodies with clinical potential include:

  • Prognostic biomarker development:

    • Tissue microarray analysis shows GLT8D1 expression correlates with higher glioma grade

    • Patient survival analysis indicates lower survival in GLT8D1-high tumors (372 days vs. 452 days)

    • Standardized immunohistochemistry protocols could enable clinical implementation

  • Therapeutic response prediction:

    • GLT8D1 knockdown enhances temozolomide (TMZ) sensitivity

    • Antibody-based detection could identify patients likely to benefit from specific therapies

    • Multiplexed staining with other markers could improve predictive accuracy

  • Liquid biopsy development:

    • Detection of GLT8D1 in circulating tumor cells or extracellular vesicles

    • Monitoring treatment response through changes in GLT8D1 expression

    • Correlation with imaging findings for comprehensive disease monitoring

  • Therapeutic targeting:

    • Developing antibodies that can disrupt GLT8D1/CD133 complex formation

    • Employing antibody-drug conjugates targeting GLT8D1-expressing cells

    • Utilizing intrabodies to disrupt GLT8D1 function in specific cellular compartments

Clinical translation will require validation across larger patient cohorts and standardization of detection methods for reliable implementation in clinical settings.

What are the most promising methodological approaches for studying GLT8D1 in neurodegenerative disease models?

For studying GLT8D1 in neurodegenerative disease:

  • Patient-derived cellular models:

    • iPSC-derived motor neurons from ALS patients with GLT8D1 mutations

    • Isogenic iPSC lines with CRISPR-corrected mutations as controls

    • Antibody-based detection of GLT8D1 localization and processing

  • Animal models:

    • Transgenic mice expressing wild-type or mutant GLT8D1

    • AAV-mediated expression of GLT8D1 variants in specific CNS regions

    • Examination of motor function, neurodegeneration, and biochemical changes

  • Biochemical and structural approaches:

    • Cryo-EM or X-ray crystallography of wild-type vs. mutant GLT8D1

    • Glycosyltransferase activity assays with neural substrates

    • Protein-protein interaction screens in neuronal contexts

  • Therapeutic screening:

    • High-throughput screens for compounds that correct mutant GLT8D1 mislocalization

    • Assays measuring reduction in ER stress induced by mutant GLT8D1

    • Tests for restoration of glycosyltransferase activity

Research has identified ALS-associated mutations in GLT8D1, with the R92C mutation shown to be toxic in both neuronal and non-neuronal cell models , providing a foundation for these advanced approaches.

How might GLT8D1 antibodies be used to investigate connections between hypoxia response and neurodegenerative mechanisms?

To investigate hypoxia-neurodegeneration connections using GLT8D1 antibodies:

  • Comparative tissue analysis:

    • Examine GLT8D1 expression in hypoxic regions of neurodegenerative disease tissues

    • Compare with expression patterns in hypoxic regions of tumors

    • Correlate with HIF-1α and other hypoxia markers

  • Conditional expression systems:

    • Create neuronal models with HIF-1α-regulated GLT8D1 expression

    • Use antibodies to track GLT8D1 localization and processing under normoxic vs. hypoxic conditions

    • Assess neuronal viability, morphology, and function

  • Protein interaction networks:

    • Identify GLT8D1 interaction partners in neurons under normoxic vs. hypoxic conditions

    • Compare with interaction networks in cancer cells

    • Identify common pathways and divergent mechanisms

  • Glycosylation target identification:

    • Determine neuronal proteins glycosylated by GLT8D1

    • Assess how hypoxia affects substrate selection and modification

    • Evaluate functional consequences of altered glycosylation patterns

Research has established that GLT8D1 is induced by HIF-1α under hypoxic conditions through direct binding to hypoxia response elements in the GLT8D1 promoter . This regulatory mechanism may have implications for both cancer and neurodegenerative diseases where hypoxic conditions occur.

What are the most reliable GLT8D1 antibody validation strategies for ensuring reproducible research?

For ensuring reliable GLT8D1 antibody validation:

  • Multi-level verification approach:

    • Genetic controls: Test antibodies on knockout/knockdown samples

    • Expression systems: Validate against overexpressed wild-type and tagged proteins

    • Multiple applications: Verify performance across Western blot, immunocytochemistry, and immunoprecipitation

  • Cross-validation with multiple antibodies:

    • Use antibodies targeting different epitopes of GLT8D1

    • Compare monoclonal and polyclonal antibodies

    • Verify concordant results across antibodies from different vendors

  • Application-specific validation:

    • Western blot: Confirm single band of correct molecular weight

    • Immunofluorescence: Verify expected subcellular localization (Golgi apparatus)

    • Immunoprecipitation: Confirm pull-down of GLT8D1 by mass spectrometry

  • Documentation and reporting standards:

    • Record complete antibody information (vendor, catalog number, lot, dilution)

    • Document all validation experiments performed

    • Share validation data through repositories or supplementary materials

These validation approaches are essential for ensuring reproducibility in GLT8D1 research, particularly when studying subtle differences between wild-type and mutant proteins or examining expression in heterogeneous tissues.

What integrated experimental approaches provide the most comprehensive understanding of GLT8D1 function in disease models?

For comprehensive understanding of GLT8D1 function:

  • Multi-omics integration:

    • Transcriptomics: RNA-seq to identify gene expression changes

    • Proteomics: Mass spectrometry to detect protein level changes and PTMs

    • Glycomics: Analysis of glycosylation patterns

    • Integration of datasets to identify convergent pathways

  • Functional pathway analysis:

    • ER stress pathway assessment (BiP, CHOP, sXBP1 expression)

    • Cell cycle regulation (G2/M checkpoint proteins)

    • Apoptotic pathway activation

    • Stem cell maintenance networks

  • Temporal dynamics:

    • Time-course experiments following GLT8D1 manipulation

    • Inducible systems to control expression timing

    • Live-cell imaging with tagged GLT8D1 to monitor localization changes

  • In vivo validation:

    • Patient sample correlation studies

    • Animal models of GLT8D1 mutation or dysregulation

    • Therapeutic intervention studies

Research implementing these integrated approaches has revealed that GLT8D1 functions in multiple contexts, from glycosylation activity in the Golgi to protein-protein interactions affecting CD133 degradation, with mutations causing mislocalization and ER stress .

How should researchers interpret GLT8D1 antibody data across different disease contexts?

When interpreting GLT8D1 antibody data across diseases:

  • Context-specific baseline establishment:

    • Determine normal GLT8D1 expression patterns in relevant tissues

    • Establish cell type-specific expression profiles

    • Consider developmental stage-specific expression patterns

  • Disease-specific considerations:

    • Neurodegenerative diseases: Focus on mutation-specific effects and cellular stress

    • Cancer: Emphasize connections to stem cell maintenance and hypoxia response

    • Consider tissue-specific post-translational modifications

  • Comparative analysis framework:

    • Systematically compare expression, localization, and interaction partners

    • Identify common vs. disease-specific mechanisms

    • Correlate with clinical parameters where available

  • Methodological standardization:

    • Use consistent antibodies and protocols across disease studies

    • Include appropriate disease-specific controls

    • Employ quantitative methods for objective comparison

Research demonstrates disease-specific patterns, with GLT8D1 mutations causing ER stress in ALS models and hypoxia-induced GLT8D1 maintaining cancer stem cells in gliomas . Understanding these context-specific mechanisms is essential for developing targeted therapeutic approaches.

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.