GTPBP2 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Target

The GTPBP2 antibody is a polyclonal rabbit-derived IgG antibody targeting GTPBP2, a GTPase implicated in ribosomal function, Wnt/β-catenin signaling, and cancer progression . GTPBP2 regulates translation efficiency and interacts with pathways critical for neurodevelopment and tumor metastasis .

3.1. Western Blot (WB)

  • Detects GTPBP2 in human kidney and spleen tissues, with optimal dilution at 1:500–1:2000 .

  • Used to identify GTPBP2 overexpression in non-small cell lung cancer (NSCLC) tissues compared to normal tissues .

3.2. Immunohistochemistry (IHC)

  • Validated in human pancreas tissue (1:50–1:500 dilution) .

  • Revealed elevated GTPBP2 expression in NSCLC tumors, correlating with advanced TNM stages and lymph node metastasis .

3.3. Functional Studies

  • Cancer Research: GTPBP2 knockdown in NSCLC cell lines (A549, H1299) reduced invasion/migration by downregulating RhoC and MMP-9, and suppressed proliferation via cyclinD1/CDK4/c-myc pathways .

  • Mechanistic Insights: GTPBP2 modulates Wnt/β-catenin signaling by inhibiting Axin, promoting β-catenin accumulation and tumor progression .

4.1. Association with NSCLC Prognosis

A study of 112 NSCLC patients demonstrated GTPBP2’s role as a prognostic marker :

Clinical ParameterHigh GTPBP2 ExpressionLow GTPBP2 ExpressionP-value
TNM Stage (II/III vs. I)43230.005
Lymph Node Metastasis37290.014

Patients with high GTPBP2 levels had shorter 5-year survival rates, underscoring its oncogenic role .

4.2. Neurodevelopmental Disorders

While GTPBP2 is primarily studied in cancer, its paralog GTPBP1 is linked to neurodevelopmental disorders (e.g., microcephaly, epilepsy) . This highlights the antibody’s potential utility in broader translational research .

Protocols and Validation

  • WB Protocol: Requires antigen retrieval with TE buffer (pH 9.0) or citrate buffer (pH 6.0) .

  • Key Controls: Include human spleen (positive) and knockdown cell lysates (negative) to ensure specificity .

Future Directions

  • Therapeutic Targeting: GTPBP2’s role in Wnt signaling and EMT positions it as a potential target for NSCLC therapy .

  • Neurological Research: Exploring GTPBP2’s conserved function in Drosophila models could clarify its role in neurodevelopment .

Product Specs

Buffer
Phosphate Buffered Saline (PBS) with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid repeated freeze-thaw cycles.
Lead Time
We typically dispatch GTPBP2 Antibody orders within 1-3 business days of receipt. Delivery times may vary depending on the purchase method and destination. For specific delivery timeframes, please consult your local distributor.
Synonyms
GTP binding protein 2 antibody; GTP-binding protein 2 antibody; GTPB2_HUMAN antibody; GTPBP 2 antibody; GTPBP2 antibody; MGC74725 antibody; RP11-22I24.2 antibody
Target Names
GTPBP2
Uniprot No.

Q&A

What is GTPBP2 and why is it important in cellular research?

GTPBP2 (GTP-binding protein 2) is a member of the G protein superfamily with GTP hydrolase activity. It plays crucial roles in cell signal transmission, cytoskeletal regulation, and protein synthesis. GTPBP2 has significant research importance due to its highly conserved nature across species, with 92.8% and 99.1% similarity at DNA and protein levels between mouse and human respectively . This conservation suggests fundamental biological functions. Recent research has identified GTPBP2's involvement in cancer progression, particularly in non-small cell lung cancer (NSCLC), where it regulates cell invasion and migration through multiple signaling pathways . Additionally, GTPBP2 allele-inactivating variants have been associated with neurodevelopmental disorders and intellectual disabilities, making it relevant to both oncology and neuroscience research .

What are the optimal methods for detecting GTPBP2 expression in tissue samples?

For detecting GTPBP2 in tissue samples, immunohistochemistry (IHC) has proven particularly effective. The methodology involves:

  • Fixing specimens in 10% neutral formalin

  • Preparing 4μm thick slices

  • Dewaxing in xylene and hydrating with gradient alcohol

  • Performing high-pressure antigen retrieval in citrate buffer (2 minutes)

  • Incubating with GTPBP2 rabbit anti-human polyclonal antibody (1:100 dilution) overnight at 4°C

  • Applying appropriate secondary antibody, followed by DAB for color development

  • Counterstaining with hematoxylin, dehydrating and mounting

This protocol has successfully demonstrated differential GTPBP2 expression between cancer tissues and adjacent normal tissues. For quantifiable results, this approach can be complemented with Western blot and quantitative real-time PCR to validate protein and mRNA expression levels respectively .

How should researchers interpret variations in GTPBP2 staining intensity across different tissue sections?

When interpreting variations in GTPBP2 staining intensity, researchers should consider several methodological factors:

  • Tissue-specific baseline expression: Research has shown that GTPBP2 is naturally highly expressed in certain tissues (esophagus, testis) with moderate expression in others (lung, brain, kidney) . Therefore, baseline differences should be established with proper controls.

  • Quantification approach: Rather than subjective assessment, establish a scoring system that accounts for both staining intensity and percentage of positive cells. This allows statistical comparison between tissue types.

  • Correlation with clinical parameters: In NSCLC studies, GTPBP2 expression correlated significantly with TNM stage (P=0.005) and lymph node metastasis (P=0.014), while showing no correlation with age, gender, histological type, or differentiation degree . This pattern helps validate whether observed variations have biological significance.

  • Survival analysis: Kaplan-Meier analysis has shown that high GTPBP2 expression correlates with lower 5-year survival rates in NSCLC, providing functional context to expression differences .

When variations are observed, verification with alternative detection methods (Western blot, qPCR) is strongly recommended to confirm whether the differences reflect biological reality or technical artifacts.

What criteria should guide the selection of GTPBP2 antibodies for specific experimental applications?

When selecting GTPBP2 antibodies, researchers should consider application-specific criteria:

  • For Western blotting: Anti-GTPBP2 rabbit-monoclonal antibodies (e.g., PA5-56738) have demonstrated high specificity and sensitivity at concentrations of 0.1μg/ml . Monoclonals generally provide better specificity for detecting the approximately 65kDa GTPBP2 protein.

  • For immunohistochemistry: Polyclonal rabbit anti-human GTPBP2 antibodies at 1:100 dilution have been successfully employed in formalin-fixed, paraffin-embedded tissues following citrate buffer-based antigen retrieval .

  • For immunofluorescence: Rabbit anti-GTPBP2 antibodies (e.g., PA5-56738) at 1μg/ml concentration work effectively when coupled with appropriate fluorescent secondary antibodies .

  • Epitope considerations: Select antibodies targeting conserved regions if cross-species reactivity is desired. The high conservation between human and mouse GTPBP2 (99.1% at protein level) means many antibodies work across these species .

  • Validation requirements: Prioritize antibodies validated through multiple applications and techniques, especially those with knockout/knockdown controls that demonstrate specificity.

For comprehensive investigations, researchers might need different antibodies optimized for each application rather than relying on a single antibody for all experimental needs.

How can researchers effectively validate GTPBP2 antibody specificity before experimental use?

Thorough validation of GTPBP2 antibody specificity requires a multi-step approach:

  • Knockdown/knockout controls: The gold standard validation involves comparing antibody signals between wild-type samples and those with GTPBP2 knockdown (via shRNA) or knockout. Published research has demonstrated this approach by showing decreased GTPBP2 signal following knockdown in A549 cell lines .

  • Recombinant protein controls: Using full-length recombinant GTPBP2 protein as a positive control. The canine GTPBP2 cDNA has been synthetically generated based on published sequences (NCBI Reference Sequence: XM_538939.7) and cloned into expression vectors . Similar approaches with human GTPBP2 can serve as excellent positive controls.

  • Multi-technique confirmation: Validate expression patterns across complementary techniques:

    • Western blot for protein size verification

    • Immunohistochemistry/immunofluorescence for localization patterns

    • qPCR for correlation with transcript levels

  • Cross-reactivity assessment: Test the antibody against related proteins, particularly GTPBP1, which is the closest homolog to GTPBP2 . This ensures signals are not due to cross-reactivity.

  • Predictable expression patterns: Compare observed patterns with expected tissue expression (higher in esophagus and testis, moderate in lung, brain, and kidney) .

Documentation of these validation steps should be maintained to ensure experimental reproducibility and reliability.

What are the key differences between polyclonal and monoclonal GTPBP2 antibodies in research applications?

The choice between polyclonal and monoclonal GTPBP2 antibodies significantly impacts experimental outcomes:

Polyclonal GTPBP2 Antibodies:

  • Sensitivity advantage: Recognize multiple epitopes, enhancing detection signals in tissues with low GTPBP2 expression. This has been valuable in immunohistochemistry studies of normal lung tissues where GTPBP2 expression is relatively lower .

  • Broader reactivity: Useful when studying GTPBP2 across species due to recognition of multiple conserved epitopes.

  • Application strengths: Often preferred for immunoprecipitation and immunohistochemistry of formalin-fixed tissues due to recognition of multiple epitopes that may survive fixation.

  • Limitations: Batch-to-batch variation requires careful validation between lots.

Monoclonal GTPBP2 Antibodies:

  • Specificity advantage: Recognize a single epitope, providing higher specificity. Rabbit monoclonal anti-GTPBP2 (e.g., PA5-56738) has demonstrated excellent specificity in Western blot applications .

  • Consistency: Minimal batch-to-batch variation ensures reproducibility across experiments.

  • Application strengths: Preferred for Western blot, flow cytometry, and immunofluorescence where background must be minimized.

  • Limitations: May lose reactivity if the single epitope is masked or modified in certain experimental conditions.

Research involving comparison between normal and pathological states often benefits from using both types – monoclonals for precise quantification and polyclonals for robust detection in varied sample conditions.

What are the optimal protocols for using GTPBP2 antibodies in Western blot analyses?

For optimal Western blot detection of GTPBP2, the following validated protocol is recommended:

  • Sample preparation:

    • Extract proteins using standard lysis buffers containing protease inhibitors

    • Determine protein concentration (BCA or Bradford assay)

    • Load 20-30μg of total protein per lane

  • Gel electrophoresis:

    • Resolve proteins on 10% SDS-PAGE gels

    • Include molecular weight markers to verify the expected 65kDa size of GTPBP2

  • Transfer conditions:

    • Transfer to nitrocellulose membrane (preferred over PVDF for GTPBP2)

    • Use standard transfer buffer (25mM Tris, 192mM glycine, 20% methanol)

  • Blocking:

    • Block with 3% bovine serum albumin in TBS-T (TBS + 0.05% Tween-20) for one hour at room temperature

    • BSA has shown better results than milk-based blocking buffers for GTPBP2 detection

  • Antibody incubation:

    • Primary: Anti-GTPBP2 rabbit-monoclonal antibody (e.g., PA5-56738) at 0.1μg/ml concentration

    • Incubate overnight at 4°C with gentle rocking

    • Secondary: Anti-rabbit-HRP (1:30,000 dilution)

    • Incubate for 1 hour at room temperature

  • Development and imaging:

    • Develop with DAB or enhanced chemiluminescence reagents

    • Image using appropriate systems (e.g., iBright 1500)

  • Quantification:

    • Perform densitometric analysis from at least three independent experiments

    • Normalize to appropriate loading controls (e.g., GAPDH, β-actin)

This protocol has been successfully employed to detect differential GTPBP2 expression between NSCLC tissues and adjacent normal tissues, as well as between cancer cell lines and normal bronchial epithelial cells .

How can researchers optimize immunofluorescence protocols for GTPBP2 subcellular localization studies?

Optimizing immunofluorescence for GTPBP2 subcellular localization requires careful attention to fixation, permeabilization, and antibody conditions:

  • Cell preparation and fixation:

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

    • Wash thoroughly with PBS (3× 5-minute washes)

    • This fixation method preserves GTPBP2 epitopes while maintaining cellular architecture

  • Permeabilization optimization:

    • Use 0.5% Triton X-100 in PBS for 10 minutes at room temperature

    • For delicate subcellular structures, a milder permeabilization with 0.1% Triton X-100 or 0.1% saponin may be preferable

  • Blocking conditions:

    • Block with 1% BSA in PBS for 30-60 minutes at room temperature

    • Add 5-10% normal serum (from the species of the secondary antibody) to reduce background

  • Antibody selection and dilution:

    • Primary: Rabbit anti-GTPBP2 PA5-56738 at 1μg/ml concentration in 1% BSA/PBS solution

    • For co-localization studies, pair with organelle markers such as Lamp2 for lysosomes

    • Secondary: Alexa Fluor 568 (red) or Alexa Fluor 488 (green) at 2μg/ml

  • Nuclear counterstaining:

    • Use Hoechst 33342 (1μg/ml) for nuclear morphology visualization

  • Mounting and imaging:

    • Mount with anti-fade mounting medium (e.g., Mowiol) to preserve fluorescence

    • Image using confocal microscopy with appropriate laser settings (e.g., TCS-SP5 II confocal laser scanning microscope)

  • Controls and validation:

    • Include negative controls (primary antibody omission, isotype controls)

    • Validate localization patterns with subcellular fractionation and Western blotting

    • Consider GTPBP2 knockdown cells as specificity controls

This approach has successfully revealed GTPBP2 subcellular distribution patterns and allowed co-localization analysis with other cellular markers, providing insights into its functional relationships within the cell .

What quantitative methods are most reliable for measuring GTPBP2 expression levels in clinical samples?

For reliable quantification of GTPBP2 expression in clinical samples, a multi-platform approach is recommended:

  • Quantitative real-time PCR (qPCR):

    • RNA extraction using standard protocols (TRIzol or column-based methods)

    • cDNA synthesis with oligo(dT) or random primers

    • qPCR using validated GTPBP2 primers:

      • Forward: CTGGCTGAGGAGGAAATG

      • Reverse: CACACGGAGGTCTAGGAAC

    • Normalization to stable reference genes (GAPDH validated for GTPBP2 studies):

      • GAPDH Forward: GAAGGTGAAGGTCGGAGT

      • GAPDH Reverse: GAAGATGGTGATGGGATTTC

    • Analysis using the 2^-ΔΔCT method with appropriate statistical analysis

  • Immunohistochemistry (IHC) with digital quantification:

    • Standard IHC protocol as described previously

    • Digital slide scanning and analysis using software packages

    • Quantification parameters:

      • Percentage of positive cells

      • Staining intensity (0-3+ scale)

      • H-score calculation (percentage × intensity)

    • Correlation with clinical parameters (TNM stage, lymph node status, survival)

  • Western blot with densitometry:

    • Standard Western blot protocol as described previously

    • Densitometric analysis of at least three independent experiments

    • Normalization to appropriate loading controls

    • Statistical analysis comparing patient groups

  • TPM calculation from RNA-sequencing data:

    • RNA-seq analysis using Kallisto or similar software to calculate Transcripts Per Million (TPM) for GTPBP2

    • Comparison between affected and normal samples

    • This method allows integration with broader transcriptomic datasets

Each method has strengths: qPCR provides sensitive mRNA quantification, IHC reveals spatial distribution, Western blot confirms protein size, and RNA-seq places expression in broader context. Ideally, researchers should employ at least two complementary techniques for robust quantification in clinical studies.

How does GTPBP2 antibody staining correlate with cancer progression and clinical outcomes?

Research investigating GTPBP2 expression in NSCLC has revealed significant correlations with cancer progression and clinical outcomes:

  • Cancer stage correlation:

    • GTPBP2 expression shows statistically significant correlation with TNM stage (P=0.005)

    • Higher expression in more advanced stages (II+III) compared to stage I

    • The correlation data is presented in the following table:

    TNM StageCasesLow GTPBP2 ExpressionHigh GTPBP2 ExpressionP-value
    I5431230.005*
    II+III581543
  • Lymph node metastasis association:

    • High GTPBP2 expression correlates with lymph node metastasis (P=0.014)

    • Distribution data shows:

    Nodal StatusCasesLow GTPBP2 ExpressionHigh GTPBP2 ExpressionP-value
    N06031290.014*
    N1/N2/N3521537
  • Survival outcomes:

    • Kaplan-Meier analysis demonstrates that high GTPBP2 expression correlates with lower 5-year post-operative survival rates

    • This relationship was statistically significant (P<0.05)

  • Non-significant correlations:

    • GTPBP2 expression shows no statistically significant correlation with:

      • Age (P=0.129)

      • Gender (P=0.506)

      • Histological type (P=0.541)

      • Differentiation grade (P=0.146)

These findings suggest GTPBP2 antibody staining has potential as a prognostic biomarker, particularly for predicting lymph node involvement and disease progression. Methodologically, researchers should ensure standardized staining protocols, blinded assessment, and appropriate statistical analyses when investigating these correlations in new cohorts or cancer types.

What technical challenges arise when using GTPBP2 antibodies in dual immunofluorescence or multiplexed staining approaches?

Dual immunofluorescence and multiplexed staining with GTPBP2 antibodies present several technical challenges that require specific optimization strategies:

  • Antibody species compatibility:

    • Since many validated GTPBP2 antibodies are rabbit-derived , pairing with other rabbit antibodies becomes problematic

    • Solution: Use directly conjugated primary antibodies, sequential staining with Fab fragments to block first primary, or tyramide signal amplification (TSA) methods

  • Signal strength balancing:

    • GTPBP2 may have lower expression in some contexts compared to abundant proteins

    • Solution: Optimize antibody concentrations individually (typically 1μg/ml for GTPBP2) , adjust exposure settings, or employ signal amplification for weaker signals

  • Cross-reactivity with related GTPases:

    • GTPBP1 is highly homologous to GTPBP2 , potentially causing cross-reactivity

    • Solution: Pre-absorb antibodies against recombinant related proteins or validate using GTPBP2 knockdown controls alongside GTPBP1 knockdowns

  • Autofluorescence interference:

    • Particularly problematic in tissues with high natural autofluorescence (e.g., lung tissue)

    • Solution: Include Sudan Black B (0.1%) treatment, use spectral unmixing, or employ fluorophores with emission spectra distinct from autofluorescence peaks

  • Epitope masking in multiplex protocols:

    • Sequential staining may lead to epitope blocking or antibody stripping issues

    • Solution: Optimize antigen retrieval methods specifically for GTPBP2 (citrate buffer has been validated) , test different sequential staining orders

  • Subcellular localization verification:

    • Ensuring co-localization findings are not artifacts

    • Solution: Include organelle markers (e.g., Lamp2 for lysosomes has been successfully paired with GTPBP2) , validate with super-resolution microscopy

When implementing these techniques, thorough controls are critical: single-stain controls, secondary-only controls, and isotype controls should be included in every experiment to ensure reliable interpretation of co-localization or multiplexed data.

How can GTPBP2 knockdown models be used to validate antibody specificity and study protein function?

GTPBP2 knockdown models serve dual purposes in validating antibody specificity and elucidating protein function:

  • Antibody validation methodology:

    • Generate shRNA-mediated GTPBP2 knockdown in relevant cell lines (e.g., A549 for lung cancer studies)

    • Perform Western blot analysis with the GTPBP2 antibody being validated

    • Observe significant reduction in band intensity at the expected molecular weight (~65kDa)

    • Quantify the knockdown efficiency through densitometric analysis

    • This approach provides definitive evidence of antibody specificity

  • Functional studies enabled by validated knockdowns:

    • Migration assays: Knockdown studies have demonstrated that decreased GTPBP2 expression reduces migration function in A549 cell lines

    • Invasion assays: Using Matrigel-coated transwell chambers (1:9 dilution), researchers observed decreased invasion capacity following GTPBP2 knockdown

    • Proliferation analysis: Cell proliferation capacity decreases with GTPBP2 knockdown

  • Molecular pathway analysis:

    • GTPBP2 knockdown experiments revealed:

      • No significant changes in RhoA and RhoB proteins

      • Decreased RhoC expression

      • Reduced MMP-9 expression

      • Decreased expression of cyclinD1, CDK4, and c-myc

      • Increased expression of P27

    • These findings indicate that GTPBP2 influences cell invasion and proliferation through specific molecular pathways

  • Protocol optimization for knockdown studies:

    • Transfection efficiency should be monitored using co-transfected reporters

    • Multiple shRNA constructs targeting different regions should be tested

    • Knockdown confirmation should employ both protein (Western blot) and mRNA (qPCR) analysis

    • Functional assays should be performed at optimal timepoints post-knockdown (typically 48-72 hours)

  • Alternative validation approaches:

    • CRISPR-Cas9 knockout models offer complete protein elimination

    • Rescue experiments (re-introducing wild-type or mutant GTPBP2) can confirm specificity of observed phenotypes

    • Site-directed mutagenesis can generate specific variants such as the canine GTPBP2 in-frame deletion model

This integrated approach not only validates antibody specificity but also provides mechanistic insights into GTPBP2 function, demonstrating how it promotes NSCLC progression through multiple signaling pathways .

What are common pitfalls in GTPBP2 antibody-based experiments and how can researchers address them?

Researchers frequently encounter several challenges when working with GTPBP2 antibodies:

  • Weak or absent signal in Western blots:

    • Cause: Insufficient protein extraction, degradation, or low endogenous expression

    • Solution:

      • Use fresh samples with protease inhibitors

      • Increase loading amount (30-40μg total protein)

      • Extend primary antibody incubation (overnight at 4°C)

      • Use enhanced chemiluminescence substrates with longer exposure times

      • Focus on tissues known to express GTPBP2 (esophagus, testis, lung)

  • High background in immunohistochemistry:

    • Cause: Insufficient blocking, non-specific binding, or excessive antibody concentration

    • Solution:

      • Optimize blocking (3% BSA has shown good results)

      • Titrate primary antibody concentration (starting from 1:100 dilution)

      • Include additional washing steps with 0.1% Tween-20

      • Pre-absorb antibody against tissue lysates from GTPBP2 knockdown models

  • Inconsistent immunofluorescence staining patterns:

    • Cause: Fixation artifacts, antibody batch variation, or cellular heterogeneity

    • Solution:

      • Standardize fixation protocol (4% PFA for 15 minutes) and permeabilization (0.5% Triton X-100)

      • Validate with orthogonal methods (subcellular fractionation, co-localization)

      • Include positive controls (tissues/cells with known expression patterns)

  • Cross-reactivity with related proteins:

    • Cause: Antibody recognition of homologous domains in related GTPases, particularly GTPBP1

    • Solution:

      • Validate with GTPBP2 knockdown/knockout controls

      • Perform parallel Western blots with recombinant GTPBP1 and GTPBP2

      • Use monoclonal antibodies targeting unique epitopes when available

  • Inconsistent qPCR results:

    • Cause: RNA degradation, primer inefficiency, or inappropriate reference genes

    • Solution:

      • Check RNA integrity before cDNA synthesis

      • Validate primer efficiency (90-110% range)

      • Use validated GTPBP2-specific primers:

        • Forward: CTGGCTGAGGAGGAAATG

        • Reverse: CACACGGAGGTCTAGGAAC

      • Include multiple reference genes beyond GAPDH

Implementing these solutions requires systematic optimization and inclusion of appropriate controls in each experiment to ensure reliable and reproducible results.

How can researchers distinguish between true GTPBP2 signals and artifacts in their experiments?

Distinguishing genuine GTPBP2 signals from artifacts requires a multi-faceted validation approach:

  • Biological validation controls:

    • Knockdown/knockout verification: The most definitive control involves comparing staining patterns between wild-type and GTPBP2-depleted samples. Significant signal reduction in knockdown samples confirms specificity

    • Expression pattern consistency: GTPBP2 shows tissue-specific expression patterns (higher in esophagus and testis) . Signals that dramatically deviate from established patterns without biological rationale warrant scrutiny

  • Technical validation approaches:

    • Multiple antibody validation: Using antibodies targeting different GTPBP2 epitopes should yield consistent patterns

    • Size verification: In Western blots, GTPBP2 appears at approximately 65kDa. Bands at significantly different molecular weights may represent non-specific binding or degradation products

    • Multi-method confirmation: Correlating protein detection (Western blot/IHC) with mRNA expression (qPCR/RNA-seq) provides stronger evidence of true signals

  • Blocking peptide experiments:

    • Pre-incubating the antibody with excess recombinant GTPBP2 protein should abolish specific signals while non-specific binding often remains

    • This approach is particularly valuable for new antibodies or those with limited validation history

  • Subcellular localization assessment:

    • Artifacts often show unusual subcellular distribution patterns

    • Verify GTPBP2 subcellular localization through co-staining with organelle markers

    • Perform subcellular fractionation to biochemically confirm localization patterns observed in imaging studies

  • Signal quantification analysis:

    • True biological signals typically show dose-dependent responses and statistically significant differences between experimental groups

    • Isolated extreme outliers often represent technical artifacts

    • Quantifying expression across multiple independent experiments and biological replicates helps distinguish consistent signals from artifacts

By implementing these validation strategies, researchers can confidently differentiate between genuine GTPBP2 biological signals and experimental artifacts, ensuring the reliability and reproducibility of their findings.

What quality control measures should be implemented when using GTPBP2 antibodies across different experimental batches?

To ensure consistency and reliability when using GTPBP2 antibodies across different experimental batches, implement these quality control measures:

  • Antibody validation for each new lot:

    • Perform Western blot analysis on standardized positive control samples (e.g., A549 cells for GTPBP2)

    • Compare band intensity and specificity between antibody lots

    • Document lot numbers and validation results in laboratory records

    • Consider creating an antibody validation report template for standardization

  • Reference sample inclusion:

    • Maintain frozen aliquots of reference samples (cell lysates or tissue extracts)

    • Include these references in each experimental batch

    • Calculate inter-assay coefficient of variation (CV) to monitor consistency

    • Acceptable inter-assay CV should be <15% for quantitative applications

  • Standard curve development for quantitative applications:

    • Generate standard curves using recombinant GTPBP2 protein

    • Include standard points spanning the expected concentration range

    • Establish limits of detection and quantification for each antibody lot

  • Internal controls for immunostaining experiments:

    • Include tissue microarrays (TMAs) with known GTPBP2 expression patterns

    • Process control slides alongside experimental samples

    • Score staining intensity using standardized scales

    • Document imaging parameters for consistency (exposure times, gain settings)

  • Batch effect monitoring and correction:

    • Randomize samples across batches when possible

    • Include at least one identical sample in all batches as a bridging sample

    • When analyzing data, use appropriate statistical methods to identify and correct batch effects

    • Consider using mixed-effects models that account for batch as a random effect

  • Documentation and reporting standards:

    • Maintain detailed records of antibody source, catalog number, lot number, and dilution

    • Document all experimental conditions (incubation times, temperatures, buffer compositions)

    • Report validation methods and quality control outcomes in publications

    • Consider sharing antibody validation data through repositories or supplementary information

Implementation of these quality control measures significantly reduces inter-experimental variability and increases confidence in results obtained across different experimental batches or between different laboratories studying GTPBP2.

How are GTPBP2 antibodies being used to investigate the protein's role in cancer progression mechanisms?

GTPBP2 antibodies are enabling novel insights into cancer progression mechanisms through several innovative research approaches:

  • Signaling pathway delineation:

    • GTPBP2 antibodies have revealed that this protein negatively regulates Axin to promote β-catenin expression, thereby activating Wnt/β-catenin signaling in NSCLC

    • Immunoblotting after GTPBP2 knockdown demonstrated its regulatory effects on multiple cancer-associated proteins:

      • Upregulation of RhoC and MMP-9 (invasion mediators)

      • Upregulation of cyclinD1, CDK4, and c-myc (proliferation drivers)

      • Downregulation of P27 (cell cycle inhibitor)

    • These findings establish GTPBP2 as a multifunctional regulator in cancer progression

  • Clinical correlation studies:

    • Immunohistochemistry with GTPBP2 antibodies has established correlations between expression levels and:

      • TNM stage (P=0.005)

      • Lymph node metastasis (P=0.014)

      • 5-year survival rates

    • These correlations support GTPBP2's potential as a prognostic biomarker

  • Therapeutic target validation:

    • GTPBP2 knockdown experiments monitored by antibody detection have demonstrated reduced cancer cell proliferation and invasion

    • This suggests potential therapeutic value in targeting GTPBP2 or its downstream effectors

  • Microenvironmental interaction studies:

    • Dual immunofluorescence approaches combining GTPBP2 antibodies with markers for tumor-associated macrophages, cancer-associated fibroblasts, or immune cells are beginning to reveal how GTPBP2 might influence the tumor microenvironment

  • Liquid biopsy development:

    • Research is exploring whether GTPBP2 protein detected by sensitive immunoassays in circulating tumor cells or exosomes could serve as a minimally invasive biomarker

These approaches collectively demonstrate how GTPBP2 antibodies are facilitating the transition from basic mechanistic understanding to potential clinical applications in cancer research.

What new insights are emerging about GTPBP2's role in neurological disorders and how are antibodies contributing to this research?

Emerging research is revealing GTPBP2's crucial roles in neurological function, with antibody-based approaches providing key insights:

  • Neurodevelopmental disorder associations:

    • GTPBP2 allele-inactivating variants have been linked to familial neurodevelopmental disorders and intellectual disabilities

    • Immunohistochemistry is helping map GTPBP2 expression across different brain regions and developmental stages

    • This spatial and temporal expression profiling is crucial for understanding how GTPBP2 mutations lead to specific neurological phenotypes

  • Translational quality control mechanisms:

    • Related research on GTPBP1 (the closest homolog to GTPBP2) has shown its importance in resolving paused ribosomes during tRNA deficiency, which is necessary for neuronal survival

    • Antibody-based co-immunoprecipitation studies are beginning to identify neuronal-specific interaction partners for GTPBP2

    • These interactions may explain the particular vulnerability of neural tissues to GTPBP2 dysfunction

  • Animal model validation:

    • Canine models with GTPBP2 in-frame deletions show non-syndromic conditions that provide valuable insights into GTPBP2 function

    • Immunofluorescence studies in these models help correlate protein expression/localization with phenotypic outcomes

    • The high conservation of GTPBP2 between species (99.1% protein similarity between mouse and human) makes these models particularly relevant

  • Subcellular localization in neurons:

    • Advanced immunofluorescence techniques are revealing GTPBP2's distribution within neuronal compartments (soma, dendrites, axons)

    • Co-localization with organelle markers (similar to Lamp2 studies in other cell types) is providing insights into GTPBP2's functional roles in neuronal homeostasis

  • Post-translational modification analysis:

    • Phospho-specific antibodies are beginning to reveal how GTPBP2's activity is regulated in neuronal contexts

    • These studies are identifying signaling pathways that might be targeted to modulate GTPBP2 function in neurological disorders

Antibody-based approaches are thus proving instrumental in bridging genetic associations with functional mechanisms, potentially opening new therapeutic avenues for GTPBP2-associated neurological conditions.

How might single-cell analysis techniques using GTPBP2 antibodies advance our understanding of cellular heterogeneity?

Single-cell analysis techniques employing GTPBP2 antibodies are poised to transform our understanding of cellular heterogeneity in both normal and pathological contexts:

  • Single-cell protein profiling approaches:

    • Mass cytometry (CyTOF) incorporating GTPBP2 antibodies can simultaneously measure dozens of proteins at single-cell resolution

    • This approach may reveal previously unrecognized cell subpopulations with distinct GTPBP2 expression patterns

    • Correlation with other cancer-related markers (e.g., RhoC, MMP-9, cyclinD1) at the single-cell level can identify co-regulated pathways

  • Spatial transcriptomics integration:

    • Combining GTPBP2 immunofluorescence with in situ hybridization techniques (e.g., RNAscope)

    • This integration allows correlation between GTPBP2 protein expression and transcriptional programs at single-cell resolution

    • Particularly valuable in tumor samples to map cellular heterogeneity across distinct microenvironmental niches

  • Single-cell multi-omics applications:

    • Emerging technologies allowing simultaneous protein (including GTPBP2) and transcript measurement from the same cell

    • These approaches can reveal post-transcriptional regulation mechanisms affecting GTPBP2

    • Additionally, they may identify regulatory relationships between GTPBP2 and other cellular factors

  • Lineage tracing with GTPBP2 expression:

    • Combining GTPBP2 antibody staining with cell lineage markers

    • This approach can track how GTPBP2 expression changes during differentiation or disease progression

    • Particularly relevant in cancer studies to identify whether GTPBP2-high cells represent specific cancer stem cell populations

  • Methodological considerations for single-cell GTPBP2 analysis:

    • Antibody conjugation protocols must preserve epitope recognition while adding detection tags

    • Validation in cell mixtures with known GTPBP2 expression levels is essential

    • Computational pipelines need optimization to handle potential technical artifacts while preserving true biological variation

These single-cell approaches are revealing how seemingly homogeneous cell populations actually contain distinct subpopulations with varying GTPBP2 expression levels, potentially explaining differential responses to treatments and varied clinical outcomes in diseases such as NSCLC .

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.