GNG2 Antibody

Shipped with Ice Packs
In Stock

Description

Applications in Research

The GNG2 antibody has been instrumental in uncovering the protein’s role in cancer biology and signaling pathways.

Key Research Findings

Tumor Suppression in Breast Cancer

  • Mechanism: GNG2 inhibits ERK and Akt signaling by directly binding MRAS (muscle RAS oncogene homolog), suppressing proliferation and promoting apoptosis in breast cancer cells .

  • Experimental Evidence:

    • Overexpression of GNG2 reduced MCF-7 and MDA-MB-231 cell viability by 40–60% .

    • GSEA analysis linked high GNG2 expression to apoptosis and immune response pathways .

Inhibition of Melanoma Metastasis

  • Mechanism: GNG2 overexpression reduces FAK phosphorylation, impairing migration and invasion in SK-Mel28 melanoma cells .

  • Experimental Evidence:

    • GNG2-overexpressing cells showed 90% reduction in invasion compared to controls .

    • Knockdown of GNG2 in A375P melanoma cells increased FAK activity and metastasis .

Prevention of Colorectal Cancer (CRC) Brain Metastasis

  • Mechanism: GNG2 suppresses PI3K/AKT/mTOR signaling, inducing G0/G1 cell cycle arrest and downregulating MMP-2 .

  • Experimental Evidence:

    • GNG2 overexpression reduced CDK4/CDK6 levels in CRC cells, inhibiting S-phase entry .

    • Low GNG2 expression correlated with advanced CRC stages and brain metastasis .

Validation and Comparative Studies

  • WB Validation: Detects GNG2 at 8 kDa in mouse paranephros lysates .

  • IHC Validation: Strong staining in human brain and stomach tissues .

  • Cross-Reactivity: Confirmed in human, mouse, and rat samples .

Significance in Biomedical Research

GNG2 antibodies have enabled breakthroughs in understanding cancer mechanisms, particularly in metastasis and therapeutic targeting. For example:

  • Diagnostic Potential: Low GNG2 expression serves as a biomarker for metastatic CRC and melanoma .

  • Therapeutic Strategies: Enhancing GNG2 expression or disrupting GNG2–MRAS interactions could treat breast cancer .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchase method or location. Please consult your local distributor for specific delivery timelines.
Synonyms
G gamma I antibody; G gamma-I antibody; GBG2_HUMAN antibody; GNG 2 antibody; Gng2 antibody; Guanine nucleotide binding protein gamma 2 antibody; Guanine nucleotide-binding protein G(I)/G(S)/G(O) subunit gamma-2 antibody
Target Names
Uniprot No.

Target Background

Function
Guanine nucleotide-binding proteins (G proteins) play a crucial role as modulators or transducers in various transmembrane signaling systems. The beta and gamma subunits are essential for GTPase activity, facilitating the replacement of GDP with GTP, and for G protein-effector interaction.
Gene References Into Functions
  1. Elevated GNG2 expression is associated with alcoholic hepatitis. PMID: 28818508
  2. G-protein betagamma subunits act as positive regulators of Kv7.4 and native vascular Kv7 channel activity. PMID: 25941381
  3. Alterations in gene expression profiling, including GPR174 and GNG2, are linked to vasovagal syncope. PMID: 25367286
  4. Research indicates that endogenous mTOR interacts with Gbetagamma. PMID: 24462769
  5. Increased protein expression levels of GNG2 alone inhibit the proliferation of malignant melanoma cells in vitro and in vivo. PMID: 23031273
  6. The presence of Gng2 and Netrin-G2 immunoreactive elements in the insular cortex, but not in the putamen, suggests a potential shared ontogeny of the claustrum and insula. PMID: 22957104
  7. Gbetagamma inhibits Epac-induced Ca 2+ elevation in melanoma cells. The interplay of Ca 2+ signaling between Gbetagamma & Epac plays a significant role in melanoma cell migration. PMID: 21679469
  8. Data suggest that the domain I-II linker region contributes significantly to voltage-dependent Gbeta1/Ggamma2 modulation of Cav2.2 calcium channels. PMID: 20181083
  9. Studies demonstrate that activation of PLCbeta(2) by alpha(q) and beta1gamma2 differs from activation by Rac2 and from each other. PMID: 20007712
  10. Research reveals that G protein inhibition of N-type calcium channels is critically dependent on two distinct but adjacent regions, each approximately 20 amino acids long, within the Gbeta subunit, as observed with Gbetas 1 and 5 and Ggamma2. PMID: 15105422
  11. Ten genes were downregulated following treatment of T-ALL cells with 0.15 and 1.5 microg/mL of metal ores at 72 h. PMID: 15747776
  12. Fission of transport carriers at the trans-Golgi network relies specifically on PLCbeta3, which is essential for activating PKCeta and PKD in that Golgi compartment, through diacylglycerol production. PMID: 17492941
  13. A signaling pathway has been identified in which a G(i)-coupled receptor specifically induces Rac and Cdc42 activation through direct interaction of Gbetagamma with FLJ00018. PMID: 18045877

Show More

Hide All

Database Links

HGNC: 4404

OMIM: 606981

KEGG: hsa:54331

STRING: 9606.ENSP00000334448

UniGene: Hs.187772

Protein Families
G protein gamma family
Subcellular Location
Cell membrane; Lipid-anchor; Cytoplasmic side.
Tissue Specificity
Expressed in fetal tissues, including testis, adrenal gland, brain, white blood cells and brain.

Q&A

What is GNG2 and what cellular functions is it involved in?

GNG2 (guanine nucleotide binding protein gamma 2) is one of the subunits of the Gβγ-dimer that forms a heterotrimeric G protein complex with a Gα-subunit. It plays crucial roles in various biological activities including cell proliferation, differentiation, invasion and angiogenesis. GNG2 is expressed in a range of fetal tissues as well as adult testis, adrenal gland, brain, and white blood cells . Recent studies have shown that GNG2 expression levels are reduced in malignant melanoma cells compared to benign melanocytic cells, suggesting a potential tumor suppressor role in certain contexts .

What types of GNG2 antibodies are available for research applications?

Several types of GNG2 antibodies are available for research applications, including polyclonal and monoclonal variants. Polyclonal antibodies like 11693-1-AP are generated from rabbits immunized with GNG2 fusion proteins and purified through antigen affinity methods . Monoclonal antibodies such as clone 4C8 are also available for more specific epitope targeting . Both types come in various formats, including unconjugated antibodies and those conjugated with reporter molecules like HRP, FITC, or biotin for specialized applications . The selection between polyclonal and monoclonal depends on the research question, with polyclonals offering broader epitope recognition and monoclonals providing higher specificity.

What are the common applications for GNG2 antibodies in laboratory research?

GNG2 antibodies are primarily utilized in several key laboratory techniques:

ApplicationTypical Dilution RangeCommon Sample Types
Western Blot (WB)1:1000-1:6000Mouse/rat brain tissue, cell lines
Immunohistochemistry (IHC)1:200-1:800Human tissue (e.g., stomach), FFPE sections
ELISAApplication-dependentVaries by experimental design

These applications allow researchers to detect and quantify GNG2 expression in various experimental contexts, enabling studies on protein expression patterns, localization, and interactions with other cellular components . When designing experiments, it's crucial to optimize antibody concentrations for each specific application and sample type to achieve optimal signal-to-noise ratios.

How does GNG2 expression correlate with cancer progression, particularly in melanoma?

Research has established a significant inverse correlation between GNG2 expression and melanoma progression. Studies have demonstrated that GNG2 expression levels in malignant melanoma cells are substantially lower compared to benign melanocytic cells in both mouse models and human samples . This pattern suggests GNG2 may function as a tumor suppressor in melanoma. More specifically, comparative analysis between A375P melanoma cells and their highly metastatic derivative A375M revealed significantly lower GNG2 expression in the more aggressive A375M line . This expression pattern correlates with increased invasive and migratory capabilities, suggesting that GNG2 downregulation may be a biomarker for melanoma progression and metastatic potential. The mechanism involves FAK (Focal Adhesion Kinase) activity modulation, where decreased GNG2 leads to increased FAK phosphorylation, promoting cellular migration and invasion capabilities .

What validation strategies should be employed to ensure GNG2 antibody specificity?

Thorough validation of GNG2 antibodies is essential for generating reliable research data. Multiple complementary approaches should be employed:

  • Orthogonal validation: Compare results across different detection methods (e.g., mass spectrometry, RNA-seq) to confirm protein expression patterns match antibody staining .

  • Independent antibody validation: Use multiple antibodies targeting different epitopes of GNG2 to confirm staining patterns. Concordant results strongly support specificity .

  • Gene knockdown/knockout controls: Implement siRNA knockdown or CRISPR-Cas9 knockout of GNG2 to demonstrate reduced or absent antibody signal. This represents the gold standard for antibody validation .

  • Overexpression systems: Express tagged GNG2 constructs and confirm co-localization with antibody staining patterns .

  • Western blot molecular weight verification: Confirm the detected band appears at the expected molecular weight of 8 kDa for GNG2 .

The Human Protein Atlas designates antibodies as "Enhanced" validation when they pass rigorous validation through at least two independent methods .

How can GNG2 antibodies be utilized to investigate G-protein signaling pathway alterations in disease models?

GNG2 antibodies enable detailed investigation of G-protein signaling alterations in disease states through several methodological approaches:

  • Comparative expression profiling: Quantify GNG2 expression levels across normal versus diseased tissues using standardized Western blot or immunohistochemistry protocols. For tissue microarrays, use consistent antibody dilutions (typically 1:200-1:800 for IHC) and scoring systems to ensure reliable comparisons .

  • Co-immunoprecipitation assays: Employ GNG2 antibodies to pull down protein complexes and examine altered interactions with Gα subunits or downstream effectors in disease models. This reveals how signaling complex formation may be disrupted in pathological states.

  • Subcellular localization studies: Use immunofluorescence with GNG2 antibodies (1:200 dilution typical) combined with confocal microscopy to track changes in the subcellular distribution of GNG2 in response to disease stimuli, revealing potential alterations in signaling compartmentalization .

  • Functional rescue experiments: In GNG2-depleted cells (via siRNA), reintroduce wild-type or mutant GNG2 constructs and use antibodies to confirm expression levels while monitoring downstream signaling pathway activation through phospho-specific antibodies (e.g., phospho-FAK) .

  • Proximity ligation assays: Combine GNG2 antibodies with antibodies against suspected interaction partners to visualize and quantify protein interactions in situ, allowing detection of altered protein complexes in disease states.

What are the optimal protocols for using GNG2 antibodies in immunohistochemistry applications?

Optimal immunohistochemistry protocols for GNG2 detection require careful consideration of several parameters:

Protocol StepRecommended ConditionsNotes
Fixation10% neutral buffered formalin, 24hOverfixation may mask epitopes
Section thickness4-5 μmBalance between tissue integrity and antibody penetration
Antigen retrievalTE buffer, pH 9.0 (primary recommendation) or citrate buffer, pH 6.0 (alternative)Heat-induced epitope retrieval, 95-98°C for 20 minutes
Blocking5% normal serum from same species as secondary antibody, 1 hour at RTCritical for reducing background
Primary antibody1:200-1:800 dilution, overnight at 4°COptimal dilution should be determined empirically for each sample type
Detection systemHRP-polymer based systems recommendedAvidin-biotin systems may show higher background
CounterstainHematoxylin, 30-60 secondsLight counterstaining improves visualization of positive signals

For human stomach tissue, which serves as a positive control for GNG2 antibodies, the suggested antigen retrieval with TE buffer pH 9.0 typically yields optimal results . The antibody concentration should be titrated for each testing system and tissue type to achieve optimal signal-to-noise ratio. Negative controls (primary antibody omission and isotype controls) should be included in all experimental runs to verify staining specificity.

How should researchers troubleshoot weak or absent GNG2 antibody signals in Western blot applications?

When encountering weak or absent GNG2 signals in Western blots, researchers should systematically troubleshoot using the following methodological approach:

  • Sample preparation optimization:

    • Ensure proper tissue/cell lysis using buffers containing appropriate detergents (RIPA or NP-40)

    • Add protease inhibitors freshly before extraction

    • For brain tissues specifically (where GNG2 is highly expressed), minimize freeze-thaw cycles and process samples rapidly

  • Protein loading and transfer adjustments:

    • Increase protein loading (50-100 μg for tissues with lower GNG2 expression)

    • Use PVDF membranes instead of nitrocellulose for better retention of low molecular weight proteins (GNG2 is only 8 kDa)

    • Adjust transfer conditions: lower voltage (25-30V) for longer time (overnight) at 4°C for efficient transfer of small proteins

  • Antibody optimization:

    • Test different antibody concentrations, starting with 1:1000 dilution and adjusting as needed

    • Extend primary antibody incubation to overnight at 4°C

    • Use 5% BSA instead of milk for blocking and antibody dilution to reduce background

    • Consider using enhanced chemiluminescence detection systems with extended exposure times

  • Controls and validation:

    • Include positive control samples (mouse or rat brain tissue are reliable positive controls)

    • If using human samples, note that expression levels vary significantly across tissues

    • Consider running a GNG2 overexpression lysate alongside experimental samples

  • Consider post-translational modifications:

    • GNG2 undergoes prenylation which can affect detection; sample preparation methods may influence epitope availability

If signal remains problematic after these optimizations, consider alternative antibody clones or epitopes, as some may perform better with specific sample types or experimental conditions.

What are the key considerations when designing GNG2 knockdown or overexpression experiments?

Designing robust GNG2 knockdown or overexpression experiments requires careful attention to several methodological aspects:

  • Knockdown strategy selection:

    • siRNA approach: Use 2-3 independent siRNA sequences targeting different regions of GNG2 mRNA to confirm specificity of observed effects

    • For transient knockdown, optimal assessment timepoint is typically 48-72 hours post-transfection

    • For stable knockdown, consider shRNA or CRISPR-Cas9 approaches with appropriate selection markers

  • Overexpression considerations:

    • Select expression vectors with appropriate promoters (CMV for high expression, tissue-specific promoters for physiological levels)

    • Include epitope tags (FLAG, HA, etc.) that don't interfere with GNG2 function for easier detection

    • For functional studies, confirm that tags do not disrupt prenylation of GNG2, which is critical for membrane localization and function

  • Controls:

    • Include scrambled siRNA/non-targeting gRNA controls for knockdown experiments

    • Use empty vector controls for overexpression studies

    • Create rescue experiments by expressing siRNA-resistant GNG2 in knockdown cells to confirm specificity

  • Verification methods:

    • Confirm knockdown/overexpression at both mRNA level (qRT-PCR) and protein level (Western blot)

    • Target 70-90% knockdown efficiency for studying loss-of-function without triggering compensatory mechanisms

    • For overexpression, aim for physiologically relevant levels (2-5 fold increase) to avoid artifacts

  • Functional readouts:

    • Based on established GNG2 functions, assess migration and invasion capabilities using transwell assays

    • Measure FAK phosphorylation levels as a key downstream readout of GNG2 activity

    • For melanoma studies, include both in vitro proliferation and in vivo xenograft growth assessments

The metastatic potential of GNG2-manipulated cells can be assessed using well-established experimental systems like the A375P/A375M comparative model, where GNG2 knockdown in A375P cells enhances migration and invasion activities with increased FAK phosphorylation, mimicking the more metastatic A375M phenotype .

How can researchers effectively use GNG2 antibodies to study melanoma progression and metastasis?

Researchers can leverage GNG2 antibodies to investigate melanoma progression and metastasis through several strategic approaches:

  • Expression profiling across progression stages:

    • Apply immunohistochemistry with GNG2 antibodies (1:200-1:800 dilution) to tissue microarrays containing benign nevi, primary melanomas, and metastatic samples

    • Develop standardized scoring systems that account for both staining intensity and percentage of positive cells

    • Correlate GNG2 expression patterns with established clinicopathological parameters and patient outcomes

  • Mechanistic studies using cell line models:

    • Compare GNG2 expression between paired cell lines with different metastatic potential (e.g., A375P vs. A375M) using Western blot (1:1000-1:6000 dilution)

    • Implement GNG2 overexpression in metastatic lines with low endogenous expression to assess functional rescue

    • Measure key downstream events including:

      • FAK phosphorylation status via phospho-specific antibodies

      • Cell migration using wound healing assays

      • Invasion capabilities using Matrigel-coated transwell chambers

      • Cell-matrix adhesion dynamics

  • In vivo metastasis models:

    • Develop GNG2-modulated melanoma cells (overexpression or knockdown)

    • Employ tail vein injection models to assess lung colonization capacity

    • Use orthotopic intradermal injections to evaluate local invasion and lymph node metastasis

    • Apply immunohistochemistry with GNG2 antibodies to analyze metastatic foci and correlate with primary tumor expression

  • Pathway integration analysis:

    • Combine GNG2 antibodies with other G-protein signaling components in multiplex immunofluorescence to map pathway alterations

    • Correlate GNG2 expression with established melanoma progression markers

    • Investigate relationships between GNG2 expression and response to targeted therapies or immunotherapies

Research has demonstrated that GNG2 overexpression inhibits metastasis in human malignant melanoma cells by decreasing FAK activity, suggesting that GNG2 could be a potential molecular target for preventing and treating melanoma metastasis .

What are the common sources of inconsistency when using GNG2 antibodies across different experimental systems?

Several factors can contribute to inconsistent results when using GNG2 antibodies across different experimental systems:

  • Antibody clone and epitope variability:

    • Different antibodies recognize distinct epitopes (amino acids 19-52, 44-62, or 1-71) which may be differentially accessible depending on experimental conditions

    • Polyclonal antibodies may show batch-to-batch variation in epitope recognition

    • Solution: Validate multiple antibody clones on the same samples and select the most consistent performer

  • Sample preparation differences:

    • Fixation methods and duration significantly impact epitope preservation, especially for IHC applications

    • For Western blot, small proteins like GNG2 (8 kDa) may be lost during standard preparation protocols

    • Solution: Standardize preparation protocols across experiments; for small proteins, use specialized extraction methods and transfer conditions

  • Expression level variations across tissues and cell types:

    • GNG2 expression varies substantially across tissues, being particularly high in brain but lower in other tissues

    • Background expression levels affect detection sensitivity requirements

    • Solution: Include appropriate positive controls (brain tissue) and concentration curves for each new system

  • Post-translational modifications:

    • GNG2 undergoes prenylation which affects membrane localization and potentially antibody accessibility

    • Phosphorylation states may alter epitope recognition

    • Solution: Characterize antibody epitope sensitivity to known modifications

  • Technical variables in detection systems:

    • Secondary antibody selection, detection reagents, and imaging parameters contribute to variability

    • Solution: Implement standardized protocols with detailed documentation of all reagents and acquisition parameters

  • Cross-reactivity considerations:

    • G-protein gamma subunits share sequence similarities that may cause cross-reactivity

    • Solution: Perform specificity controls including knockdown/knockout validation and peptide competition assays

To minimize inconsistencies, researchers should maintain detailed records of antibody lots, protocols, and imaging parameters, and implement standardized positive and negative controls across all experiments.

How should researchers interpret discrepancies between GNG2 antibody detection and mRNA expression data?

When faced with discrepancies between GNG2 protein detection using antibodies and mRNA expression data, researchers should consider several potential explanations and follow a systematic interpretation approach:

  • Post-transcriptional regulation mechanisms:

    • GNG2 mRNA may be subject to microRNA regulation affecting translation efficiency

    • Alternative splicing variants might exist that aren't detected by standard primers or antibodies

    • Approach: Examine 3'UTR regions for regulatory elements and design experiments to assess mRNA stability and translation rates

  • Protein stability and turnover differences:

    • GNG2 protein may have context-dependent stability profiles not reflected at the mRNA level

    • Heterotrimeric G-protein assembly can affect subunit stability (unincorporated subunits may be rapidly degraded)

    • Approach: Perform protein half-life studies using cycloheximide chase experiments, comparing different cell types or conditions

  • Technical considerations in detection methods:

    • Antibody sensitivity thresholds versus qPCR detection limits

    • Epitope masking in certain cellular contexts or due to protein interactions

    • Approach: Use multiple antibodies targeting different epitopes and implement quantitative Western blot with recombinant protein standards

  • Spatial or temporal expression differences:

    • mRNA and protein expression may be asynchronous due to delays in translation

    • Subcellular localization may affect detection in certain assays

    • Approach: Perform time-course studies and subcellular fractionation experiments

  • Biological interpretation framework:

    • In cancer progression contexts, discrepancies may reflect disease-specific dysregulation

    • Consider that in melanoma, post-transcriptional suppression mechanisms may be particularly relevant

    • Approach: Systematically compare normal versus diseased tissue/cells using parallel RNA-seq and proteomics approaches

For rigorous data interpretation, researchers should implement an integrated analysis approach that includes:

  • Multiple antibody validation techniques

  • Parallel RNA and protein quantification from the same samples

  • Assessment of protein-protein interactions that might affect epitope availability

  • Consideration of technical limitations in both protein and mRNA detection methods

This comprehensive approach provides a framework for distinguishing genuine biological phenomena from technical artifacts when interpreting GNG2 expression data.

What emerging applications of GNG2 antibodies are being developed in cancer research?

Emerging applications of GNG2 antibodies in cancer research are expanding beyond traditional detection methods to include several innovative approaches:

  • Biomarker development for melanoma progression: GNG2 expression profiling using validated antibodies is being developed as a potential prognostic biomarker, with decreased expression correlating with increased metastatic potential in melanoma . This application involves standardized immunohistochemical protocols with GNG2 antibodies applied to clinical specimens.

  • Therapeutic target validation: GNG2 antibodies are instrumental in validating this protein as a potential therapeutic target, particularly in melanoma where its restoration could potentially suppress metastasis . This involves using antibodies to confirm target engagement in preclinical models of experimental therapeutics designed to modulate GNG2 function.

  • Liquid biopsy applications: Development of highly sensitive detection methods for GNG2 in circulating tumor cells or exosomes using specialized antibody-based capture and detection systems. This emerging application could enable non-invasive monitoring of cancer progression.

  • Functional imaging: Labeled GNG2 antibodies or antibody fragments are being explored for in vivo imaging of tumors with altered G-protein signaling, potentially allowing for patient stratification and therapeutic response monitoring.

  • Antibody-drug conjugates: Although still in early research phases, the differential expression of GNG2 between normal and cancerous tissues suggests potential for targeted delivery approaches using GNG2 antibodies conjugated to therapeutic payloads.

These emerging applications highlight the expanding utility of GNG2 antibodies beyond traditional research tools to potential clinical applications in cancer diagnostics and therapeutics.

What are the current challenges in GNG2 antibody research that require further investigation?

Several significant challenges remain in GNG2 antibody research that warrant further investigation:

  • Epitope-specific functional correlations: Current antibodies target different epitopes (amino acids 19-52, 44-62, or 1-71) , but limited research exists on how these different epitope recognitions correlate with functional aspects of GNG2. Future studies should comprehensively map epitope accessibility across different cellular contexts.

  • Isoform-specific detection: Potential splice variants of GNG2 are inadequately characterized, and current antibodies may not distinguish between these variants. Developing isoform-specific antibodies would enable more precise functional studies.

  • Post-translational modification detection: GNG2 undergoes prenylation and potentially other modifications that affect its function. Current antibodies rarely discriminate between modified forms. Development of modification-specific antibodies would provide valuable insights into regulatory mechanisms.

  • Cross-reactivity with other G-protein gamma subunits: The high sequence similarity among G-protein gamma family members creates potential for cross-reactivity. More rigorous validation across the entire family is needed to ensure specificity.

  • Quantitative standardization across laboratories: The lack of universal standards for quantitative GNG2 detection limits cross-study comparisons. Establishing calibrated reference materials and standardized protocols would enhance data reproducibility.

  • Integration with emerging single-cell technologies: Adapting GNG2 antibodies for compatibility with single-cell proteomics and spatial transcriptomics represents a significant technical challenge that would enable unprecedented insights into heterogeneous expression patterns within tumors.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.