GNG12 Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Description

FITC Conjugation Methodology

FITC conjugation involves covalent attachment of fluorescein isothiocyanate to primary amines (lysine residues) on the antibody. Key considerations include:

  • Optimal Conjugation Ratio: 3–6 FITC molecules per antibody to balance brightness and solubility .

  • Critical Steps:

    • Complete removal of sodium azide (inhibits conjugation) .

    • Immediate use of solubilized FITC due to instability .

  • Quality Control: Staining titration to evaluate brightness and background signal .

3.1. GNG12 in Gliomas

GNG12 is upregulated in gliomas and correlates with poor prognosis. Key studies include:

FindingSource
High GNG12 expression linked to immune cell infiltration (CD4+ T cells, dendritic cells) in glioblastoma (GBM) and low-grade glioma (LGG)
siRNA-mediated GNG12 knockdown reduces glioma cell proliferation and migration in vitro
Positive correlation with immune checkpoints (PD-L1, PD-L2) in GBM and LGG

3.2. Mechanistic Insights

  • Immune Microregulation: GNG12 expression inversely correlates with B cell, CD8+ T cell, and macrophage infiltration in LGG, suggesting immune evasion mechanisms .

  • Therapeutic Potential: Combined targeting of GNG12 and PD-1/PD-L1 may enhance glioma immunotherapy efficacy .

Experimental Validation

  • Immunofluorescence: FITC-conjugated GNG12 antibody used to visualize subcellular localization in glioma tissues .

  • Limitations: ProClin 300 preservative requires trained personnel for handling .

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 of receiving your order. Delivery times may vary depending on the purchase method or location. Please contact your local distributor for specific delivery details.
Synonyms
GNG12Guanine nucleotide-binding protein G(I)/G(S)/G(O) subunit gamma-12 antibody
Target Names
Uniprot No.

Target Background

Function
Guanine nucleotide-binding proteins (G proteins) play a crucial role as modulators or transducers in a variety of transmembrane signaling systems. The beta and gamma subunits are essential for GTPase activity, facilitating the exchange of GDP for GTP, and mediating G protein-effector interactions.
Database Links

HGNC: 19663

OMIM: 615405

KEGG: hsa:55970

STRING: 9606.ENSP00000360021

UniGene: Hs.431101

Protein Families
G protein gamma family
Subcellular Location
Cell membrane; Lipid-anchor; Cytoplasmic side.

Q&A

What is GNG12 and why is it an important research target?

GNG12 (G Protein gamma 12) functions as a signal transducer in G-protein coupled receptor protein signaling pathways, making it a crucial component in cellular communication networks. The protein participates in transmitting extracellular signals to intracellular effectors, regulating numerous physiological processes including sensory perception, cell growth, and hormonal responses. As a member of the G protein gamma family, GNG12 forms heterodimers with G protein beta subunits, creating the βγ complex that regulates various downstream effectors like ion channels and enzymes. The study of GNG12 is particularly important in understanding GPCR-mediated signal transduction in both normal cellular physiology and pathological conditions . Research targeting GNG12 can provide insights into fundamental cellular processes and potential therapeutic interventions for diseases involving altered G protein signaling.

What are the key applications for GNG12 Antibody, FITC conjugated?

GNG12 Antibody conjugated with FITC has several important research applications:

  • Immunofluorescence (IF) microscopy: The FITC conjugation enables direct visualization of GNG12 protein localization within cells and tissues without requiring secondary antibody steps. Recommended dilutions for IF applications range from 1:50 to 1:200 .

  • Immunohistochemistry (IHC): Used for detecting GNG12 expression in paraffin-embedded (IHC-P) and frozen tissue sections (IHC-fro) with dilutions between 1:20 and 1:200 .

  • Flow cytometry: The fluorescent properties of FITC (excitation ~495nm, emission ~519nm) make this conjugated antibody suitable for multi-parameter flow cytometry analysis.

  • Protein co-localization studies: Particularly valuable when examining the spatial relationship between GNG12 and other G protein subunits or downstream effectors.

  • ELISA and other immunological assays: While unconjugated antibodies might be preferred for some applications, FITC-conjugated antibodies can be used in specialized fluorescence-based ELISA formats .

The direct conjugation eliminates potential background issues associated with secondary antibody systems, providing cleaner signals when examining GNG12 distribution and expression patterns in both normal and pathological samples.

How does the epitope specificity affect experimental outcomes?

The epitope specificity of GNG12 antibodies critically determines experimental outcomes and interpretations. Available GNG12 antibodies target different amino acid regions, including AA 2-17, AA 25-55, and AA 30-69, each potentially yielding different results . The choice of epitope region impacts:

  • Antibody accessibility: Some epitopes may be masked in certain conformational states or protein complexes, affecting detection sensitivity. The AA 2-17 region targeted by some GNG12 antibodies is located near the N-terminus, which may have different accessibility compared to other regions.

  • Cross-reactivity profiles: Epitopes with higher sequence conservation across species show broader cross-reactivity. The AA 2-17 epitope-targeted antibodies demonstrate reactivity with human samples, while antibodies targeting AA 25-55 show broader reactivity with human, mouse, rat, dog, monkey, and rabbit samples .

  • Protein interaction detection: Antibodies targeting regions involved in protein-protein interactions may fail to recognize GNG12 when it's engaged in complexes with Gβ subunits or other interacting proteins.

  • Post-translational modification interference: If the epitope contains sites for post-translational modifications, antibody binding may be hindered when these modifications are present.

To mitigate epitope-specific limitations, researchers should validate results using antibodies targeting different epitopes of GNG12 and complement antibody-based detection with alternative approaches such as fluorescent protein tagging or mass spectrometry when possible.

What controls should be included when using GNG12 Antibody, FITC conjugated?

For rigorous experimental design with GNG12 Antibody, FITC conjugated, the following controls are essential:

  • Isotype control: Use a FITC-conjugated rabbit IgG isotype control at the same concentration as the GNG12 antibody to assess non-specific binding and establish background fluorescence levels.

  • Blocking peptide control: Pre-incubate the antibody with the immunizing peptide (competitive inhibition) to verify signal specificity. For GNG12 antibodies targeting AA 2-17, using the specific peptide sequence from Human Guanine nucleotide-binding protein G(I)/G(S)/G(O) subunit gamma-12 protein (2-17AA) would be appropriate .

  • Negative tissue/cell control: Include samples known to lack or express minimal GNG12 through genetic validation (knockout/knockdown) or tissue specificity.

  • Positive control: Include samples with confirmed GNG12 expression. Based on the available data, human, mouse, and rat samples would be suitable positive controls for antibodies with demonstrated reactivity to these species .

  • Autofluorescence control: Examine unstained samples to establish natural autofluorescence, particularly important when working with tissues containing lipofuscin, collagen, or elastin.

  • Single-color controls: When performing multi-color fluorescence experiments, include single-color controls to establish compensation settings and assess spectral overlap.

  • Absorption controls: For tissues with high levels of endogenous peroxidase or biotin, appropriate quenching steps should be included and controlled for.

Documenting these controls systematically ensures the reliability and reproducibility of results obtained with the GNG12 antibody.

How can I optimize immunofluorescence protocols for GNG12 Antibody, FITC conjugated in different tissue types?

Optimizing immunofluorescence protocols for GNG12 Antibody, FITC conjugated requires tissue-specific adjustments to maximize signal-to-noise ratio and preserve antigen integrity:

Brain tissue optimization:

  • Fixation: Use 4% paraformaldehyde for 24 hours at 4°C to preserve GNG12 epitopes while maintaining tissue architecture.

  • Antigen retrieval: Employ heat-induced epitope retrieval in citrate buffer (pH 6.0) for 20 minutes at 95°C, which has been shown to enhance detection of G protein subunits in neural tissues.

  • Permeabilization: Use 0.3% Triton X-100 for 30 minutes at room temperature to improve antibody penetration without disrupting membrane structures where GNG12 localizes.

  • Blocking: Apply 10% normal goat serum with 1% BSA for 2 hours to reduce non-specific binding, particularly important in lipid-rich brain tissue.

  • Antibody dilution: Start with 1:50 dilution and titrate as needed .

  • Counterstaining: Use DAPI (1:1000) for nuclear visualization and neuronal markers like NeuN to establish cellular context.

Immune cell optimization:

  • Fixation: Use 2% paraformaldehyde for 10 minutes to preserve surface epitopes.

  • Permeabilization: Apply 0.1% saponin instead of Triton X-100 for gentler membrane permeabilization.

  • Blocking: Use 5% normal rabbit serum with 0.5% BSA.

  • Antibody dilution: Begin with 1:100 dilution .

  • Counterstaining: Complement with lymphocyte subset markers (CD3, CD4, CD8) to identify specific immune cell populations.

Epithelial tissue optimization:

  • Fixation: Use 10% neutral buffered formalin for 12-24 hours.

  • Antigen retrieval: Perform protease-induced epitope retrieval using proteinase K (20μg/mL) for 15 minutes at 37°C.

  • Blocking: Apply 5% milk protein in PBS with 0.05% Tween-20.

  • Antibody dilution: Start with 1:200 dilution and adjust based on tissue-specific expression levels .

  • Counterstaining: Include E-cadherin or cytokeratin staining to highlight epithelial structures.

For all tissue types, include appropriate controls as outlined in question 1.4 and optimize incubation times and temperatures based on preliminary results.

What are the potential cross-reactivity concerns with GNG12 antibodies and how can they be addressed?

Cross-reactivity concerns with GNG12 antibodies stem from the high sequence homology between different G protein gamma subunits and require systematic validation approaches:

Potential cross-reactivity sources:

G Protein Subunit% Homology with GNG12Tissue Expression PatternPotential Cross-Reactivity
GNG267%Widespread, high in brainModerate to high
GNG358%Primarily neuronalModerate
GNG454%Brain, retinaLow to moderate
GNG549%UbiquitousLow
GNG761%Brain, endocrine tissuesModerate
GNG1172%Vascular tissuesHigh
GNG1345%Taste and sensory tissueLow

Validation strategies to address cross-reactivity:

  • Western blot analysis: Perform side-by-side comparison using recombinant GNG family proteins to establish antibody specificity profiles. For the AA 2-17 epitope-targeted antibodies, particular attention should be paid to GNG11 due to high sequence homology.

  • Genetic validation: Employ siRNA knockdown or CRISPR/Cas9 knockout of GNG12 to confirm signal specificity. The signal should diminish proportionally to knockdown efficiency in truly specific antibodies.

  • Peptide competition: Conduct blocking experiments using peptides corresponding to the GNG12 epitope as well as homologous regions from other gamma subunits. A specific antibody will be blocked only by the GNG12 peptide.

  • Mass spectrometry validation: Perform immunoprecipitation followed by mass spectrometry to identify all proteins captured by the antibody in complex biological samples.

  • Tissue-specific expression pattern analysis: Compare antibody staining patterns with known tissue-specific expression profiles of different gamma subunits. Discrepancies may indicate cross-reactivity.

  • Dual-labeling approaches: Co-stain with validated antibodies targeting different epitopes of GNG12 and other gamma subunits to assess signal overlap patterns.

  • Heterologous expression systems: Test antibody specificity using cells overexpressing individual gamma subunits tagged with orthogonal reporter systems.

Research using GNG12 antibodies should include appropriate cross-reactivity controls and clearly document validation steps to ensure accurate interpretation of experimental results.

How does FITC conjugation affect antibody binding kinetics and sensitivity compared to other fluorophores?

FITC conjugation to GNG12 antibodies influences binding kinetics and detection sensitivity through several mechanisms that differ from other fluorophore conjugations:

Binding kinetics effects:

Sensitivity comparison with other fluorophores:

FluorophoreExcitation/Emission (nm)Quantum YieldPhotobleaching ResistanceRelative BrightnesspH Sensitivity
FITC495/5190.85LowMediumHigh (pKa ~6.4)
Alexa Fluor 488495/5190.92HighHighLow
PE (Phycoerythrin)565/5780.84MediumVery highLow
Cy3550/5700.15MediumMediumLow
APC (Allophycocyanin)650/6600.68MediumHighLow

FITC-conjugated GNG12 antibodies show several distinct characteristics:

  • pH sensitivity: FITC fluorescence decreases significantly below pH 7.0, potentially compromising sensitivity in acidic cellular compartments. This is particularly relevant when studying GNG12 in endocytic pathways or lysosomes, where Alexa Fluor 488 would provide more reliable detection.

  • Photobleaching: FITC photobleaches more rapidly than newer generation fluorophores, limiting extended imaging sessions or repeated scanning. When quantitative comparisons of GNG12 expression levels are needed, more photostable fluorophores like Alexa Fluor 488 are preferable.

  • Tissue autofluorescence: FITC emission overlaps with tissue autofluorescence, potentially reducing signal-to-noise ratio in tissues with high levels of intrinsic fluorescence (particularly formalin-fixed tissues). For such applications, red-shifted fluorophores like Cy3 may provide better contrast.

  • Multiplexing capacity: When studying GNG12 alongside other markers, FITC's spectral characteristics limit multiplexing options compared to fluorophores with narrower emission spectra.

For optimal results, selection between FITC and alternative fluorophores should be guided by the specific experimental requirements, including pH conditions, imaging duration, and multiplexing needs.

What are the current challenges in studying GNG12 signaling networks using antibody-based approaches?

Studying GNG12 signaling networks using antibody-based approaches presents several methodological challenges that require specialized techniques to overcome:

1. Temporal dynamics detection:
G protein signaling occurs on millisecond to second timescales, while traditional antibody-based detection typically provides static snapshots. Addressing this limitation requires:

  • Development of real-time biosensors using FRET or BRET technology to monitor GNG12 interactions with binding partners

  • Live-cell antibody fragment (Fab) conjugates that can access intracellular GNG12 without fixation

  • Optimization of rapid fixation protocols (<5 seconds) to capture transient signaling states

  • Correlation with functional readouts such as calcium flux or MAPK phosphorylation

2. Distinguishing free vs. complexed GNG12:
GNG12 exists both as free monomer and in complexes with Gβ subunits and various effectors. Current antibody approaches may not discriminate between these states, necessitating:

  • Development of conformation-specific antibodies that selectively recognize different GNG12 states

  • Proximity ligation assays (PLA) to specifically detect GNG12 in complex with particular partners

  • Co-immunoprecipitation followed by quantitative mass spectrometry to determine complex composition

  • Native PAGE Western blotting to preserve protein complexes during separation

3. Post-translational modification detection:
GNG12 undergoes various post-translational modifications (PTMs) including prenylation, which affects its membrane localization and function. Studying these modifications requires:

  • Development of PTM-specific antibodies (e.g., recognizing prenylated vs. non-prenylated GNG12)

  • Use of mass spectrometry to identify and quantify specific modifications

  • Combining click chemistry approaches with antibody detection for studying lipid modifications

  • Correlation of PTM status with functional readouts of downstream signaling

4. Subcellular localization resolution:
GNG12 distributes between plasma membrane, endomembrane systems, and potentially the nucleus. Accurately mapping this distribution requires:

  • Super-resolution microscopy techniques (STORM, PALM, SIM) with properly validated antibodies

  • Combination of electron microscopy with immunogold labeling for nanometer-scale resolution

  • Subcellular fractionation approaches combined with sensitive Western blotting

  • Comparison with orthogonal techniques like proximity labeling (BioID, APEX)

5. Context-dependent signaling detection:
GNG12 signaling outcomes vary between cell types and physiological states, requiring:

  • Single-cell analysis approaches to account for cellular heterogeneity

  • Tissue-specific conditional knockout models for validation studies

  • Systems biology approaches integrating antibody-based detection with transcriptomics and proteomics

  • Development of computational models to predict signaling network responses

How can GNG12 Antibody, FITC conjugated be used in multiparameter flow cytometry?

Integrating GNG12 Antibody, FITC conjugated into multiparameter flow cytometry experiments requires careful panel design, optimization, and analysis strategies:

Panel design considerations:

  • Spectral compatibility: FITC (excitation 495nm, emission 519nm) requires appropriate compensation with other fluorochromes. Recommended compatible fluorochromes include:

    • PE (R-Phycoerythrin): Minimal spectral overlap with FITC

    • APC (Allophycocyanin): Far red emission with negligible FITC overlap

    • PE-Cy7: Provides good separation from FITC signal

    • BV421 (Brilliant Violet 421): Blue-violet excitation separated from FITC

  • Marker selection: Strategic selection of complementary markers enables comprehensive characterization of GNG12-expressing cells:

PurposeMarkerRecommended FluorochromeRationale
Lineage identificationCD3, CD19, CD14, CD56APC, PE-Cy7, BV421, APC-Cy7Identifies major immune cell populations
Activation stateCD69, CD25, HLA-DRPE, APC, BV650Correlates GNG12 with activation status
G protein signalingGβ subunitsPEExamines co-expression with binding partners
Functional readoutpERK, pAKTPE-Cy7, BV421Links GNG12 to downstream signaling
ViabilityFixable viability dyeBV510Excludes dead cells that may bind non-specifically

Sample preparation optimization:

  • Fixation protocol: Use 2% paraformaldehyde for 10 minutes at room temperature to maintain cell integrity while preserving GNG12 epitope accessibility.

  • Permeabilization strategy: For intracellular GNG12 detection, 0.1% saponin provides optimal permeabilization while minimizing epitope degradation. For membrane-associated GNG12, 0.1% Triton X-100 may provide better access to membrane-proximal epitopes.

  • Blocking strategy: Implement 5% normal rabbit serum in PBS with 0.5% BSA for 20 minutes prior to antibody staining to reduce non-specific binding.

  • Antibody titration: Perform titration experiments to determine optimal GNG12-FITC antibody concentration, typically starting at 1:50 dilution and testing 2-fold serial dilutions to identify the concentration that maximizes signal-to-noise ratio .

  • Staining buffer selection: Use pH-stabilized buffers (pH 7.4-7.6) to maximize FITC fluorescence intensity and stability.

Gating and analysis strategies:

  • Compensation setup: Use single-stained controls for each fluorochrome to establish proper compensation matrix, particularly important for FITC which may have spillover into PE channel.

  • Fluorescence minus one (FMO) controls: Essential for setting accurate GNG12 positivity thresholds, particularly in cell populations with variable autofluorescence.

  • Quantitative analysis: Consider using quantitative flow cytometry with calibration beads to establish standardized molecules of equivalent soluble fluorochrome (MESF) values for GNG12 expression.

  • Bivariate analysis: Create bivariate plots comparing GNG12-FITC signal with G protein beta subunits to assess stoichiometry and co-expression patterns.

  • Phospho-flow integration: For signaling studies, implement phospho-flow protocols to correlate GNG12 expression with activation of downstream pathways like ERK, AKT, or calcium mobilization.

  • tSNE or UMAP analysis: Apply dimensionality reduction algorithms to identify novel cell populations with distinct GNG12 expression patterns in heterogeneous samples.

This comprehensive approach enables robust integration of GNG12-FITC antibodies into multiparameter flow cytometry panels for detailed characterization of GNG12 expression patterns and correlations with cellular phenotypes and functions.

How can GNG12 Antibody, FITC conjugated be used to investigate protein-protein interactions in G protein signaling complexes?

GNG12 Antibody, FITC conjugated offers several specialized approaches for investigating protein-protein interactions within G protein signaling complexes:

Förster Resonance Energy Transfer (FRET) applications:

  • Acceptor photobleaching FRET: When pairing GNG12-FITC (donor) with antibodies against potential interaction partners conjugated to suitable acceptor fluorophores (e.g., Gβ subunits with Cy3), FRET efficiency can be measured by photobleaching the acceptor and quantifying donor dequenching. This technique provides spatial resolution of protein interactions at ~1-10nm scale.

  • Sensitized emission FRET: By comparing the emission of the acceptor when excited at donor vs. acceptor wavelengths, this approach allows dynamic measurements of GNG12 interactions in fixed or living cells without destroying the acceptor.

  • FRET optimization parameters:

    • Maintain consistent donor:acceptor ratio (ideally 1:1)

    • Perform appropriate controls including donor-only and acceptor-only samples

    • Calculate FRET efficiency using established algorithms (e.g., Clegg method)

    • Validate interactions using mutant proteins with disrupted binding interfaces

Proximity Ligation Assay (PLA) applications:

  • PLA protocol optimization for GNG12 interactions:

    • Primary antibody selection: Use GNG12-FITC alongside unconjugated antibodies against interaction partners (e.g., Gβ1-4, adenylyl cyclase, PLC-β)

    • Secondary probes: Anti-FITC and appropriate species-specific PLA probes for the interacting protein antibody

    • Signal development: Rolling circle amplification with red fluorescent oligos provides contrast against FITC signal

    • Controls: Include technical controls (omitting one primary antibody) and biological controls (using mutants with disrupted interactions)

  • Quantitative PLA analysis:

    • Measure dot number, intensity, and spatial distribution

    • Calculate interaction density per cell or subcellular compartment

    • Compare interaction profiles across different cellular conditions (stimulated vs. basal)

Co-immunoprecipitation with direct fluorescence detection:

  • Use GNG12-FITC antibody for immunoprecipitation followed by:

    • Direct fluorescence scanning of separation gels

    • Western blotting for interaction partners

    • Mass spectrometry analysis of co-immunoprecipitated proteins

  • Optimization strategies:

    • Cross-linking before lysis (e.g., DSP, formaldehyde) to stabilize transient interactions

    • Detergent selection critical for membrane protein complexes (digitonin or CHAPS preferable to stronger detergents)

    • Include appropriate negative controls (isotype control antibodies, competing peptides)

Time-resolved microscopy approaches:

  • Fluorescence Recovery After Photobleaching (FRAP):

    • Visualize GNG12-FITC dynamics in membrane microdomains

    • Compare recovery rates before and after receptor stimulation

    • Analyze diffusion coefficients to assess complex formation

  • Single-molecule tracking:

    • When used at very low concentrations, GNG12-FITC antibodies can enable tracking of individual complexes

    • Analyze trajectory patterns, diffusion rates, and confinement zones

    • Correlate with functional measures of signaling activity

These approaches provide complementary information about GNG12 interactions, with each method offering distinct advantages in terms of spatial resolution, temporal dynamics, and quantitative capacity.

What are the considerations for using GNG12 Antibody, FITC conjugated in live cell imaging experiments?

Live cell imaging with GNG12 Antibody, FITC conjugated presents unique challenges and requires specific optimization strategies to maintain cell viability while achieving sufficient signal:

Cell entry strategies:

  • Antibody fragment approaches:

    • Fab fragment preparation: Enzymatic digestion of GNG12-FITC with papain followed by purification to obtain monovalent fragments with improved cell penetration

    • scFv engineering: Recombinant single-chain variable fragments against GNG12, secondarily labeled with FITC

    • Optimization of fragment concentration: Typically 5-10μg/mL provides sufficient signal while minimizing toxicity

  • Membrane permeabilization techniques:

    • Gentle detergents: Brief treatment with 0.01% saponin allows antibody entry while maintaining cell viability

    • Pore-forming toxins: Streptolysin O at 0.2U/mL creates transient pores for antibody delivery

    • Hypotonic shock: Exposure to 50% osmolarity buffer for 30 seconds can facilitate antibody entry

    • Recovery periods: Allow 30-60 minutes in complete media after permeabilization before imaging

  • Physical delivery methods:

    • Microinjection: Precise delivery of GNG12-FITC (0.5-1mg/mL) directly into cytoplasm

    • Cell-penetrating peptides: Conjugation of TAT or polyarginine sequences to Fab fragments

    • Electroporation: Optimized voltage parameters (typically 250V/cm, 10ms pulses) for antibody delivery

Cell viability considerations:

  • Phototoxicity mitigation:

    • Light exposure: Limit excitation to <100ms per frame with >5s intervals to reduce ROS generation

    • Antifade agents: Include oxyrase (1U/mL) or vitamin C (100μM) in imaging media

    • Neutral density filters: Reduce excitation intensity to 10-30% of maximum

    • LED illumination: Prefer over mercury or xenon sources for reduced phototoxicity

  • Buffer composition optimization:

    • Use phenol red-free media to reduce background fluorescence

    • Supplement with 10mM HEPES to maintain pH stability during imaging

    • Add 1% BSA to reduce non-specific binding

    • Consider including 2-5% serum to maintain cell health during extended imaging

  • Temperature control:

    • Maintain at physiological temperature (37°C) for normal GNG12 trafficking

    • Pre-warm all solutions and the microscope stage to avoid thermal shock

    • If room temperature imaging is necessary, account for reduced protein trafficking rates

Signal optimization strategies:

  • FITC-specific considerations:

    • pH stability: Maintain imaging buffer at pH 7.4-7.6 for optimal FITC fluorescence

    • Photobleaching compensation: Use reference standards for intensity normalization

    • Background reduction: Pre-incubate cells in 0.1% Sudan Black to reduce autofluorescence

  • Imaging parameters:

    • Use high NA objectives (≥1.3) to maximize light collection

    • Set camera exposure and gain to utilize 70-80% of dynamic range

    • Apply minimal processing (2×2 binning maximum) to preserve signal fidelity

    • Establish z-step size at no more than 0.5μm for 3D acquisition

  • Temporal resolution considerations:

    • Balance temporal resolution needs with photobleaching constraints

    • For rapid G protein activation events, sacrifice spatial resolution for temporal sampling

    • Consider using spinning disk confocal for reduced photodamage during time-lapse imaging

Analysis accommodations:

  • Photobleaching correction:

    • Apply exponential decay models based on control regions

    • Use reference structures for intensity normalization

    • Consider photoactivatable fluorescent proteins as complementary approaches

  • Cell movement compensation:

    • Implement rigid or non-rigid registration algorithms

    • Use nuclear labeling (e.g., low concentration Hoechst) as registration reference

    • Apply trajectory analysis for highly motile cells

  • Quantification approaches:

    • Measure membrane/cytoplasm intensity ratios rather than absolute intensities

    • Implement ratiometric analysis when possible to control for expression level variations

    • Develop custom segmentation algorithms optimized for GNG12 subcellular distribution patterns

By systematically addressing these considerations, researchers can successfully implement live-cell imaging experiments using GNG12 Antibody, FITC conjugated while maintaining physiological relevance and quantitative rigor.

How can GNG12 Antibody, FITC conjugated be used alongside other tools to study G protein signaling dynamics?

Integrating GNG12 Antibody, FITC conjugated with complementary approaches creates powerful experimental systems for comprehensively studying G protein signaling dynamics:

Complementary genetic tools:

  • CRISPR/Cas9 genome editing:

    • Generate GNG12 knockout cell lines for antibody validation and loss-of-function studies

    • Create knock-in lines with fluorescent tags for orthogonal validation of antibody localization

    • Engineer point mutations in key GNG12 functional domains to assess antibody epitope accessibility

    • Implementation protocol: Use guide RNAs targeting non-conserved regions of GNG12, screen clones via Western blot, and validate with the GNG12-FITC antibody

  • RNA interference approaches:

    • siRNA/shRNA targeting GNG12 creates partial knockdown models for dose-response studies

    • Combine with rescue experiments using RNAi-resistant constructs to establish specificity

    • Optimization: Typically 25-50nM siRNA provides sufficient knockdown within 48-72 hours

  • Overexpression systems:

    • Tagged GNG12 constructs (FLAG, HA, HIS) provide alternative detection methods

    • Mutant GNG12 constructs (e.g., prenylation-deficient variants) help dissect membrane targeting

    • Inducible expression systems (Tet-On/Off) enable temporal control of GNG12 levels

Complementary biochemical approaches:

  • G protein activity assays:

    • GTPγS binding assays measure nucleotide exchange rates in membrane preparations

    • GTPase activity assays quantify hydrolysis rates to assess GAP protein interactions

    • [35S]GTPγS scintillation proximity assays provide high-throughput activity measurement

    • Correlation with GNG12 antibody staining: Normalize activity measures to GNG12 expression levels

  • Second messenger assays:

    • Real-time cAMP sensors (EPAC-based FRET reporters) measure adenylyl cyclase activation

    • Calcium indicators (Fluo-4, Fura-2) assess Gq-coupled signaling

    • IP3 accumulation assays detect PLC activation

    • Protocol integration: Perform live cell calcium imaging followed by fixation and GNG12-FITC staining in the same cells

  • Effector interaction assays:

    • Bioluminescence resonance energy transfer (BRET) between Gβγ and effectors

    • Split luciferase complementation assays for protein interaction dynamics

    • AlphaScreen/AlphaLISA for high-throughput interaction screening

    • Data integration: Correlate interaction strength with GNG12 expression levels determined by flow cytometry

Multiplexed imaging approaches:

  • Multi-color immunofluorescence panels:

    • Combine GNG12-FITC with antibodies against other G protein subunits (Gα, Gβ)

    • Include receptor antibodies to assess co-localization with signaling complexes

    • Add markers for subcellular compartments (caveolin-1, clathrin, Rab GTPases)

    • Recommended fluorophore combinations: GNG12-FITC + Gβ-Cy3 + Gα-Cy5 + DAPI

  • Super-resolution techniques:

    • Stimulated Emission Depletion (STED) microscopy achieves ~50nm resolution

    • Stochastic Optical Reconstruction Microscopy (STORM) reaches ~20nm resolution

    • Structured Illumination Microscopy (SIM) provides ~100nm resolution with standard fluorophores

    • Implementation strategy: Use GNG12-FITC in STED applications with appropriate depletion laser settings

  • Expansion microscopy:

    • Physical expansion of specimens by factor of 4-10x improves effective resolution

    • Protocol adaptation: Apply standard expansion protocols with additional validation of epitope preservation

    • Signal enhancement: May require additional amplification steps due to dilution effect

Functional correlation strategies:

  • Electrophysiology:

    • Patch-clamp recording of G protein-regulated ion channels

    • Protocol integration: Combine patch-clamp with GNG12-FITC imaging in fixed cells post-recording

    • Analysis approach: Correlate current amplitude with GNG12 expression level or localization

  • Cell migration assays:

    • Transwell migration studies assess chemokine receptor function

    • Wound healing assays monitor directional cell movement

    • Time-lapse microscopy tracks individual cell trajectories

    • Implementation: Fix and stain cells with GNG12-FITC antibody after migration period

  • Receptor internalization assays:

    • Surface biotinylation followed by internalization tracking

    • Flow cytometry-based endocytosis assays

    • ELISA-based receptor surface expression quantification

    • Data integration: Normalize receptor internalization rates to GNG12 expression levels

By systematically combining these approaches with GNG12-FITC antibody detection, researchers can build multidimensional datasets that connect GNG12 expression and localization with functional outcomes of G protein signaling under various physiological and pathological conditions.

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.