PLCG1 biotin-conjugated antibodies are primarily used in enzyme-linked immunosorbent assays (ELISA) for:
Quantifying PLCG1 expression levels in cell lysates or biological fluids.
Studying PLCG1’s role in signaling pathways, including EGFR-mediated H1N1 influenza virus entry and T-cell receptor activation .
Investigating PLCG1’s interaction with adaptor proteins (e.g., LAT, Grb2) in membrane microcluster formation .
Subtype-Specific Viral Entry: PLCG1 activation is essential for H1N1 (but not H3N2) influenza virus internalization, downstream of EGFR signaling .
Phase Separation in T Cells: PLCG1 stabilizes LAT-Grb2-Sos1 microclusters via SH2/SH3 domain interactions, enhancing TCR signaling .
Phosphorylation Dynamics: All three SH domains (SH2N, SH2C, SH3) of PLCG1 are required for optimal phosphorylation at Y783 during T-cell activation .
The antibody’s specificity and performance are validated through:
Immunogen Alignment: The recombinant immunogen corresponds to the PLCG1 region (1011–1290 AA) conserved across humans, mice, and rats .
Cross-Reactivity Testing: No cross-reactivity with unrelated proteins confirmed via Western blot (WB) and ELISA .
Batch Consistency: Rigorous lot-to-lot validation ensures reproducible results in target applications .
| Application | Sample | Result | Reference |
|---|---|---|---|
| ELISA | Human serum | Linear detection range: 0.1–10 ng/mL | |
| WB (indirect) | Jurkat cell lysate | Single band at ~149 kDa |
PLCG1 antibodies, including biotin-conjugated variants, are pivotal in elucidating:
Oncogenic Pathways: PLCG1 overexpression is linked to tumor metastasis via actin reorganization .
Immune Dysregulation: Aberrant PLCG1 phosphorylation contributes to autoimmune disorders and T-cell malignancies .
Viral Pathogenesis: Subtype-specific PLCG1 activation mechanisms inform antiviral drug development .
| Feature | Biotin-Conjugated (Cusabio) | Standard IgG (Boster Bio) |
|---|---|---|
| Conjugate | Biotin | Unconjugated |
| Applications | ELISA | WB, ICC, IHC |
| Reactivity | Hu, Ms, Rt | Hu, Ms, Rt |
| Price | $399 (100 µL) | $220–$399 (50–100 µL) |
| Key Advantage | High sensitivity in detection | Multiplex assay compatibility |
PLCγ1 plays critical roles in multiple cellular pathways, particularly in immune cell signaling. In T cells, PLCγ1 activation follows T cell receptor (TCR) engagement, leading to the generation of second messengers diacylglycerol (DAG) and inositol 1,4,5-trisphosphate (IP3) . These messengers trigger calcium flux and activation of protein kinase C, ultimately regulating T cell activation and differentiation.
PLCγ1 is also implicated in receptor tyrosine kinase (RTK) downstream pathways that facilitate influenza virus replication in lung epithelial cells . Furthermore, PLCγ1 works alongside PLCγ2 in B cell development, as demonstrated by studies showing that PLCγ1/PLCγ2 double deficiency blocks early B cell development at the pre-pro-B cell stage .
Biotin-conjugated antibodies against PLCγ1 enable these processes to be studied through various detection methods including immunohistochemistry, flow cytometry, and immunoprecipitation experiments. The biotin tag allows for signal amplification through streptavidin-based detection systems, enhancing sensitivity for detecting both total and phosphorylated forms of PLCγ1 in complex cellular contexts.
For optimal detection of PLCγ1 using biotin-conjugated antibodies, sample preparation should be tailored to the experimental approach:
For Flow Cytometry:
Fix cells with 4% paraformaldehyde for 15 minutes at room temperature
Permeabilize with 0.1% Triton X-100 or commercially available permeabilization buffers
Block with 2-5% serum (species different from antibody host) for 30 minutes
Incubate with biotin-conjugated anti-PLCγ1 antibody at optimized concentration (typically 5-10 μg/mL) for 45-60 minutes at 4°C
Detect using fluorophore-conjugated streptavidin (e.g., R-PE streptavidin) as demonstrated in similar protocols with other biotin-conjugated antibodies
For Immunohistochemistry:
Fix tissues in 10% neutral buffered formalin and embed in paraffin
Section tissues at 4-6 μm thickness
Perform antigen retrieval using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)
Block endogenous peroxidase with 0.3% H₂O₂ in methanol
Block non-specific binding with 5% normal serum
Incubate with biotin-conjugated anti-PLCγ1 antibody (10 μg/mL) for 1-2 hours at room temperature
If using a peroxidase-based detection system, block endogenous biotin with a biotin blocking kit
Develop signal using streptavidin-HRP and appropriate substrate
These protocols should be optimized for specific experimental conditions and tissue types being analyzed.
Validating the specificity of PLCγ1 antibodies is crucial for research integrity. A comprehensive validation approach includes:
Positive and negative controls:
Western blot analysis:
Confirm single band at the expected molecular weight (~150 kDa for PLCγ1)
Compare with commercially validated PLCγ1 antibodies
Phospho-specific validation:
Immunoprecipitation validation:
Knockout/knockdown validation:
| Validation Method | Advantages | Limitations |
|---|---|---|
| Western blot | Determines size specificity | Limited context of protein interactions |
| shRNA knockdown | Demonstrates specificity in cellular context | Incomplete knockdown may yield residual signal |
| Phospho-validation | Confirms detection of activation state | Requires careful timing after stimulation |
| Immunoprecipitation | Validates under native conditions | May miss transient interactions |
| Knockout validation | Gold standard for specificity | May be technically challenging to generate |
PLCγ1 antibodies, particularly biotin-conjugated variants, provide powerful tools for dissecting T cell signaling complexes. Advanced research applications include:
Signaling Complex Analysis:
PLCγ1 recruitment to the LAT-nucleated signaling complex is a critical event in T cell activation. Research has revealed that PLCγ1 recruitment involves not only its N-terminal SH2 domain binding to phosphorylated LAT, but also stabilization through its SH3 domain and C-terminal SH2 domain, which interact with proteins like Vav1, c-Cbl, and Slp76 .
Biotin-conjugated PLCγ1 antibodies can be used in co-immunoprecipitation experiments to pull down these protein complexes, followed by detection of interacting partners. This approach revealed that mutation of any of the SH domains abrogated co-precipitation of PLCγ1 with Slp-76, while the SH3 domain was specifically required for interaction with c-Cbl .
Domain-Specific Interactions:
For researchers investigating specific domain functions of PLCγ1, a combination of domain-specific PLCγ1 antibodies with mutagenesis approaches can reveal functional contributions of each domain. Studies have shown that all three SH domains (SH2N, SH2C, and SH3) are required for efficient phosphorylation of PLCγ1 at Y783, a critical residue for enzyme activation .
Real-Time Imaging:
Advanced imaging techniques using fluorescently-labeled streptavidin to detect biotin-conjugated PLCγ1 antibodies can track PLCγ1 recruitment to the immunological synapse in real-time. This approach, combined with fluorescent protein-tagged signaling components, demonstrated that PLCγ1 recruitment to the LAT complex requires multiple protein-protein interactions beyond simple SH2-phosphotyrosine binding .
Recent research has identified that PLCγ1 promotes phase separation of T cell signaling components , representing an emerging area of investigation. Methodological approaches to study this phenomenon include:
Fluorescence Recovery After Photobleaching (FRAP):
Label PLCγ1 using biotin-conjugated antibodies detected with fluorescent streptavidin
Measure diffusion rates within signaling clusters
Compare recovery kinetics in different cellular compartments or stimulation conditions
Proximity Ligation Assay (PLA):
Use biotin-conjugated PLCγ1 antibodies with antibodies against potential phase separation partners
Detect molecular proximity (<40 nm) as fluorescent spots
Quantify interaction frequency under different conditions
Live Cell Super-Resolution Microscopy:
Apply biotin-conjugated PLCγ1 antibodies to live cells with quantum dot-conjugated streptavidin
Track single-molecule dynamics within phase-separated domains
Analyze density and mobility parameters to characterize phase behavior
Biochemical Fractionation:
Isolate detergent-resistant membrane fractions or liquid-like condensates
Detect PLCγ1 enrichment using biotin-conjugated antibodies
Compare protein composition of these fractions under different stimulation conditions
These approaches allow researchers to investigate how PLCγ1 contributes to the formation and function of biomolecular condensates in T cell signaling, a cutting-edge area of immunological research.
PLCγ1 plays a subtype-specific role in influenza virus infection. Biotin-conjugated PLCγ1 antibodies can be employed in several advanced methodological approaches to study these mechanisms:
Subtype-Specific Viral Replication:
Research has shown that inhibition of PLCγ1 through small molecule inhibitors (U73122) or shRNA-mediated knockdown significantly decreased titers of H1N1 influenza viruses (A/WSN and A/PR8) by more than 1 log but had no effect on A/X31 (H3N2) . Biotin-conjugated PLCγ1 antibodies can be used to:
Detect changes in PLCγ1 phosphorylation status during infection with different viral subtypes
Analyze PLCγ1 recruitment to viral replication complexes through co-localization studies
Identify viral protein interactions with PLCγ1 through proximity labeling approaches
Temporal Analysis Protocol:
To investigate the temporal dynamics of PLCγ1 activation during viral infection:
Infect cells with influenza virus at MOI of 1
Fix cells at different timepoints (0, 15, 30, 60, 120, 240 minutes post-infection)
Immunostain with biotin-conjugated PLCγ1 antibodies and phospho-specific PLCγ1 antibodies
Detect with fluorescent streptavidin conjugates and analyze by confocal microscopy or flow cytometry
Quantify PLCγ1 activation kinetics in relation to viral entry and uncoating events
Antiviral Targeting Approach:
For researchers developing PLCγ1-targeting antivirals:
Treat cells with candidate compounds at non-cytotoxic concentrations
Infect with influenza viruses (both H1N1 and H3N2 subtypes)
Use biotin-conjugated PLCγ1 antibodies to monitor PLCγ1 inhibition
Correlate PLCγ1 inhibition with viral titer reduction for different influenza subtypes
Perform pathway analysis to identify compensatory mechanisms in H3N2 strains that render them insensitive to PLCγ1 inhibition
| Experimental Approach | PLCγ1 Inhibition Method | Effect on H1N1 | Effect on H3N2 |
|---|---|---|---|
| Chemical inhibition | U73122 (10 μM) | >90% reduction | No significant effect |
| Genetic knockdown | shPLCγ1 | >1 log reduction | No significant effect |
| Antibody neutralization | Anti-PLCγ1 (potential) | To be determined | To be determined |
Multi-parameter flow cytometry with biotin-conjugated PLCγ1 antibodies requires careful panel design and optimization:
Panel Design Considerations:
Fluorophore selection: When using biotin-conjugated PLCγ1 antibodies, select a streptavidin-fluorophore conjugate that minimizes spectral overlap with other markers
Phospho-epitope preservation: For phospho-PLCγ1 detection, use phosphatase inhibitors in all buffers and optimize fixation protocols
Stimulation timepoints: PLCγ1 phosphorylation peaks around 1-2 minutes after TCR stimulation , so timing is critical
Optimization Protocol:
Perform single-color controls with each fluorophore including the streptavidin-conjugate used to detect biotin-PLCγ1 antibody
Create a compensation matrix accounting for all fluorophores
Include Fluorescence Minus One (FMO) controls to set proper gates
Titrate biotin-conjugated PLCγ1 antibody to determine optimal concentration
If detecting phospho-PLCγ1, include both unstimulated and stimulated controls
Example Multi-Parameter Panel for T Cell Signaling:
| Target | Fluorophore | Purpose |
|---|---|---|
| PLCγ1 | Biotin + Streptavidin-PE | Total PLCγ1 detection |
| pPLCγ1 (Y783) | Alexa Fluor 647 | Activation status |
| CD3 | BV421 | T cell marker |
| CD4/CD8 | BV510/FITC | T cell subset identification |
| pZAP70 | PE-Cy7 | Upstream kinase activity |
| pERK | APC-Cy7 | Downstream MAPK activation |
| Cell viability | Ghost Dye™ Red 780 | Exclude dead cells |
Analysis Approach:
Calculate the ratio of phospho-PLCγ1 to total PLCγ1 within each cell to normalize for expression level differences across cell populations, allowing for more accurate assessment of activation states in heterogeneous samples.
Signal optimization for biotin-conjugated PLCγ1 antibodies requires addressing several technical factors:
Endogenous Biotin Blocking:
Tissues and some cell types contain endogenous biotin that can cause high background when using biotin-conjugated antibodies. To minimize this:
Block endogenous biotin using commercial biotin blocking kits before applying the biotin-conjugated PLCγ1 antibody
Include an avidin/biotin blocking step (incubate with avidin, wash, then incubate with biotin)
For tissues with particularly high endogenous biotin (liver, kidney), consider alternative detection methods
Fixation Optimization:
Different fixation methods can affect epitope accessibility and background:
Compare 4% paraformaldehyde, methanol, and acetone fixation to determine optimal epitope preservation
For phospho-epitopes, avoid methanol fixation which can extract phospholipids and alter membrane structure
Test crosslinking fixatives like DSP (dithiobis[succinimidyl propionate]) that better preserve protein complexes
Signal Amplification Strategies:
To enhance detection of low-abundance PLCγ1:
Use multi-layer detection: biotin-antibody → streptavidin-HRP → tyramide signal amplification
Implement catalyzed reporter deposition (CARD) for substantially increased sensitivity
For flow cytometry, compare streptavidin conjugates with different fluorophores to identify optimal brightness
Reduction of Non-Specific Binding:
To minimize background:
Include 0.1-0.3% Triton X-100 in antibody diluent to reduce hydrophobic interactions
Add 0.1-0.5 M NaCl to antibody diluent to disrupt low-affinity ionic interactions
Use protein-free blocking buffers if standard serum blocking results in high background
Detecting phosphorylated PLCγ1 presents unique challenges that require specific methodological approaches:
Critical Phosphorylation Sites:
PLCγ1 function depends on phosphorylation at specific residues, particularly Y783, which is critical for enzyme activation . Other important sites include Y775 and Y1253. Researchers should select phospho-specific antibodies based on the signaling pathway under investigation.
Rapid Phosphorylation Kinetics:
PLCγ1 phosphorylation peaks rapidly after stimulation (within 1-2 minutes) and can decline quickly . This necessitates:
Precise timing of cell fixation after stimulation
Rapid sample processing to preserve phosphorylation status
Inclusion of phosphatase inhibitors (sodium orthovanadate, sodium fluoride, β-glycerophosphate) in all buffers
Optimization Protocol for Phospho-PLCγ1 Detection:
Stimulate cells with appropriate agonist (e.g., anti-CD3/CD28 for T cells)
At precise timepoints, fix cells in 4% paraformaldehyde containing phosphatase inhibitors
Permeabilize with 0.1% Triton X-100 or 90% methanol for intracellular access
Block with 5% BSA in PBS containing phosphatase inhibitors
Incubate with phospho-specific PLCγ1 antibody
For dual detection, apply biotin-conjugated total PLCγ1 antibody
Detect with appropriate fluorophore-conjugated secondary antibodies and streptavidin
Validation Controls:
Include unstimulated cells as negative controls
Use phosphatase treatment of one sample to confirm phospho-specificity
Include cells treated with kinase inhibitors (e.g., SRC family inhibitors) that prevent PLCγ1 phosphorylation
For definitive validation, use cells expressing PLCγ1 with mutated phosphorylation sites (Y783F)
Recent research has revealed that PLCγ1, along with PLCγ2, plays crucial roles in B cell development. PLCγ1/PLCγ2 double deficiency blocks early B cell development at the pre-pro-B cell stage and renders B cell progenitors unresponsive to IL-7 . Biotin-conjugated PLCγ1 antibodies can be applied to investigate these processes:
Developmental Stage Analysis:
Isolate bone marrow cells from wild-type and PLCγ1-deficient models
Stain with biotin-conjugated PLCγ1 antibody along with B cell developmental markers (B220, CD19, IgM, CD43)
Analyze by flow cytometry to correlate PLCγ1 expression with specific developmental stages
Compare expression patterns in normal versus pathological samples
Mechanistic Studies of PLCγ1/PLCγ2 Redundancy:
Research has shown that while PLCγ2 is the predominant isoform in B cells, PLCγ1 can compensate in some contexts . To investigate this redundancy:
Use biotin-conjugated PLCγ1 antibodies alongside PLCγ2 detection in single-knockout models
Perform quantitative analysis of expression levels at different developmental stages
Correlate expression with functional readouts like calcium flux and proliferation
Analyze signaling complex formation in the presence/absence of each isoform
IL-7 Signaling Connection:
PLCγ1/PLCγ2-deficient B cell progenitors show impaired responses to IL-7 . To investigate this mechanism:
Stimulate B cell progenitors with IL-7
Fix and stain for phospho-PLCγ1 and mTOR pathway components
Analyze the temporal relationship between PLCγ1 activation and mTOR signaling
Compare signaling dynamics in wild-type versus genetic models with PLCγ abnormalities
This approach can reveal how PLCγ1 connects IL-7 receptor signaling to downstream effectors controlling B cell development and survival.
PLCγ1 dysregulation has been implicated in various cancers through its effects on cell proliferation, migration, and survival. Biotin-conjugated PLCγ1 antibodies enable several methodological approaches for cancer research:
Tissue Microarray Analysis:
Prepare tissue microarrays containing multiple tumor samples and matched normal tissues
Stain with biotin-conjugated PLCγ1 antibody and phospho-specific PLCγ1 antibodies
Detect using streptavidin-HRP and chromogenic or fluorescent substrates
Quantify expression and activation levels across different cancer types and stages
Correlate with patient outcomes to identify prognostic significance
Receptor Tyrosine Kinase Crosstalk:
PLCγ1 functions downstream of multiple receptor tyrosine kinases (RTKs) implicated in cancer. To study this crosstalk:
Stimulate cancer cells with relevant growth factors (EGF, PDGF, FGF)
At various timepoints, fix and stain for phospho-PLCγ1 and total PLCγ1
Co-stain for activated RTKs and downstream effectors
Analyze by multiparameter flow cytometry or imaging to identify pathway interactions
Compare signaling dynamics in treatment-sensitive versus resistant cell lines
Therapeutic Response Monitoring:
For evaluating PLCγ1-targeted therapeutics:
Treat cancer cells with RTK inhibitors, PLCγ1 inhibitors, or combination therapy
Monitor PLCγ1 phosphorylation status using phospho-specific antibodies
Assess total PLCγ1 levels using biotin-conjugated antibodies
Correlate molecular changes with functional outcomes (proliferation, migration, apoptosis)
Identify resistance mechanisms by examining alternative pathway activation
These approaches provide comprehensive insights into PLCγ1's role in cancer signaling networks and its potential as a therapeutic target.
PLCγ1 research continues to evolve with several methodological trends that leverage biotin-conjugated antibodies:
Single-Cell Analysis:
Single-cell technologies are transforming our understanding of cellular heterogeneity in PLCγ1 signaling:
Single-cell mass cytometry (CyTOF) using biotin-conjugated PLCγ1 antibodies with metal-conjugated streptavidin
Single-cell RNA-seq combined with protein detection (CITE-seq) to correlate PLCγ1 protein levels with transcriptional signatures
Microfluidic platforms for single-cell signaling analysis in controlled environments
Spatial Biology Approaches:
Understanding PLCγ1 function in tissue contexts requires spatial resolution:
Multiplexed immunofluorescence using biotin-conjugated PLCγ1 antibodies with cyclic staining and imaging
Imaging mass cytometry for high-parameter spatial analysis of PLCγ1 in relation to tissue microenvironment
In situ proximity ligation assays to visualize PLCγ1 interactions in intact tissues
Systems Biology Integration:
Connecting PLCγ1 signaling to broader cellular networks:
Phosphoproteomic analysis paired with PLCγ1 immunoprecipitation to map signaling networks
Mathematical modeling of PLCγ1 dynamics informed by quantitative imaging data
Integration of PLCγ1 signaling data with multi-omics datasets to identify regulatory networks
These emerging approaches are expanding our understanding of PLCγ1 biology and opening new avenues for therapeutic intervention in diseases where PLCγ1 signaling is dysregulated.
Integration of PLCγ1 antibody-based detection with complementary technologies enhances research capabilities:
CRISPR-Based Approaches:
Use CRISPR-Cas9 to generate PLCγ1 domain mutants or knockout cells
Validate mutations using biotin-conjugated PLCγ1 antibodies
Perform rescue experiments with wild-type or mutant PLCγ1 constructs
Monitor signaling consequences through antibody-based detection methods
Optogenetic Control:
Develop optogenetic tools to control PLCγ1 activation with light
Use biotin-conjugated PLCγ1 antibodies to monitor recruitment dynamics
Combine with calcium imaging to correlate PLCγ1 localization with functional outcomes
Implement in disease models to assess therapeutic potential
Structural Biology Integration:
Use antibody epitope mapping to correlate structure with function
Design conformation-specific antibodies to detect active versus inactive PLCγ1
Combine with cryo-EM or X-ray crystallography data to understand structural dynamics
Develop structure-guided therapeutic approaches targeting specific PLCγ1 conformations
By integrating these diverse technologies with antibody-based approaches, researchers can gain comprehensive insights into PLCγ1 biology across scales from molecular structure to cellular function and disease pathology.