PLCG2 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Biological Context

PLCG2 antibodies are immunoreagents designed to detect and quantify the PLCγ2 enzyme encoded by the PLCG2 gene . This enzyme hydrolyzes phosphatidylinositol 4,5-bisphosphate (PIP2) into secondary messengers (IP3 and DAG), facilitating signal transduction in immune cells such as B cells, natural killer (NK) cells, and mast cells . Dysregulation of PLCγ2 is linked to immune dysregulation syndromes (e.g., PLAID, APLAID) and hematologic cancers .

Immune Dysregulation Studies

PLCG2 antibodies have been instrumental in characterizing mutations causing:

  • PLAID: Germline PLCG2 deletions leading to cold-induced urticaria, hypogammaglobulinemia, and recurrent infections .

  • APLAID: Gain-of-function mutations associated with systemic autoinflammation, interstitial lung disease, and enterocolitis .

Functional studies using these antibodies revealed impaired calcium flux and ERK phosphorylation in B cells with PLCG2 loss-of-function variants .

Cancer Research

In chronic lymphocytic leukemia (CLL), PLCG2 antibodies help identify acquired mutations (e.g., R665W, S707Y) that confer resistance to BTK inhibitors like ibrutinib . These mutations drive constitutive B-cell receptor signaling, promoting lymphoma progression .

Mechanistic Insights from Recent Studies

  • B-Cell Signaling: PLCG2 antibodies confirmed the enzyme’s role in B-cell survival, antibody production, and antigen receptor signaling . Knockout models show arrested B-cell maturation and increased apoptosis .

  • NK Cell Dysfunction: Antibody-based assays linked PLCG2 loss-of-function variants to impaired NK cell cytotoxicity and herpesviral susceptibility .

  • Mast Cell Activation: Studies using PLCG2 antibodies demonstrated aberrant cold-induced PLCγ2 activity in mast cells, explaining urticaria in PLAID .

Clinical and Therapeutic Implications

PLCG2 antibodies are critical for:

  • Diagnosing PLCγ2-associated immune dysregulation via immunohistochemical staining .

  • Developing targeted therapies for CLL and autoimmune diseases by monitoring PLCγ2 activation states .

Limitations and Future Directions

Current gaps include:

  • Limited data on PLCG2 antibody cross-reactivity in non-human models.

  • Need for standardized protocols to quantify PLCγ2 activity in clinical samples .

Product Specs

Form
Rabbit IgG in phosphate buffered saline, pH 7.4, containing 150mM NaCl, 0.02% sodium azide and 50% glycerol.
Lead Time
Typically, we are able to ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the purchasing method or location. For specific delivery details, please consult your local distributor.
Synonyms
1 phosphatidylinositol 4 5 bisphosphate phosphodiesterase gamma 2 antibody; 1-phosphatidylinositol-4 antibody; 5-bisphosphate phosphodiesterase gamma-2 antibody; EC 3.1.4.11 antibody; Phosphoinositide phospholipase C antibody; Phosphoinositide phospholipase C-gamma-2 antibody; Phospholipase C gamma 2 antibody; Phospholipase C, gamma 2 (phosphatidylinositol specific) antibody; Phospholipase C-gamma-2 antibody; Phospholipase C-IV antibody; PLC 2 antibody; PLC gamma 2 antibody; PLC IV antibody; PLC-gamma-2 antibody; PLC-IV antibody; Plcg2 antibody; PLCG2_HUMAN antibody
Target Names
Uniprot No.

Target Background

Function
Phospholipase C gamma 2 (PLCG2) plays a crucial role in transmembrane signaling by mediating the production of the second messenger molecules diacylglycerol (DAG) and inositol 1,4,5-trisphosphate (IP3). Activation of PLCG2 enzymes initiates a cascade of events that ultimately lead to cellular responses.
Gene References Into Functions
  1. Research has shown that mutations or polymorphisms in two putative calcium-regulated domains of PLCG2 are associated with ibrutinib-resistant chronic lymphocytic leukemia (CLL). This finding supports the notion that complex regulatory shifts within the PLCG2 protein are likely to occur during the development of resistance. PMID: 28366935
  2. Studies suggest a novel mechanism of PLCgamma activation with unique thermodynamic characteristics and assign a new regulatory role to its spPH domain. PMID: 27196803
  3. Data demonstrate that protein-altering changes occur in PLCG2, ABI3, and TREM2 genes, which are highly expressed in microglia. These alterations highlight an immune-related protein-protein interaction network implicated in Alzheimer's disease. PMID: 28714976
  4. Ocular manifestations of phospholipase-Cgamma2-associated antibody deficiency and immune dysregulation reveal mutations in the PLC[gamma]2 gene. These mutations result in aberrant function of immune cells and overproduction of interleukin-1 [beta] (IL-1[beta]). PMID: 27442322
  5. Mutations R665W and L845F are considered allomorphic rather than hypermorphic mutations of PLCG2. Rerouting of transmembrane signals originating from the BCR and converging on PLCgamma2 through Rac in ibrutinib-resistant CLL cells may offer novel drug treatment strategies to overcome ibrutinib resistance mediated by PLCG2 mutations or to prevent its development in ibrutinib-treated CLL patients. PMID: 27542411
  6. Research findings show that phospholipase Cgamma2 (PLCgamma2) is strongly expressed in B cell non-Hodgkin lymphoma, particularly in a significant subset of Diffuse large B-cell lymphoma (DLBCL). PMID: 25012946
  7. The study characterizes the effect of a missense point-mutation at R665W in PLCG2 on signaling mechanisms of ibrutinib resistance in chronic lymphocytic leukemia cells. PMID: 25972157
  8. Amarogentin inhibits platelet activation by suppressing the PLC gamma2-PKC cascade and MAPK pathway. PMID: 24868545
  9. A PLCG2 missense mutation has been identified as a risk factor in the development of steroid-sensitive nephrotic syndrome in childhood. PMID: 25349203
  10. The autoinhibitory C-terminal SH2 domain of phospholipase C-gamma2 contributes to the stabilization of B cell receptor signalosome assembly. PMID: 25227611
  11. The correlation between the upstream tyrosine kinase SYK and its target, PLCgamma2, demonstrates high predictive power and distinguishes chronic lymphocytic leukemia from healthy controls. PMID: 24489640
  12. Single-nucleotide polymorphisms in the PLCG2 gene have been linked to breast cancer risk after menopausal hormone replacement therapy. PMID: 24080446
  13. Research has identified three distinct mutations in PLCgamma2 in two patients exhibiting resistance to ibrutinib. PMID: 24869598
  14. Early Ca(2+) fluxing provides feed-forward signal amplification by promoting the anchoring of the PLCgamma2 C2 domain to phospho-SLP65. PMID: 24166973
  15. BANK1 and BLK play roles in B-cell signaling through phospholipase C gamma 2. PMID: 23555801
  16. Down-regulation of the PLCgamma2-beta-catenin pathway occurs in both mice and humans, leading to myeloid-derived suppressor cells-mediated tumor expansion. PMID: 24127488
  17. No associations were found between treatment response and the expression levels of Lyn, Syk, PLCgamma2, and ERK. PMID: 23039362
  18. Overexpression of the altered p.Ser707Tyr protein and ex vivo experiments using leukocytes from affected individuals demonstrated clearly enhanced PLCgamma2 activity. PMID: 23000145
  19. PLCgamma2 participates in T cell receptor (TCR) signal transduction and plays a role in T cell selection in a transgenic mouse model. PMID: 22837484
  20. Genomic deletions in PLCG2 cause a gain of PLCgamma(2) function, resulting in signaling abnormalities across multiple leukocyte subsets and a phenotype encompassing both excessive and deficient immune function. PMID: 22236196
  21. Data indicate a role for PLCgamma2 and Ca(2+) signaling in the modulation of MEK/ERK in IL3/GM-csf stimulated human hematopoietic stem/progenitor cells. PMID: 21506110
  22. Rac2 binding in the absence of lipid surfaces was unable to activate phospholipase C gamma 2. PMID: 21245382
  23. SYK, along with phospholipase Cgamma2, may serve as potential biomarkers for predicting dasatinib therapeutic response in patients. PMID: 20068106
  24. Research suggests that bile acid reflux present in patients with Barrett's esophagus (BE) may increase reactive oxygen species production and cell proliferation. This occurs through activation of PI-PLCgamma2, ERK2 MAP kinase, and NADPH oxidase NOX5-S, potentially contributing to the development of esophageal adenocarcinoma (EA). PMID: 20086178
  25. RTX treatment results in a time-dependent inhibition of the BCR-signaling cascade involving Lyn, Syk, PLC gamma 2, Akt, and ERK, as well as calcium mobilization. PMID: 19965664
  26. The collagen receptor glycoprotein VI and alphaIIbbeta3 trigger distinct patterns of receptor signaling in platelets, leading to tyrosine phosphorylation of PLCgamma2 (integrin alphaiibbeta3). PMID: 12049640
  27. Two tyrosine residues play a regulatory role in the activity of PLCgamma2. PMID: 12181444
  28. The full-length cDNA for human PLCgamma2 was cloned and expressed in E. coli using the expression vector pT5T. PMID: 12359094
  29. PLCG2 plays a signaling role in platelet glycoprotein Ib alpha calcium flux and cytoskeletal reorganization. PMID: 12813055
  30. In gastric cancer, protein translocation of PLCgamma2 and PKCalpha is a critical event in the process of apoptosis induction. PMID: 14606067
  31. PLC-gamma2 undergoes phosphorylation on Y753, Y759, and Y1217 in response to engagement of the B-cell receptor. PMID: 15509800
  32. PLCgamma2 is present in the majority of mediastinal B cell lymphomas. PMID: 15744341
  33. A novel mechanism of PLCgamma(2) activation by Rac GTPases has been identified, involving neither protein tyrosine phosphorylation nor PI3K-mediated generation of PtdInsP(3). PMID: 16172125
  34. Intracellular mediators and pathways activated by leptin downstream of JAK2 include phosphatidylinositol-3 kinase, phospholipase Cgamma2, and protein kinase C, as well as the p38 MAP kinase-phospholipase A(2) axis. PMID: 18000612
  35. Plasmacytoid dendritic cells express a signalosome consisting of Lyn, Syk, Btk, Slp65 (Blnk), and PLCgamma2. Triggering CD303 leads to tyrosine phosphorylation of Syk, Slp65, PLCgamma2, and cytoskeletal proteins. PMID: 18022864
  36. Rac regulates its effector phospholipase Cgamma2 through interaction with a split pleckstrin homology domain. PMID: 18728011

Show More

Hide All

Database Links

HGNC: 9066

OMIM: 600220

KEGG: hsa:5336

STRING: 9606.ENSP00000352336

UniGene: Hs.372303

Involvement In Disease
Familial cold autoinflammatory syndrome 3 (FCAS3); Autoinflammation, antibody deficiency, and immune dysregulation PLCG2-associated (APLAID)

Q&A

What is PLCG2 and why is it important in immunological research?

PLCG2 (Phospholipase C gamma 2) is an enzyme that plays a critical role in immune cell signaling pathways. It catalyzes the hydrolysis of phosphatidylinositol 4,5-bisphosphate to generate second messengers inositol 1,4,5-triphosphate (IP3) and diacylglycerol (DAG) . These messengers regulate cellular processes including calcium mobilization and protein kinase C activation, which are essential for immune cell function .

PLCG2 is particularly important in:

  • B cell receptor (BCR) signaling and development

  • Natural killer (NK) cell function

  • Myeloid cell responses including monocytes, macrophages, and dendritic cells

  • Inflammatory response regulation

Mutations in PLCG2 are associated with immune dysregulation syndromes, making it a significant target for immunological and autoinflammatory disease research .

Which validation methods should be used to confirm PLCG2 antibody specificity?

To confirm PLCG2 antibody specificity, researchers should employ multiple validation approaches:

  • Knockout validation: Compare antibody reactivity between PLCG2 wild-type and knockout cell lines to verify specific binding

  • Overexpression systems: Test antibody performance in cells transfected with PLCG2 expression vectors (e.g., EGFP-PLCG2 plasmid)

  • Western blot analysis: Confirm single band at expected molecular weight (approximately 148 kDa)

  • Immunoprecipitation followed by mass spectrometry: Verify pulled-down protein identity

  • Epitope mapping: Ensure antibody recognizes the intended region of PLCG2 protein

Multiple antibody clones recognizing different epitopes should be tested when possible to increase confidence in results, particularly for phospho-specific antibodies targeting key residues like Y753, Y759, Y1197, and Y1217 .

What are the recommended applications for PLCG2 antibodies in immune cell research?

Based on the literature, PLCG2 antibodies are valuable tools for multiple applications in immune cell research:

ApplicationRecommended DilutionCell TypesNotes
Western blot1:2000-1:8000B cells, NK cells, myeloid cellsCan detect native and phosphorylated forms
Immunohistochemistry1:50-1:500Lymphoid tissues (tonsil, spleen)Antigen retrieval with TE buffer pH 9.0 recommended
ImmunoprecipitationVaries by antibodyLymphocytes, transfected cell linesUseful for protein-protein interaction studies
Flow cytometryTypically 1:100Peripheral blood mononuclear cellsFor intracellular signaling analysis
Immunofluorescence1:100-1:500B cell lines, primary immune cellsFor subcellular localization studies

Most validated in: Daudi cells, Raji cells, Ramos cells, and primary human lymphocytes .

How can PLCG2 antibodies be used to study downstream signaling pathways in immune cells?

PLCG2 antibodies can be used to map complex signaling networks through multiple methodologies:

  • Phosphorylation status analysis:

    • Use phospho-specific antibodies targeting Y753, Y759, Y1197, and Y1217 to monitor activation

    • Combine with flow cytometry to quantify phosphorylation in heterogeneous cell populations

    • Western blot time-course experiments to determine kinetics of phosphorylation following receptor stimulation

  • Signalosome complex analysis:

    • Immunoprecipitation with PLCG2 antibodies to capture interacting proteins

    • Mass spectrometry to identify novel binding partners

    • Co-immunoprecipitation to confirm association with known partners (Syk, Btk, CD19)

    • Proximity ligation assays to visualize protein-protein interactions in situ

  • Functional readouts:

    • Calcium flux assays paired with PLCγ2 detection to correlate enzyme activation with calcium release

    • ERK phosphorylation assessment as a downstream readout

    • NF-κB and NFAT pathway activation monitoring through reporter assays

  • Single-cell analysis:

    • Couple with scRNA-seq to correlate protein expression with transcriptional profiles

    • Mass cytometry to simultaneously measure multiple pathway components

These approaches have been successfully used to characterize various gain-of-function and loss-of-function PLCG2 mutations and their effects on immune signaling .

What protocols are recommended for detecting both wild-type and mutant PLCG2 proteins in patient samples?

For clinical research involving PLCG2 variants, the following protocols have been validated:

Protocol for peripheral blood mononuclear cells (PBMCs):

  • Isolate PBMCs using density gradient centrifugation

  • Perform BCR stimulation (anti-IgM for 5 minutes)

  • Fix cells with paraformaldehyde (2-4%)

  • Permeabilize with methanol for phospho-epitope exposure

  • Stain with anti-PLCG2 and phospho-PLCG2 antibodies

  • Analyze by flow cytometry or imaging cytometry

For genetic variant functional assessment:

  • Generate mutant PLCG2 constructs using site-directed mutagenesis

  • Express constructs in PLCG2-deficient cell lines (e.g., DT-40 B cells)

  • Measure functional readouts:

    • B-cell receptor-induced calcium flux by flow cytometry

    • ERK phosphorylation by Western blot or flow cytometry

    • IP3 production using enzyme-linked immunosorbent assay

For patient cell immunoblotting:

  • Lyse cells in RIPA buffer with phosphatase inhibitors

  • Separate proteins on 4-20% SDS-PAGE gel

  • Transfer to PVDF membrane

  • Block and probe with anti-PLCG2 antibodies

  • Use phospho-specific antibodies to detect activation status

These methods have successfully differentiated between gain-of-function (GOF) and loss-of-function (LOF) PLCG2 variants in patients with immune dysregulation .

How do phospho-specific PLCG2 antibodies differ in application from total PLCG2 antibodies?

FeaturePhospho-specific PLCG2 AntibodiesTotal PLCG2 Antibodies
Epitope recognitionTarget specific phosphorylated residues (Y753, Y759, Y1197, Y1217)Recognize regions independent of phosphorylation status
Activation state detectionIndicate active/signaling state of PLCG2Measure total protein expression levels
Sample preparationRequire rapid fixation to preserve phosphorylation; phosphatase inhibitors essentialLess sensitive to post-collection processing
Stimulation requirementsOften require cell stimulation (e.g., BCR crosslinking) to detect signalDetectable in both resting and activated cells
Timing considerationsSignal typically transient; optimal detection window may be seconds to minutesStable detection window
Buffer requirementsSpecial buffers needed to preserve phospho-epitopesStandard buffers sufficient
ApplicationsOptimal for signaling dynamics studiesBetter for expression level assessment
Cross-reactivity concernsMay recognize similar phospho-motifs in related proteinsUsually more specific to PLCG2 regardless of activation

For functional studies of PLCG2 mutations, a combination of both phospho-specific and total PLCG2 antibodies provides the most complete characterization of signaling abnormalities .

How can PLCG2 antibodies help differentiate between PLAID and APLAID syndromes in research?

PLCG2 antibodies are crucial tools for investigating the molecular mechanisms underlying PLAID and APLAID syndromes. These approaches help differentiate between these related but distinct conditions:

Functional characterization strategies:

  • Domain-specific antibodies: Antibodies targeting the cSH2 domain can detect structural abnormalities in PLAID (typically affected by genomic deletions), while antibodies against catalytic domains help characterize APLAID (typically affected by point mutations)

  • Temperature-dependent activation analysis:

    • For PLAID research: Monitor PLCG2 activation at reduced temperatures (4-20°C)

    • For APLAID research: Assess constitutive activation at physiological temperature

    • Use phospho-specific antibodies to compare activation patterns

  • Downstream signaling profiling:

    • PLAID: Characterized by selective calcium signaling defects

    • APLAID: Shows enhanced signaling through multiple pathways (MAPK, NF-κB, NFAT)

    • Use antibody panels to monitor these pathways simultaneously

  • Cell-type specific effects:

    • Compare PLCG2 activation in B cells versus NK cells and myeloid cells

    • PLAID typically shows B cell defects with preserved myeloid signaling

    • APLAID demonstrates broader signaling dysregulation

This multimodal antibody-based approach helps classify PLCG2 variants along the phenotypic spectrum from PLAID to APLAID, which is critical for understanding genotype-phenotype correlations in these disorders .

What are the challenges in using PLCG2 antibodies to study heterozygous loss-of-function variants?

Studying heterozygous PLCG2 loss-of-function (LOF) variants presents several technical challenges that require specific methodological solutions:

  • Distinguishing mutant from wild-type protein:

    • Challenge: Both proteins are present in patient cells at similar molecular weights

    • Solution: Develop allele-specific antibodies or use epitope tagging in model systems

  • Measuring partial functional defects:

    • Challenge: Heterozygous variants often show subtle signaling abnormalities

    • Solution: Use highly quantitative readouts such as flow cytometry or phospho-flow with precise gating strategies

  • Cell type-specific effects:

    • Challenge: LOF variants may affect NK cells more than B cells

    • Solution: Compare antibody detection across multiple immune cell populations using multiparameter flow cytometry

  • Dose-dependent signaling:

    • Challenge: Loss of one functional allele may be compensated by the wild-type allele

    • Solution: Titrate stimulation conditions to reveal signaling defects at submaximal stimulation

  • Background genetic factors:

    • Challenge: Other genetic variants may influence PLCG2 phenotype

    • Solution: Use isogenic cell lines with CRISPR-engineered mutations for controlled studies

Research has shown that heterozygous PLCG2 LOF variants are associated with a novel form of immune dysregulation including humoral immune deficiency, autoinflammation, herpesvirus susceptibility, and NK cell dysfunction, highlighting the importance of developing robust methods to study these mutations .

How do researchers validate novel PLCG2 variants with functional assays using antibodies?

Validation of novel PLCG2 variants requires a systematic approach combining genetic analysis, molecular modeling, and functional studies with antibodies:

Complete validation protocol:

  • Initial screening:

    • Bioinformatic analysis of variant (conservation, CADD score, SiPhy)

    • Structural modeling of mutation's potential impact on protein domains

    • Comparison with known pathogenic variants

  • Expression system validation:

    • Generate mutant PLCG2 constructs using site-directed mutagenesis

    • Express in PLCG2-deficient cell lines (e.g., PLCG2-knockout DT-40 B cells)

    • Use anti-PLCG2 antibodies to confirm equivalent expression levels by Western blot

  • Functional readouts:

    • Calcium flux assays following BCR stimulation

    • Phospho-ERK detection by flow cytometry

    • IP3 production measurement

    • NFAT and NF-κB reporter assays (for suspected gain-of-function variants)

  • Protein-protein interaction studies:

    • Immunoprecipitation with anti-PLCG2 antibodies

    • Assess binding to upstream regulators (Syk, Btk, BLNK)

    • Evaluate autoinhibitory domain interactions in gain-of-function variants

  • Patient cell validation:

    • Compare antibody staining patterns in patient cells versus controls

    • Measure multiple signaling nodes to detect pathway dysregulation

    • Perform cell-type specific analyses (B cells, NK cells, myeloid cells)

This approach has successfully classified variants as gain-of-function, loss-of-function, or benign, correlating with specific clinical phenotypes ranging from autoinflammation to antibody deficiency .

How should experimental controls be designed when studying PLCG2 variants with antibodies?

Robust experimental design for PLCG2 variant studies requires careful consideration of controls:

Essential controls for PLCG2 antibody experiments:

  • Genetic controls:

    • PLCG2 knockout cells (negative control)

    • Wild-type PLCG2 expression (baseline control)

    • Known pathogenic variants (positive controls)

      • Gain-of-function: S707Y, L848P, M1141K for APLAID-like phenotypes

      • Loss-of-function: heterozygous LOF variants from validated cases

  • Stimulation controls:

    • Unstimulated baseline

    • Dose titration of BCR stimulation (anti-IgM)

    • Time-course sampling to capture signaling kinetics

    • Pharmacological controls (e.g., PLC inhibitors like U73122)

  • Antibody controls:

    • Isotype-matched control antibodies

    • Secondary antibody-only controls

    • Phospho-specific antibody validation with phosphatase treatment

    • Multiple antibody clones targeting different epitopes

  • Cell type controls:

    • Non-B cells for B-cell specific phenotypes

    • Cell lines with varying PLCG2 expression levels

    • Primary cells from healthy donors spanning age ranges

  • Experimental system validation:

    • Reproducibility across multiple biological replicates

    • Validation in both overexpression systems and primary cells

    • Cross-validation with functional readouts (calcium flux, pERK)

This comprehensive control strategy ensures reliable interpretation of PLCG2 antibody-based experiments, particularly when studying novel variants of uncertain significance .

What troubleshooting steps should be taken when PLCG2 antibodies show inconsistent results?

When facing inconsistent results with PLCG2 antibodies, follow this systematic troubleshooting approach:

Antibody-specific issues:

  • Verify antibody quality:

    • Check antibody validation data using knockout controls

    • Test multiple antibody lots

    • Validate with another independent antibody targeting a different epitope

  • Optimize antibody conditions:

    • Titrate antibody concentration

    • Modify incubation time and temperature

    • Try different blocking agents to reduce background

    • For phospho-antibodies, ensure rapid sample processing with phosphatase inhibitors

Sample preparation issues:

  • Cell lysis optimization:

    • Test multiple lysis buffers (RIPA, NP-40, Triton X-100)

    • Include protease and phosphatase inhibitor cocktails

    • Minimize freeze-thaw cycles of samples

  • Protein denaturation:

    • For Western blot, compare reducing vs. non-reducing conditions

    • Test different denaturation temperatures

    • For conformational epitopes, try native conditions

Technical considerations:

  • Signaling dynamics:

    • Ensure precise timing of stimulation

    • Check cell viability before stimulation

    • Control temperature during all steps (especially important for PLAID research)

  • Cell type variations:

    • PLCG2 effects may differ between cell types

    • Verify expression levels in your specific cell system

    • Consider genetic background effects

  • Antibody cross-reactivity:

    • Test specificity against related proteins (e.g., PLCG1)

    • Perform peptide competition assays

    • For phospho-antibodies, validate with phosphatase treatment

These troubleshooting steps have helped researchers resolve inconsistencies in PLCG2 antibody experiments, particularly when working with novel variants or complex signaling pathways .

How can researchers optimize protocols for studying PLCG2 phosphorylation in primary immune cells?

Optimizing PLCG2 phosphorylation detection in primary immune cells requires attention to several critical factors:

Sample collection and processing:

  • Minimize time between collection and fixation:

    • Process samples within 60 minutes of collection

    • Keep samples at physiological temperature until fixation

    • For blood samples, use heparin (not EDTA) as anticoagulant

  • Stimulation protocol optimization:

    • For B cells: anti-IgM (5-10 μg/ml) for 1-5 minutes

    • For NK cells: IL-2 priming followed by target cell exposure

    • Include positive control stimuli (e.g., PMA/ionomycin)

    • Perform kinetic analyses to identify optimal timepoints

Phospho-epitope preservation:

  • Fixation and permeabilization:

    • Fix with 1.6% paraformaldehyde for 10 minutes at 37°C

    • Permeabilize with ice-cold methanol (15 minutes at -20°C)

    • Alternative: Use commercial phospho-flow buffers (BD Phosflow, Thermo PerFix)

    • Include phosphatase inhibitors (sodium orthovanadate, sodium fluoride, β-glycerophosphate)

  • Antibody selection:

    • Use antibodies specifically validated for phospho-flow

    • Target key phosphorylation sites: Y753, Y759, Y1197, Y1217

    • Include total PLCG2 antibody to normalize for expression levels

Analysis strategies:

  • Multi-parameter analysis:

    • Include lineage markers to identify cell populations

    • Measure multiple signaling nodes simultaneously

    • Calculate phosphorylation index relative to unstimulated cells

    • Consider dimensionality reduction techniques (tSNE, UMAP) for complex datasets

  • Data normalization:

    • Include standardized control samples in each experiment

    • Use stimulation index rather than raw MFI values

    • For patient samples, analyze alongside healthy controls processed identically

These optimized protocols have successfully identified phosphorylation defects in patients with PLCG2 variants, enabling accurate classification of variants and correlation with clinical phenotypes .

How can PLCG2 antibodies be used in single-cell analysis to study immune cell heterogeneity?

PLCG2 antibodies are increasingly being integrated into single-cell analysis platforms to reveal immune cell functional heterogeneity:

Single-cell technologies with PLCG2 antibody integration:

  • Mass cytometry (CyTOF):

    • Metal-conjugated PLCG2 antibodies enable high-parameter analysis

    • Can simultaneously measure multiple phosphorylation sites

    • Allows correlation of PLCG2 activity with dozens of other proteins

    • Enables identification of rare cell populations with distinct signaling profiles

  • Single-cell proteogenomics:

    • CITE-seq approach combining antibody detection with transcriptomics

    • Correlate PLCG2 protein levels with gene expression programs

    • Identify regulatory networks associated with PLCG2 activation

    • Link genetic variants to protein expression and function

  • Imaging mass cytometry:

    • Spatial distribution of PLCG2 activation in tissue contexts

    • Visualize PLCG2 signaling within lymphoid structures

    • Map cell-cell interactions influencing PLCG2 activation

  • Phospho-flow cytometry:

    • High-throughput analysis of signaling at single-cell resolution

    • Capture signaling heterogeneity within phenotypically similar cells

    • Identify differential responses to stimulation or inhibition

    • Quantify rare cell populations with aberrant signaling

These approaches have revealed unexpected heterogeneity in PLCG2 signaling among seemingly homogeneous cell populations and identified distinct signaling signatures associated with disease states like PLAID/APLAID .

What approaches can detect structural changes in PLCG2 protein conformation using antibodies?

Detecting PLCG2 conformational changes is critical for understanding activation mechanisms, particularly for gain-of-function mutations. Several antibody-based approaches can reveal these structural alterations:

  • Conformation-specific antibodies:

    • Antibodies that specifically recognize active vs. inactive conformations

    • Can detect spontaneous activation in gain-of-function variants

    • Useful for monitoring the autoinhibited state disruption

  • Domain-exposure assays:

    • Antibodies targeting normally hidden epitopes that become exposed upon activation

    • Particularly useful for autoinhibitory domain interactions

    • Can detect disrupted interdomain interactions in variants like D993Y

  • FRET-based approaches:

    • Fluorophore-conjugated antibodies against different PLCG2 domains

    • Measure FRET efficiency as indicator of domain proximity

    • Can detect conformational changes in live cells

    • Allows real-time monitoring of activation dynamics

  • Limited proteolysis combined with epitope-specific antibodies:

    • Controlled digestion reveals differential domain accessibility

    • Epitope-specific antibodies detect fragments

    • Compare proteolytic patterns between wild-type and mutant PLCG2

    • Reveals structural differences not apparent in native protein

  • Cross-linking mass spectrometry with immunoprecipitation:

    • Chemical cross-linking captures domain interactions

    • Immunoprecipitation with PLCG2 antibodies enriches the protein

    • Mass spectrometry identifies cross-linked peptides

    • Maps intramolecular interactions altered in disease variants

These methods have successfully demonstrated how mutations like D993Y alter PLCG2 conformation by disrupting autoinhibitory interactions between catalytic and regulatory domains, leading to constitutive enzyme activation in APLAID syndrome .

How can PLCG2 antibodies be used to study its role in neuroinflammation and neurodegenerative diseases?

Recent research has implicated PLCG2 in neuroinflammation and neurodegenerative disorders, creating demand for specialized antibody applications in this emerging field:

Neuroimmunological applications of PLCG2 antibodies:

  • Microglia-specific PLCG2 analysis:

    • Multiplex immunofluorescence combining PLCG2 with microglial markers (IBA1, TMEM119)

    • Flow cytometry of isolated microglia from brain tissue

    • Phospho-specific antibodies to assess activation state in disease models

    • Compare activation patterns in different brain regions

  • Neuroinflammation assessment:

    • Track PLCG2 activation in response to amyloid-β and tau pathology

    • Co-staining with inflammatory markers to correlate with microglial activation

    • Examine relationship with TREM2 signaling pathway

    • Monitor responses to immunomodulatory therapies

  • Genetic variant functional characterization:

    • Antibody-based functional studies of P522R variant (protective against Alzheimer's)

    • Compare signaling properties with antibodies against multiple phosphorylation sites

    • Assess impact on phagocytosis and inflammatory cytokine production

    • Validate findings in iPSC-derived microglia

  • Brain tissue analysis:

    • Post-mortem studies comparing PLCG2 expression and activation

    • Regional variations across neurodegenerative disease progression

    • Correlation with pathological hallmarks (plaques, tangles, inflammation)

    • Co-localization with other microglial function markers

These applications help elucidate how PLCG2 signaling in microglia contributes to neuroinflammation and potentially influences neurodegenerative disease progression, opening new avenues for therapeutic development targeting this pathway .

How should researchers interpret discrepancies between phospho-PLCG2 antibody results and functional calcium flux assays?

When phospho-PLCG2 antibody results conflict with calcium flux measurements, systematic analysis is required:

Potential causes and interpretations:

  • Temporal dynamics discrepancies:

    • PLCG2 phosphorylation typically peaks 1-5 minutes after stimulation

    • Calcium flux occurs within seconds and may resolve before peak phosphorylation

    • Solution: Perform detailed time-course with both readouts simultaneously

    • Interpretation: Different kinetics rather than contradictory results

  • Threshold effects:

    • Small changes in phosphorylation may produce large calcium responses

    • Non-linear relationship between phosphorylation and functional output

    • Solution: Dose-response studies with quantitative analysis of both parameters

    • Interpretation: System may operate at different points on response curve

  • Alternative pathway activation:

    • Calcium flux may occur through PLCG2-independent mechanisms

    • IP3 receptor sensitization can amplify small PLCG2 signals

    • Solution: Use IP3 receptor blockers or PLCG2 inhibitors to assess contribution

    • Interpretation: Multiple pathways contribute to observed calcium response

  • Cell type-specific factors:

    • Buffering capacity and calcium channel expression vary between cell types

    • Signaling thresholds differ in primary cells versus cell lines

    • Solution: Compare multiple cell models and primary cells

    • Interpretation: Context-dependent signaling relationships

  • Technical considerations:

    • Antibody may recognize only subset of phosphorylated forms

    • Calcium dye loading efficiency introduces variability

    • Solution: Use multiple antibodies against different phospho-sites

    • Interpretation: Methodological limitations rather than biological discrepancy

These analytical approaches have helped researchers resolve apparent contradictions between phosphorylation data and functional outcomes in studies of PLCG2 variants associated with immune dysregulation .

What are the key considerations when using PLCG2 antibodies for longitudinal studies of patient samples?

Longitudinal studies using PLCG2 antibodies require special attention to ensure comparable results across timepoints:

Critical factors for consistent longitudinal analysis:

  • Sample standardization:

    • Process samples within consistent timeframe after collection

    • Standardize anticoagulant and storage conditions

    • Use identical cell isolation protocols across timepoints

    • Include reference control samples in each experimental batch

  • Antibody lot consistency:

    • Purchase sufficient antibody from single lot for entire study

    • Alternatively, perform lot-to-lot normalization with reference samples

    • Document detailed antibody information for each experiment

    • Maintain consistent antibody storage conditions

  • Stimulation protocol standardization:

    • Prepare master stock of stimulation reagents

    • Use consistent stimulation strength and duration

    • Control temperature precisely during stimulation

    • Document exact timing between steps

  • Instrumentation calibration:

    • For flow cytometry, use calibration beads at each session

    • Document PMT voltages and compensation matrices

    • For Western blot, include standard curve samples

    • For microscopy, use reference slides for intensity calibration

  • Data normalization strategies:

    • Include internal control samples at each timepoint

    • Calculate fold change rather than absolute values

    • Consider using stimulation index (stimulated/unstimulated)

    • For flow cytometry, convert to molecules of equivalent soluble fluorochrome (MESF)

  • Clinical correlation:

    • Document concurrent medications and clinical activity

    • Account for potential confounding factors (infections, procedures)

    • Consider seasonal variations in immune parameters

    • Correlate with other laboratory values (e.g., immunoglobulin levels)

These approaches have enabled successful longitudinal tracking of PLCG2 signaling in patients with PLAID/APLAID during disease flares and treatment interventions .

How can researchers distinguish between primary PLCG2 defects and secondary dysregulation in complex immune disorders?

Differentiating primary PLCG2 abnormalities from secondary effects requires a multi-faceted approach:

Differential diagnostic strategies:

  • Genetic-functional correlation:

    • Confirm presence of PLCG2 variant with sequencing

    • Test variant's functional impact in isolation using cell models

    • Compare patient cells with engineered cell lines expressing the variant

    • Assess if functional phenotype segregates with variant in family studies

  • Pathway dissection:

    • Examine upstream regulators of PLCG2 (Syk, Btk, BLNK)

    • Test multiple downstream effectors (calcium, ERK, NFAT, NF-κB)

    • Use inhibitors to block specific pathway components

    • Determine if abnormality is restricted to PLCG2-dependent pathways

  • Cell type specificity:

    • Compare PLCG2 signaling across multiple immune cell types

    • Assess if abnormality is present in all PLCG2-expressing cells

    • Pattern of cell-specific effects may distinguish primary from secondary effects

    • NK cell abnormalities are particularly informative for PLCG2 defects

  • Kinetic analysis:

    • Primary PLCG2 defects typically affect early signaling events

    • Secondary dysregulation may involve compensatory changes

    • Time-course experiments can distinguish immediate from delayed effects

    • Recovery kinetics after stimulation may differ

  • Response to targeted therapy:

    • Primary PLCG2 defects may respond to PLC inhibitors or downstream blockers

    • Secondary dysregulation might respond to broader immunomodulation

    • Therapeutic response pattern can provide diagnostic insights

    • Ex vivo drug testing can predict clinical responses

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.