PLCG1 Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to PLCG1 Antibody

PLCG1 antibodies are immunodetection tools targeting phospholipase C gamma 1 (PLCG1), a 148–150 kDa enzyme critical for intracellular signaling via hydrolysis of phosphatidylinositol 4,5-bisphosphate (PIP2) into inositol trisphosphate (IP3) and diacylglycerol (DAG) . These antibodies enable researchers to study PLCG1’s roles in cancer progression, immune regulation, and neuronal development by detecting its expression, phosphorylation status, and subcellular localization .

Cancer Biomarker Studies

  • Expression Correlation: Elevated PLCG1 levels in IDH wild-type gliomas correlate with poor prognosis and tumor invasiveness . siRNA-mediated PLCG1 knockdown reduces migration and proliferation in LGG cell lines (SW1088, SW1783) .

  • Therapeutic Targeting: PLCG1 amplification in RAS wild-type colorectal cancer associates with cetuximab resistance. In vivo studies show PLCG1 inhibition suppresses metastasis .

Immune Dysregulation

  • Gain-of-Function Variants: The p.S1021F variant in PLCG1 causes hyperactive ERK/NF-κB pathways, leading to autoimmune and inflammatory phenotypes. IP3 production increases 1.5–2 fold in patient-derived T cells .

Neuronal Development

PLCG1 antibodies have been used to study synaptic plasticity and actin cytoskeleton dynamics in neuronal cells, though this application is less documented in the reviewed literature .

Validation and Quality Control

  • Knockout Validation: Abcam’s ab109501 demonstrates specificity via loss of signal in PLCG1 knockout HEK-293T cells .

  • Dilution Optimization: Proteintech’s 28362-1-AP recommends 1:1000–1:4000 for WB and 1:200–1:800 for IF/ICC .

  • Phosphorylation-Specific Detection: Cell Signaling’s #4510 antibody detects Ser1248 phosphorylation, a key activation marker .

Technical Considerations

  • Storage: Most antibodies are stable at -20°C in PBS with 50% glycerol and 0.02% sodium azide .

  • Cross-Reactivity: Affinity’s AF6210 shows broad reactivity across mammals, while Proteintech’s 28362 is human-specific .

  • Buffer Compatibility: Avoid repeated freeze-thaw cycles; aliquot volumes ≤20 µl contain stabilizing BSA .

Emerging Therapeutic Insights

PLCG1 is a promising target for small-molecule inhibitors (e.g., PLCγ1 inhibitor U73122) and antibody-drug conjugates. Preclinical studies show that blocking PLCG1 activity reverses NF-κB hyperactivation in immune dysregulation and enhances cetuximab efficacy in colorectal cancer .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days after receiving them. Delivery time may vary depending on your location and shipping method. Please contact your local distributor for specific delivery details.
Synonyms
1 phosphatidyl D myo inositol 4 5 bisphosphate antibody; 1 phosphatidylinositol 4 5 bisphosphate phosphodiesterase gamma 1 antibody; 1-phosphatidylinositol-4,5-bisphosphate phosphodiesterase gamma-1 antibody; Inositoltrisphosphohydrolase antibody; Monophosphatidylinositol phosphodiesterase antibody; NCKAP3 antibody; Phosphatidylinositol phospholipase C antibody; Phosphoinositidase C antibody; Phosphoinositide phospholipase C antibody; Phosphoinositide phospholipase C-gamma-1 antibody; Phospholipase C 148 antibody; Phospholipase C gamma 1 antibody; Phospholipase C-gamma-1 antibody; Phospholipase C-II antibody; PLC gamma 1 antibody; PLC II antibody; PLC-148 antibody; PLC-gamma-1 antibody; PLC-II antibody; PLC1 antibody; PLC148 antibody; Plcg1 antibody; PLCG1_HUMAN antibody; PLCgamma1 antibody
Target Names
Uniprot No.

Target Background

Function
PLCG1 antibody targets phospholipase C gamma 1 (PLCG1), a key enzyme involved in signal transduction pathways. It mediates the production of diacylglycerol (DAG) and inositol 1,4,5-trisphosphate (IP3), both of which act as second messenger molecules within cells. PLCG1 plays a critical role in regulating intracellular signaling cascades. Its activation is triggered by the ligand-mediated activation of receptor-type tyrosine kinases, including PDGFRA, PDGFRB, EGFR, FGFR1, FGFR2, FGFR3, and FGFR4. Furthermore, PLCG1 is involved in actin reorganization and cell migration.
Gene References Into Functions
  1. Research indicates that FGFR3 with mutations found in SADDAN (but not FGFR3 with mutations found in TDII) affects cytoskeleton organization in chondrocytes by inducing tyrosine hyperphosphorylation of paxillin. The binding of FGFR3 to PLCG1 appears to be involved in this process. (FGFR3 = fibroblast growth factor receptor 3; SADDAN = Severe Achondroplasia with Developmental Delay and Acanthosis Nigricans; TDII = Thanatophoric Dysplasia type II) PMID: 29242050
  2. PLCgamma2 plays a crucial role in calcium flux in human corneal epithelial cells (HCECs) stimulated by Aspergillus fumigatus hyphae. Syk acts upstream of PLCgamma2 in the Dectin-1 signaling pathway. PMID: 30005593
  3. Studies have revealed a previously unrecognized role for PLC-gamma1 in the positive regulation of Zap-70 and T-cell receptor tyrosine phosphorylation. Conversely, PLC-gamma1 negatively regulated the phosphorylation of SLP-76-associated proteins, including established Lck substrate phosphorylation sites within this complex. PMID: 28644030
  4. Syk-induced signals in bone marrow stromal cell lines are mediated by phospholipase C gamma1 (PLCgamma1) in osteogenesis and PLCgamma2 in adipogenesis. PMID: 28786489
  5. PLCG1, together with ITGA4, is regulated by miR-30b in clinical samples of coronary artery cells from coronary atherosclerosis patients. PMID: 27464494
  6. The IL-2-R/Lck/PLCgamma/PKCtheta;/alphaPIX/Rac1/PYGM signaling pathway plays a pivotal role in controlling fundamental cellular processes such as T cell migration and proliferation. PMID: 27519475
  7. LAT and phospholipase C-gamma dephosphorylation by SHP-1 inhibits natural killer cell cytotoxicity. PMID: 27221712
  8. The products of PLC-gamma activity mediate the innate immune response by regulating respiratory burst, phagocytosis, cell adhesion, and cell migration. (Review) PMID: 27707630
  9. 1,25(OH)2D3 indirectly modulates the differentiation of Treg/Th17 cells by affecting the VDR/PLC-gamma1/TGF-beta1 pathway. These findings suggest that administering 1,25(OH)2D3 supplements may be a beneficial treatment for organ transplantation recipients. PMID: 28926770
  10. Research has demonstrated that PLCgamma-1 activation enhances skin cell transformation. PMID: 28574619
  11. These studies indicate that immobilized EGF increases collective keratinocyte displacement via an increase in single-cell migration persistence, resulting from altered EGFR trafficking and PLCgamma1 activation. PMID: 27025961
  12. High FLC gamma expression is associated with radioresistance in glioblastoma. PMID: 26896280
  13. High PLC gamma expression is associated with breast cancer. PMID: 28112359
  14. Research has shown that the decrease in PI(4,5)P2 level under non-stimulated conditions inhibits PTEN activity, leading to the aberrant activation of the oncoprotein Akt. In addition to defining a novel mechanism of Akt phosphorylation with important therapeutic implications, these findings also demonstrate that differential expression levels of FGFR2, Plc11, and Grb2 correlate with patient survival. PMID: 26212011
  15. The PLCgamma-1 signaling pathway plays a crucial role in the H1N1-induced inflammatory responses. Studies suggest that targeting the PLCgamma-1 signaling pathway could be a potential antiviral therapy against H1N1 by inhibiting both viral replication and excessive inflammation. PMID: 27310357
  16. These findings indicate that PP1 is recruited to the extracellular calcium-dependent E-cadherin-catenin-PIP5K1a complex in the plasma membrane to activate PIP5K1a, which is required for PLC-g1 activation leading to keratinocyte differentiation. PMID: 27340655
  17. FGFR1 dimers form a complex with its effector PLCgamma1. PMID: 26482290
  18. High PLC gamma1 expression is associated with gastric adenocarcinoma. PMID: 26811493
  19. Research has reported PLCG1 genetic alterations in angiosarcomas. PMID: 26735859
  20. The expression of PLC-gamma1 and PIKE positively correlated with the tumor differentiation of oral squamous cell carcinoma. PMID: 26464646
  21. In a transgenic mouse model, PLCgamma1 is the dominant signaling effector by which activation of TrkB promotes epilepsy. PMID: 26481038
  22. hsa-miR-665 and hsa-miR-95 were downregulated in GSRCC but upregulated in intestinal gastric adenocarcinoma. The relatively differential expression of these miRNAs negatively controls their target genes, GLI2 and PLCG1. PMID: 25964059
  23. Research provides evidence that PTPRB and PLCG1 mutations are driving events in a subset of secondary angiosarcomas. PMID: 24795022
  24. PLLG1 protein mutations are uncommon in cutaneous T-cell lymphomas. PMID: 25910029
  25. PLCgamma1 is part of the molecular mechanism. PMID: 25491205
  26. Recurrent presence of the PLCG1 S345F mutation is associated with nodal peripheral T-cell lymphomas. PMID: 25304611
  27. The degradation of zonula occludens-1 (ZO-1) and claudin-2 exhibits a significant dependence on the activation of the transient receptor potential melastatin (TRPM) 2 channel, phospholipase Cgamma1 (PLCgamma1), and the protein kinase Calpha (PKCalpha) signaling cascade. PMID: 23629676
  28. Data from structural, genetic, and mechanistic studies on the role of PLCG1 in cell biology suggest that dysfunctional forms of PLCG1 are linked to immune disorders and cancer. [REVIEW] PMID: 25456276
  29. This SOCS7 knockdown-attributed effect could be due to a precise anti-PLCg-1 role. PMID: 25162020
  30. The activation of the gamma1 isoform of phospholipase C (PLCgamma1) is critical for pressure sensing in cerebral arteries and subsequent vasoconstriction. PMID: 24866019
  31. These findings indicate that the PLCgamma1-R707Q mutation causes constitutive activation of PLCgamma1 and may represent an alternative way of activating KDR/PLCg1 signaling besides KDR activation in angiosarcomas. PMID: 25252913
  32. Research reveals that PLCG1 is genetically altered in a significant portion of Cutaneous T-cell lymphomas. PMID: 24706664
  33. A portion of PLC-gamma1 phosphorylated on tyrosine 783 is not found at LAT-containing clusters but instead is located at TCR-containing clusters. PMID: 24412752
  34. Extracellular K(+) concentration regulated the levels of activated PLC-gamma1, chromosome X, and carbachol-stimulated intracellular Ca(2+) mobilization in human endothelial cells. PMID: 24785188
  35. Increased proliferative and survival mechanisms in cutaneous T-cell lymphoma may partially depend on the acquisition of somatic mutations in PLCG1 and other genes essential for normal T-cell differentiation. PMID: 24497536
  36. PLCG1, a signal transducer of tyrosine kinases, encoded a recurrent, likely activating p.Arg707Gln missense variant in 3 of 34 cases of angiosarcoma. PMID: 24633157
  37. Phospholipase C gamma1 plays a key role in cell migration and invasion. [review] PMID: 23925006
  38. PLCgamma1 signaling is the dominant pathway in promoting limbic epileptogenesis. PMID: 24502564
  39. Metastatic outcome can be dictated by the constitutive competition between Grb2 and Plcgamma1 for the phosphorylation-independent binding site on FGFR2. PMID: 24440983
  40. A study showed that PLC-gamma directly binds c-Src through its SH2 domains, and this interaction is necessary for carbachol-mediated inhibition of NHE3 activity in Caco-2/BBe/NHE3 cells. PMID: 23703528
  41. PLC-gamma1 is highly expressed in the brain and participates in neuronal cell functions mediated by neurotrophins. (Review) PMID: 23063587
  42. High expression of PLCgamma1, and of its activated forms, is associated with a worse clinical outcome. PMID: 22847294
  43. The role of four domains of human PLCG1 defined by structural and biochemical investigation. PMID: 23063561
  44. Data indicate that Akt expression was up-regulated with high glucose and insulin in both cell lines, whereas PLCgamma expression was enhanced in colon cancer cells only. PMID: 22554284
  45. Analysis of two distinct mechanisms by which phospholipase C-gamma1 mediates epidermal growth factor-induced keratinocyte migration and proliferation. PMID: 22749651
  46. T cell receptor (TCR)-mediated proliferation is impaired in PLCgamma1/PLCgamma2 double-deficient T cells compared with PLCgamma1 single-deficient T cells. PMID: 22837484
  47. The oncogenic truncation of this region elicits conformational changes that interfere with the Vav1-mediated activation of PLCgamma1 and inhibit calcium mobilization. PMID: 22474331
  48. Report the interplay of HER2/HER3/PI3K and EGFR/HER2/PLC-gamma1 signaling in breast cancer cell migration and dissemination. PMID: 22262199
  49. Translocation of PLC-gamma 1 to the cell membrane and the associated calcium signal were enhanced only in mast cells responding to EP3 prostaglandin E2 receptor agonist sulprostone. PMID: 21798286
  50. Our approach, which is applicable to any set of interval scale traits that are heritable and exhibit evidence of phenotypic clustering, identified three new loci in or near APOC1, BRAP, and PLCG1, which were associated with multiple phenotype domains. PMID: 22022282

Show More

Hide All

Database Links

HGNC: 9065

OMIM: 172420

KEGG: hsa:5335

STRING: 9606.ENSP00000244007

UniGene: Hs.268177

Subcellular Location
Cell projection, lamellipodium. Cell projection, ruffle.

Q&A

What is PLCG1 and what cellular functions does it regulate?

PLCG1 (phospholipase C, gamma 1) is a 149 kDa enzyme that catalyzes the hydrolysis of 1-phosphatidyl-1D-myo-inositol 4,5-bisphosphate to produce 1D-myo-inositol 1,4,5-trisphosphate and diacylglycerol, two important second messengers in signal transduction pathways . PLCG1 functions as a major substrate for heparin-binding growth factor 1 (acidic fibroblast growth factor)-activated tyrosine kinase and plays critical roles in the regulation of cell growth, differentiation, migration, invasion, cell spreading, and immune responses .

The protein is found in various subcellular compartments, including the cytosol and plasma membrane, allowing it to participate in multiple signaling pathways . PLCG1 is particularly important in the intracellular transduction of receptor-mediated tyrosine kinase activators, forming complexes with receptors such as EGF and PDGF, which leads to phosphorylation of PLCG1 at specific tyrosine residues (Tyr771, 783, and 1248) . Dysregulation of PLCG1 has been implicated in several pathological conditions, including cancer, autoimmune disorders, and neurological conditions, making it an important research target .

What types of PLCG1 antibodies are available for research applications?

Several types of PLCG1 antibodies are available for research purposes, each with specific characteristics and applications:

Antibody TypeHost/IsotypeClassReactivityPrimary ApplicationsSource Example
Anti-PLCG1 (Total)Rabbit/IgGPolyclonalHumanELISAProteintech (19981-1-AP)
Phospho-PLCG1 (Ser1248)RabbitPolyclonalHuman, Mouse, Rat, MonkeyWestern BlottingCell Signaling Technology (#4510)
PLCG1 PolyclonalRabbitPolyclonalHumanWestern BlotAssay Genie (CAB21545)
Anti-PLCG1RabbitMonoclonalHuman, RatWestern BlotBoster Bio (M00677)

When selecting an antibody, researchers should consider the specific phosphorylation site they wish to detect (if applicable), the host species, the experimental application, and the species reactivity required for their research model . For detection of phosphorylation-dependent activation, phospho-specific antibodies like the Phospho-PLCγ1 (Ser1248) are essential to distinguish between inactive and active forms of the protein .

How can I verify the specificity of a PLCG1 antibody?

Verifying antibody specificity is critical for generating reliable research data. For PLCG1 antibodies, several validation approaches are recommended:

  • Western blot analysis: Confirm a single specific band at approximately 149 kDa (the expected molecular weight of PLCG1) . Multiple bands may indicate non-specific binding or protein degradation. Compare results using various cell lines known to express PLCG1, such as Jurkat, MCF-7, HepG2, SH-SY5Y (human) or C6, RH35 (rat) .

  • Positive and negative controls: Include a positive control (e.g., cell lines known to express PLCG1) and a negative control (e.g., PLCG1 knockdown cells using siRNA) .

  • Phospho-specificity validation: For phospho-specific antibodies, treat samples with and without phosphatase to confirm the antibody's specificity for the phosphorylated form of the protein .

  • Cross-reactivity testing: If the antibody is intended for use across multiple species, verify reactivity in each species separately before conducting comparative studies .

  • Immunoprecipitation followed by mass spectrometry: This approach can provide definitive evidence of antibody specificity by identifying the precise proteins being recognized.

The antibody datasheet often provides validation data, but researchers should perform their own validation in the specific experimental context to ensure reliable results .

What are the optimal conditions for using PLCG1 antibodies in Western blotting?

Successful Western blotting with PLCG1 antibodies requires careful optimization of several parameters:

Sample preparation and electrophoresis conditions:

  • Use 5-20% SDS-PAGE gels for optimal separation, as PLCG1 is a large protein (149 kDa)

  • Run at 70V (stacking gel) and 90V (resolving gel) for 2-3 hours for proper separation

  • Load approximately 30 μg of protein per lane under reducing conditions

Transfer and detection protocol:

  • Transfer proteins to a nitrocellulose membrane at 150 mA for 50-90 minutes

  • Block the membrane with 5% non-fat milk in TBS for 1.5 hours at room temperature

  • Incubate with primary antibody (e.g., anti-PLCG1) at a dilution of 1:1000 overnight at 4°C

  • Wash with TBS-0.1% Tween three times for 5 minutes each

  • Probe with an appropriate secondary antibody (e.g., goat anti-rabbit IgG-HRP)

Phospho-specific considerations:

  • For phospho-specific antibodies (e.g., Phospho-PLCγ1 Ser1248), maintain phosphorylation status by including phosphatase inhibitors in lysis buffers

  • Consider using both phospho-specific and total PLCG1 antibodies to calculate the phosphorylation ratio, which often provides more meaningful data than absolute phosphorylation levels

The expected band size for PLCG1 is approximately 149 kDa, and researchers should be vigilant about verifying this molecular weight to ensure specificity .

How should I design immunohistochemistry experiments to detect PLCG1 in tissue samples?

Immunohistochemistry (IHC) requires specific optimization for PLCG1 detection in tissue samples:

Tissue preparation and antigen retrieval:

  • Fix tissue samples with 4% paraformaldehyde to preserve protein structure

  • Perform heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) to expose antigenic sites that may be masked during fixation

Staining protocol and optimization:

  • Incubate sections with rabbit polyclonal anti-PLCG1 antibody at a 1:100 dilution in 1% bovine serum albumin (BSA)/phosphate-buffered saline (PBS) at room temperature for 1 hour

  • Counterstain with hematoxylin and mount with appropriate mounting medium

  • Include positive control tissues known to express PLCG1 (e.g., certain cancer tissues) and negative controls (primary antibody omitted)

Scoring and quantification:

  • Implement a semi-quantitative scoring system that considers both percentage of PLCG1-positive cells and staining intensity

  • Score percentage of stained cells as: 0 (0%), 1 (1–10%), 2 (11–50%), 3 (51–80%), 4 (>80%)

  • Evaluate staining intensity as: 0 (negative), 1+ (weak), 2+ (moderate), 3+ (strong)

  • Calculate final scores by multiplying the percentage score with the intensity score

This scoring system allows for quantitative comparison between different samples and experimental conditions, facilitating statistical analysis of results .

What approaches can be used to study PLCG1 gene amplification in tumor samples?

Two primary approaches are used to study PLCG1 gene amplification in tumor samples:

Fluorescence in situ hybridization (FISH):

  • Use fluorescent PLCG1 FISH probe alongside a control probe (e.g., CEP2/CDC42EP2)

  • Fix samples with 4% paraformaldehyde and immobilize on slides

  • Co-incubate overnight with FISH probes (50 nmol/L) at 37°C

  • Visualize using a fluorescence microscope and digitally capture images

  • Calculate the ratio of PLCG1 signals to control probe signals to determine amplification status

TCGA copy number variation (CNV) analysis:

  • Extract CNV data from cancer genomics databases like TCGA and cBioportal

  • Transform CNV values into segment means using the formula: Segment mean = log2(CNV/2)

  • Calculate copy number from segment means: 2 * (2^segment mean)

  • Interpret results: diploid regions have segment mean = 0, amplified regions have positive values, and deletions have negative values

  • Correlate PLCG1 copy number with mRNA expression levels using Pearson correlation analysis

These complementary approaches provide robust data on PLCG1 amplification, with FISH offering direct visualization in individual samples and TCGA analysis enabling large-scale genomic investigations across patient populations .

How is PLCG1 activity regulated through phosphorylation?

PLCG1 activity is tightly regulated through a complex phosphorylation pattern involving multiple kinases and phosphorylation sites:

Key phosphorylation sites and responsible kinases:

  • Tyrosine phosphorylation: PLCγ forms complexes with growth factor receptors (e.g., EGF and PDGF receptors), leading to phosphorylation at Tyr771, Tyr783, and Tyr1248

  • Syk kinase phosphorylates PLCγ1 at Tyr783, which is crucial for activating its enzymatic activity

  • EGFR phosphorylates PLCγ1 at Tyr783, causing a conformational change that enables interaction between the PLCγ1 SH3 domain and Akt proline-rich motifs

  • Akt subsequently phosphorylates PLCγ1 at Ser1248, further modulating its activity

Functional consequences of phosphorylation:

  • Phosphorylation at Tyr783 is particularly critical for activating PLCG1's catalytic function

  • The conformational change induced by phosphorylation exposes the catalytic domain and facilitates substrate binding

  • Sequential phosphorylation at multiple sites creates a regulatory cascade with precise temporal control

Inhibitory phosphorylation:

  • While not specific to PLCG1, studies on related phospholipase C isoforms (e.g., PLCβ3) show that phosphorylation by PKA or PKC at certain sites (e.g., Ser1105 in PLCβ3) can inhibit activity

  • This bidirectional regulation through activating and inhibitory phosphorylation provides fine-tuned control over PLCG1 signaling

The phosphorylation status of PLCG1 can be studied using phospho-specific antibodies, which recognize PLCG1 only when phosphorylated at specific residues (e.g., Phospho-PLCγ1 (Ser1248) Antibody) .

What role does PLCG1 play in cancer progression and how can this be studied?

PLCG1 has emerged as an important factor in cancer progression, with multiple mechanisms that can be investigated using various experimental approaches:

PLCG1's oncogenic mechanisms:

  • Elevated PLCG1 expression is associated with tumor growth and worse prognosis in IDH wild-type lower-grade gliomas (LGGs)

  • PLCG1 regulates cancer cell migration, invasion, and spreading, contributing to metastatic potential

  • Recurrent PLCG1 mutations causing constitutive activation have been described in various cancers

  • PLCG1 mediates intracellular signaling cascades downstream of tyrosine kinase receptors that drive proliferation

Experimental approaches to study PLCG1 in cancer:

  • Expression analysis:

    • Analyze PLCG1 mRNA and protein expression in tumor vs. normal tissues using qPCR and Western blotting

    • Correlate expression levels with clinical parameters and patient outcomes using databases like TCGA and CGGA

  • Functional studies:

    • Conduct PLCG1 knockdown using targeted siRNA (e.g., si1303) in cancer cell lines

    • Assess effects on proliferation, migration, and invasion in vitro

    • Evaluate tumor growth in mouse models following PLCG1 depletion

  • Mechanistic investigations:

    • Study PLCG1 amplification using FISH and CNV analysis

    • Investigate the relationship between PLCG1 amplification and overexpression using correlation analysis

    • Examine PLCG1 phosphorylation status in tumor samples using phospho-specific antibodies

  • Therapeutic targeting:

    • Test PLC inhibitors (e.g., U73122) on cancer cell lines in vitro and in mouse models

    • Measure tumor growth suppression and pathway inhibition as indicators of therapeutic efficacy

Research has demonstrated that higher PLCG1 expression correlates with poor clinical outcomes in certain cancers, suggesting its potential as both a prognostic biomarker and therapeutic target .

How can I overcome common challenges when using PLCG1 antibodies in Western blotting?

Researchers frequently encounter technical challenges when working with PLCG1 antibodies in Western blotting. Here are solutions to common problems:

High molecular weight protein detection issues:

  • Problem: Poor transfer of large proteins like PLCG1 (149 kDa)

  • Solutions:

    • Use a wet transfer system rather than semi-dry

    • Extend transfer time to 90 minutes at 150 mA

    • Reduce gel percentage to 5-20% to improve protein migration

    • Add SDS (0.1%) to transfer buffer to facilitate large protein transfer

Weak or absent signal:

  • Problem: Insufficient antibody binding or protein degradation

  • Solutions:

    • Optimize antibody concentration (1:1000 is typically effective)

    • Extend primary antibody incubation to overnight at 4°C

    • Include protease inhibitors in lysis buffer to prevent degradation

    • Store samples at -20°C with glycerol (50%) and sodium azide (0.02%) for stability

Multiple or non-specific bands:

  • Problem: Non-specific antibody binding

  • Solutions:

    • Increase blocking time to 1.5 hours using 5% non-fat milk

    • Wash thoroughly with TBS-0.1% Tween (3 times, 5 minutes each)

    • Use antigen affinity-purified antibodies to improve specificity

    • Validate with positive controls (e.g., Jurkat, MCF-7, HepG2, SH-SY5Y cells for human samples)

Phospho-specific detection issues:

  • Problem: Loss of phosphorylation signal

  • Solutions:

    • Include phosphatase inhibitors in all buffers

    • Avoid repeated freeze-thaw cycles

    • Prepare fresh lysates whenever possible

    • Use phosphorylation-promoting treatments in positive controls (e.g., EGF stimulation)

Recommended sample preparation:

  • Optimal cell lysis buffer: PBS with protease inhibitors, phosphatase inhibitors

  • Sample storage: -20°C in buffer containing 0.02% sodium azide and 50% glycerol (pH 7.3)

  • Protein loading: 30 μg per lane under reducing conditions

Following these troubleshooting strategies should significantly improve PLCG1 detection in Western blotting experiments.

What considerations are important when selecting and validating PLCG1 siRNAs for knockdown experiments?

Successful PLCG1 knockdown experiments require careful consideration of siRNA design, validation, and experimental controls:

siRNA design and selection:

  • Test multiple siRNA sequences targeting different regions of PLCG1 mRNA (e.g., si336, si1303, si1384)

  • Select siRNAs that avoid regions with secondary structure that might impede binding

  • Verify that siRNA sequences do not have significant homology with other genes to prevent off-target effects

  • Note that not all designed siRNAs will be effective (e.g., in published research, si336 and si1384 showed no obvious knockdown effect, while si1303 was effective)

Transfection optimization:

  • Use appropriate transfection reagents (e.g., Rfect infection reagent)

  • Replace transfection medium with fresh medium approximately 8 hours after transfection

  • Optimize cell density to ensure efficient transfection without toxicity

Validation of knockdown efficiency:

  • Assess PLCG1 mRNA levels by qRT-PCR at 24-48 hours post-transfection

  • Confirm protein knockdown by Western blot, using validated PLCG1 antibodies

  • Quantify knockdown efficiency by densitometry, normalizing to housekeeping genes/proteins

  • Monitor cell viability to ensure observed effects are due to PLCG1 knockdown rather than cytotoxicity

Essential controls:

  • Include a non-targeting siRNA control (siCtrl) with similar length and GC content

  • Use untransfected cells as an additional control

  • Consider rescue experiments (re-expression of siRNA-resistant PLCG1) to confirm specificity

  • For phosphorylation studies, include appropriate stimulation controls (e.g., growth factor treatment)

Downstream functional assays:

  • After confirming knockdown, assess effects on cell growth, migration, and invasiveness

  • Design time-course experiments to distinguish between immediate and secondary effects

  • Consider in vivo validation in appropriate animal models for cancer studies

These considerations are essential for generating reliable data on PLCG1 function through knockdown experiments while minimizing experimental artifacts and misinterpretation.

How can I design experiments to investigate PLCG1 phosphorylation dynamics in response to stimuli?

Investigating PLCG1 phosphorylation dynamics requires careful experimental design and time-resolved analysis:

Stimulus selection and optimization:

  • Choose appropriate stimuli based on cellular context (e.g., EGF, PDGF for cells expressing corresponding receptors)

  • Determine optimal stimulus concentration through dose-response experiments

  • Consider using multiple stimuli to compare pathway-specific responses

Time-course design:

  • Capture early phosphorylation events with closely spaced time points (e.g., 0, 1, 2, 5, 10, 30 minutes)

  • Include longer time points (1, 2, 6, 24 hours) to assess sustained phosphorylation

  • Prepare multiple plates simultaneously and stop reactions at specific time points to ensure consistency

Detection methods:

  • Western blotting with phospho-specific antibodies:

    • Use Phospho-PLCγ1 (Ser1248) Antibody (1:1000 dilution) for Western blotting

    • Always probe for both phospho-PLCG1 and total PLCG1 on separate blots or after stripping

    • Calculate phospho-to-total PLCG1 ratio to normalize for expression differences

  • Phospho-flow cytometry:

    • Fix cells with formaldehyde and permeabilize with methanol

    • Stain with fluorophore-conjugated phospho-specific antibodies

    • Enables single-cell analysis and detection of population heterogeneity

  • Proximity ligation assay (PLA):

    • Visualize and quantify protein interactions and phosphorylation events in situ

    • Provides spatial information about phosphorylation in different cellular compartments

Inhibitor studies to dissect pathways:

  • Use specific kinase inhibitors to block upstream regulators (e.g., EGFR inhibitors, PI3K inhibitors)

  • Include the PLC inhibitor U73122 to block downstream signaling and assess feedback mechanisms

  • Compare phosphorylation at different sites (Tyr771, Tyr783, Tyr1248) to establish phosphorylation sequence

Data analysis and presentation:

  • Plot phosphorylation kinetics as phospho/total PLCG1 ratio versus time

  • Use statistical analysis to determine significance of changes

  • Consider computational modeling to integrate complex phosphorylation dynamics

These approaches allow for comprehensive characterization of PLCG1 phosphorylation dynamics in response to various stimuli, providing insights into the temporal regulation of PLCG1 signaling.

What is the evidence for PLCG1 as a potential therapeutic target in cancer and other diseases?

Emerging research has highlighted PLCG1 as a promising therapeutic target across multiple disease contexts:

Evidence in cancer:

  • Higher PLCG1 expression correlates with tumor growth and worse prognosis in IDH wild-type lower-grade gliomas (LGGs)

  • PLCG1-targeted siRNA dramatically affects growth, migration, and invasiveness of IDH wild-type LGG cell lines

  • The PLC inhibitor U73122 significantly suppresses tumor growth of IDH wild-type LGG cell lines in vitro and in mouse models

  • PLCG1 amplification serves as a key mechanism of PLCG1 upregulation in certain cancers, suggesting genomic alteration as a driver event

Validation through multiple approaches:

  • Both in vitro and in vivo experiments demonstrate anti-tumor effects of PLCG1 inhibition

  • Genomic and transcriptomic data from large patient cohorts (TCGA and CGGA databases) verify the association between elevated PLCG1 expression and poor clinical outcomes

  • Multiple cancer cell lines show sensitivity to PLCG1 inhibition, suggesting broad relevance across cancer types

Beyond cancer:

  • PLCG1 dysregulation has been implicated in autoimmune disorders and neurological conditions

  • The enzyme's central role in signaling cascades downstream of receptor tyrosine kinases makes it relevant to diseases involving aberrant growth factor signaling

Therapeutic strategies under investigation:

  • Direct PLCG1 inhibition: PLC inhibitors like U73122 show promise in preclinical models

  • Targeting PLCG1 phosphorylation: Inhibitors of upstream kinases (e.g., Syk inhibitors) could prevent PLCG1 activation

  • Combination approaches: PLCG1 inhibition may sensitize tumors to existing therapies

  • Mutation-specific strategies: For cancers with activating PLCG1 mutations, mutation-specific inhibitors might offer precision targeting

The converging evidence from cellular studies, animal models, and patient data strongly supports PLCG1 as a viable therapeutic target, particularly in cancers where its overexpression drives disease progression .

How can multi-omics approaches be integrated to better understand PLCG1 function in different cellular contexts?

Integration of multi-omics approaches provides a comprehensive understanding of PLCG1 function across diverse cellular contexts:

Genomic approaches:

  • Analyze PLCG1 copy number variations using FISH and computational methods from databases like TCGA

  • Assess PLCG1 mutations through targeted sequencing or whole-exome sequencing

  • Examine regulatory elements through ChIP-seq and ATAC-seq to understand transcriptional control

  • Calculate segment means from CNV data using the formula: Segment mean = log2(CNV/2)

Transcriptomic analyses:

  • Quantify PLCG1 mRNA expression levels across different tissue types and disease states

  • Perform RNA-seq after PLCG1 knockdown to identify downstream gene expression changes

  • Use single-cell RNA-seq to reveal cell-type-specific PLCG1 expression patterns

  • Correlate PLCG1 expression with patient outcomes using databases like TCGA and CGGA

Proteomic integration:

  • Map the PLCG1 interactome through immunoprecipitation coupled with mass spectrometry

  • Characterize post-translational modifications, especially phosphorylation at key residues (Tyr771, Tyr783, Tyr1248)

  • Employ reverse-phase protein arrays (RPPA) to quantify PLCG1 and related proteins across large sample sets

  • Use phospho-specific antibodies to track activation states in different contexts

Metabolomic connections:

  • Measure changes in phospholipid metabolism and second messengers (IP3, DAG) following PLCG1 modulation

  • Trace calcium flux dynamics as a downstream consequence of PLCG1 activity

  • Assess metabolic pathway alterations in PLCG1-overexpressing vs. PLCG1-depleted cells

Computational integration:

  • Develop pathway models incorporating PLCG1 signaling nodes

  • Use machine learning to identify biomarkers that predict sensitivity to PLCG1 inhibition

  • Integrate multi-omics data to reveal context-dependent PLCG1 functions

Examples of successful multi-omics applications:

  • Combined genomic (CNV) and transcriptomic analyses revealed a strong correlation between PLCG1 amplification and overexpression in LGGs

  • Integration of PLCG1 expression data with patient survival information identified PLCG1 as a prognostic marker in IDH wild-type LGGs

  • Proteomic and genomic integration helped establish PLCG1 as a potential therapeutic target, validated through functional studies

This multi-layered approach allows researchers to move beyond correlative observations to mechanistic understanding, facilitating the development of context-specific interventions targeting PLCG1.

What methodological advances are needed to better study PLCG1 in complex tissue microenvironments?

Studying PLCG1 in complex tissue microenvironments presents unique challenges requiring methodological innovations:

Advanced spatial analysis technologies:

  • Multiplexed immunofluorescence/immunohistochemistry: Simultaneously detect PLCG1, its phosphorylated forms, and contextual markers (cell type, activation state) within tissue sections

  • Imaging mass cytometry (IMC)/Multiplexed ion beam imaging (MIBI): Analyze dozens of proteins simultaneously at subcellular resolution in tissues

  • Spatial transcriptomics: Map PLCG1 mRNA expression patterns while preserving spatial context

  • In situ proximity ligation assay (PLA): Visualize protein-protein interactions involving PLCG1 within intact tissues

Improved tissue models:

  • 3D organoids: Develop patient-derived organoids that better recapitulate tissue architecture

  • Tissue-on-chip: Create microfluidic systems that mimic organ-specific environments for PLCG1 functional studies

  • Co-culture systems: Establish models incorporating multiple cell types to study PLCG1 in heterotypic cellular interactions

  • Ex vivo tissue slice cultures: Maintain native tissue architecture while enabling experimental manipulation

Single-cell analysis approaches:

  • Single-cell phospho-proteomics: Measure PLCG1 phosphorylation at the single-cell level to capture heterogeneity

  • Single-cell Western blotting: Detect PLCG1 protein in individual cells within complex populations

  • Mass cytometry (CyTOF): Simultaneously measure multiple parameters including PLCG1 expression and phosphorylation in thousands of individual cells

In vivo imaging methods:

  • Intravital microscopy with fluorescent reporters: Monitor PLCG1 activity in live tissues

  • FRET/BRET sensors: Develop biosensors to track PLCG1 activation dynamics in real-time

  • PET tracers for PLCG1: Design tracers that bind to PLCG1 or its substrates for non-invasive imaging

Computational tools for integration:

  • Spatial statistics: Develop methods to quantify PLCG1 expression patterns and correlations with microenvironmental features

  • Deep learning image analysis: Train neural networks to recognize complex patterns in PLCG1 distribution and activation

  • Agent-based modeling: Simulate PLCG1 signaling in heterogeneous cellular environments to generate testable hypotheses

Validation strategies:

  • Spatial validation of knockdown effects: Use in situ approaches to verify PLCG1 knockdown in specific regions within complex tissues

  • Cell type-specific genetic manipulation: Employ conditional knockout models to study PLCG1 function in specific cell types within intact tissues

  • Correlative light and electron microscopy (CLEM): Connect PLCG1 localization with ultrastructural features

These methodological advances would transform our understanding of PLCG1 biology by revealing context-dependent functions and regulatory mechanisms within complex tissue microenvironments, potentially leading to more effective therapeutic strategies.

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.