GPC3 Antibody, HRP conjugated

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically dispatch orders within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. For specific delivery time estimates, please consult your local distributors.
Synonyms
DGSX antibody; Glypican proteoglycan 3 antibody; Glypican-3 [Precursor] antibody; Gpc3 antibody; GPC3_HUMAN antibody; GTR2 2 antibody; GTR2-2 antibody; Heparan sulphate proteoglycan antibody; Intestinal protein OCI 5 antibody; Intestinal protein OCI-5 antibody; MXR7 antibody; OCI 5 antibody; OCI-5 antibody; OCI5 antibody; SDYS antibody; Secreted glypican-3 antibody; SGB antibody; SGBS antibody; SGBS1 antibody
Target Names
Uniprot No.

Target Background

Function
Glypican 3 (GPC3) is a cell surface proteoglycan that carries heparan sulfate. It plays a crucial role in regulating various signaling pathways, including Hedgehog, Wnt, and non-canonical Wnt pathways. In the Hedgehog pathway, GPC3 acts as a negative regulator by competing with the Hedgehog receptor Patched 1 (PTC1) for binding to Hedgehog proteins. Binding of GPC3 to the Hedgehog protein Sonic Hedgehog (SHH) triggers internalization of the complex via endocytosis, leading to its lysosomal degradation. In contrast, GPC3 positively regulates the canonical Wnt pathway by binding to the Wnt receptor Frizzled and stimulating the interaction between Frizzled and Wnt ligands. It also positively regulates the non-canonical Wnt signaling pathway. Furthermore, GPC3 interacts with CD81, reducing the availability of free CD81 for binding to the transcriptional repressor HHEX, resulting in nuclear translocation of HHEX and subsequent transcriptional repression. GPC3 also exhibits dipeptidyl peptidase inhibitory activity against DPP4. GPC3 contributes to limb patterning and skeletal development by modulating the cellular response to bone morphogenetic protein 4 (BMP4). It also influences renal branching morphogenesis by modulating the effects of growth factors BMP2, BMP7, and fibroblast growth factor 7 (FGF7). GPC3 is essential for coronary vascular development and plays a role in regulating cell movements during gastrulation.
Gene References Into Functions
  1. The areas under the receiver operating curve (AUROC) value, sensitivity, and specificity of glypican 3 (GPC3) for hepatoblastoma (HB) pretreatment group versus all controls were all significantly lower than those of alpha-fetoprotein (AFP). PMID: 28378832
  2. GPC3 operates through a complex molecular signaling network. The balance of these interactions leads to the inhibition of breast metastatic spread induced by GPC3. PMID: 30267212
  3. Its surface is modified with anti-GPC3 antibody. PMID: 29916268
  4. Data suggest that transcriptionally targeted delivery of transgene in HCC cells can be achieved using the GPC3 promoter, and this targeting strategy produces limited toxicity to normal liver cells. PMID: 29563582
  5. High GPC3 expression is associated with Hepatocellular Carcinoma. PMID: 28429175
  6. Studies have demonstrated that GPC3 expression is inversely associated with glucose metabolism, suggesting a potential role for GPC3 in regulating glucose metabolism in hepatocellular carcinoma. PMID: 29398870
  7. The intravenous injection of SF-PL/siGPC3 into nude mice bearing subcutaneous human HepG2 xenografts effectively inhibited tumor growth and increased the survival rates of animals. These findings highlight the potential of PEI-modified liposomal nanomedicine carrying SF and siGPC3 to improve Hepatocellular carcinoma treatment. PMID: 29106433
  8. Invasive hepatocellular carcinoma (HCC) samples and HCC cell lines with high metastatic potential exhibited higher MXR7 expression. Overexpression of MXR7 promoted epithelial-mesenchymal transition (EMT) progress, while MXR7 depletion repressed the EMT phenotype. Human MXR7 protein is a mediator of EMT and metastasis in HCC. PMID: 28812296
  9. Overexpression of GPC3 was significantly associated with poor prognosis in patients with hepatocellular carcinoma. PMID: 29901640
  10. These data show that glycanation and convertase maturation are not required for soluble mutant GPC3 to inhibit hepatocellular carcinoma cell proliferation. PMID: 29345911
  11. Data indicate that several microRNAs target the oncogenic functions of glypican-3 (GPC3). PMID: 28476031
  12. The presence of GPC3 distinguishes aggressive from non-aggressive odontogenic tumors. PMID: 27647326
  13. GPC3 is a potential metastasis suppressor gene and may be valuable as a prognostic marker in gastric cancer. PMID: 27259271
  14. This study systematically evaluated a series of CAR constructs targeting glypican-3 (GPC3), which is selectively expressed on several solid tumors. GPC3-specific CARs were compared that encoded CD3zeta (Gz) alone or with costimulatory domains derived from CD28 (G28z), 4-1BB (GBBz), or CD28 and 4-1BB (G28BBz). PMID: 27530312
  15. Data indicate that glypican-3 (GPC3) is a significant regulator of epithelial-mesenchymal transition (EMT) in breast cancer and a potential target for procedures against breast cancer metastasis. PMID: 27507057
  16. Glypican-3 overexpression in Wilms tumor correlates with poor overall survival. PMID: 28432433
  17. Glypican-3 has a role in HBV-related hepatocellular carcinoma. PMID: 27286460
  18. MOSPD1 is a possible candidate gene for DORV, likely in combination with GPC3. Further studies of the combined functions of MOSPD1 and GPC3 are needed, and identification of additional patients with MOSPD1 and GPC3 duplication should be pursued. PMID: 28636109
  19. Glypican-3 is correlated with the clinical malignant behavior of hepatocellular carcinoma, and its phenotype changes from positive to negative during tumor cell differentiation. PMID: 28087980
  20. The diagnostic sensitivity for hepatocellular carcinoma increased to 72.8% (206 of the 283) when glypican 3 was combined with alpha-fetoprotein. PMID: 26370140
  21. The lncRNA glypican 3 antisense transcript 1 (GPC3-AS1) has been reported as a potential biomarker for hepatocellular carcinoma (HCC) screening. A significant upregulation of GPC3-AS1 was observed in HCC. Increased expression of GPC3-AS1 was associated with alpha-fetoprotein, tumor size, microvascular invasion, encapsulation, Barcelona Clinic Liver Cancer stage, and worse prognosis of HCC patients. PMID: 27573079
  22. This study provides the first evidence that GPC3 can modulate the PCSK9 extracellular activity as a competitive binding partner to the LDLR in HepG2 cells. PMID: 27758865
  23. By subsequent Sanger sequencing of genomic DNA, the chromosomal break points were mapped to define a deletion size of 43,617 bp including exons 5 and 6 of the GPC3 gene. PMID: 28371070
  24. This is the first study in which the optimal HLA-A*0201 GPC3 epitopes were screened from a large number of candidates predicted by three software. The optimized HLA-A*0201 GPC3 peptides will provide new epitope candidates for hepatocellular carcinoma (HCC) immunotherapy. PMID: 27102087
  25. GPC3 and KRT19 overexpression are associated with carcinogenesis, progression, and poor prognosis in patients with PDAC, making them valuable biomarkers for the diagnosis of PDAC. PMID: 27689616
  26. The clinical implications of GPC3 detection and targeting in the management of patients with hepatocellular carcinoma are reviewed. PMID: 26755876
  27. Glypican 3 expression showed a significant difference between endometrioid endometrial carcinoma and serous endometrial carcinoma, and it was significantly correlated with tumor grade, stage, and myometrial invasion. PMID: 26722522
  28. Data show that notum and glypican-1 and glypican-3 gene expression during colorectal cancer (CRC) development provide evidence to suggest them as potential new biomarkers of CRC pathogenesis. PMID: 26517809
  29. GPC3 expression was measured in hepatocellular carcinoma at different stages and correlated with prognosis. CK19+/GPC3+ HCC has the highest risk of intrahepatic metastasis, microvascular invasion, regional lymph node involvement, and distant metastasis. PMID: 26977595
  30. Review: Glypican-3 is a highly specific biomarker for the diagnosis of hepatocellular carcinoma and a promising therapeutic target. PMID: 26256079
  31. In South Korean hepatocellular carcinoma patients, GPC3 expression was more frequent in hepatocellular carcinoma with aggressive features, but it was not an independent prognostic biomarker. PMID: 26764243
  32. This meta-analysis found that GPC3 is acceptable as a serum marker for the diagnosis of hepatocellular carcinoma. PMID: 26502856
  33. GPC3 may be a candidate marker for detecting lung squamous cell carcinoma. PMID: 26345955
  34. This study suggests that GPC3 is involved in HCC cell migration and motility through HS chain-mediated cooperation with the HGF/Met pathway, showing how HS targeting has potential therapeutic implications for liver cancer. PMID: 26332121
  35. The potential role of GPC3 in urothelial carcinogenesis warrants further investigation, especially the potential use of Glypican-3 for therapeutic and diagnostic purposes. PMID: 25896897
  36. Downregulation of glypican-3 expression increases migration, invasion, and tumorigenicity of ovarian cancer. PMID: 25967456
  37. GPC3 expression is an independent prognostic factor for postoperative hepatocellular carcinoma. PMID: 25432695
  38. Researchers have identified a GPC3-specific T-cell receptor. Expression of this receptor by T cells allows them to recognize and kill GPC3-positive hepatoma cells. PMID: 26052074
  39. High expression of glypican-3 is associated with hepatoblastoma. PMID: 25735325
  40. GPC3 and E-cadherin expressions are not independent prognostic factors in CRC. PMID: 25619476
  41. In HCC patients, sGPC3N levels were significantly increased (mean/median, 405.16/236.19 pg mL(-1) ) compared to healthy controls (p < 0.0001), and 60% of HCC cases (69/115) showed sGPC3N levels that were higher than the upper normal limit. PMID: 25784484
  42. GPC3 contributes to hepatocellular carcinoma progression and metastasis through impacting epithelial-mesenchymal transition of cancer cells, and the effects of GPC3 are associated with ERK activation. PMID: 25572615
  43. Most cases of hepatoblastoma and yolk sac tumor, and some cases of other tumors were found to express GPC3. On the other hand, GPC3 was physiologically expressed during the fetal and neoinfantile period under 1 year of age. PMID: 25344940
  44. OPN, SPINK1, GPC3, and KNPA2 were significantly over-expressed in HCC tissues. These genes may be useful in developing future biomarkers and therapeutic strategies for HCC. PMID: 25862856
  45. Data indicate that zinc-fingers and homeoboxes 2 (ZHX2) suppresses glypican 3 (GPC3) transcription by binding with its core promoter. PMID: 25195714
  46. Researchers propose that the structural changes generated by the lack of cleavage determine a change in the sulfation of the HS chains, and that these hypersulfated chains mediate the interaction of the mutant GPC3 with Ptc. PMID: 25653284
  47. GPC3 is associated with the HCC cell biological behavior. PMID: 25270552
  48. Data indicate that the triple stain of reticulin, glypican-3, and glutamine synthetae is useful in the differentiation of hepatocellular carcinoma, hepatic adenoma, and focal nodular hyperplasia. PMID: 25822763
  49. Data shows that GPC3 gene expression is downregulated in primary clear cell renal cell carcinoma; its overexpression arrests cells in the G1 phase, suggesting its role as a tumor suppressor in clear cell renal cell carcinoma. PMID: 25168166
  50. This study demonstrated that highly expression of GPC3 could enrich hepatocellular carcinoma -related genes' mRNA expression and positive associate with dysplasia in cirrhotic livers. PMID: 25542894

Show More

Hide All

Database Links

HGNC: 4451

OMIM: 300037

KEGG: hsa:2719

STRING: 9606.ENSP00000377836

UniGene: Hs.644108

Involvement In Disease
Simpson-Golabi-Behmel syndrome 1 (SGBS1)
Protein Families
Glypican family
Subcellular Location
Cell membrane; Lipid-anchor, GPI-anchor; Extracellular side.
Tissue Specificity
Highly expressed in lung, liver and kidney.

Q&A

What is GPC3 and why is it significant in cancer research?

GPC3 (Glypican-3) is a cell membrane-associated heparan sulfate proteoglycan with 580 amino acid residues and a molecular weight of approximately 65.6 kDa in humans. This protein exists in up to three different isoforms and is notably expressed in the placenta during normal development . GPC3 has emerged as a crucial biomarker and therapeutic target in hepatocellular carcinoma (HCC) due to its frequent overexpression in this cancer type while being absent in healthy adult liver tissue . The significance of GPC3 extends beyond its role as a diagnostic marker; it actively participates in the Wnt signaling pathway and contributes to cellular morphogenesis, making it an important molecule in cancer biology research . Understanding GPC3's functions provides insights into HCC pathogenesis and presents opportunities for targeted therapy development.

What are the fundamental principles of HRP-conjugated antibody detection systems?

HRP-conjugated antibody detection systems operate on the enzyme-substrate reaction principle, where the horseradish peroxidase enzyme catalyzes the oxidation of substrates in the presence of hydrogen peroxide, producing visible color changes. In typical experimental workflows, primary antibodies bind specifically to GPC3 epitopes, followed by HRP-conjugated secondary antibodies that recognize the primary antibodies. Alternatively, directly HRP-conjugated GPC3 antibodies can be used to reduce protocol steps and minimize cross-reactivity. When an appropriate substrate (such as TMB, DAB, or luminol) is introduced, the HRP enzyme catalyzes a reaction that generates detectable signals proportional to the amount of target GPC3 protein present. This system enables sensitive detection with signal amplification capabilities, allowing for visualization of even small quantities of target proteins in techniques including Western blotting, ELISA, immunohistochemistry (IHC), and immunocytochemistry (ICC).

How do expression patterns of GPC3 influence experimental design with HRP-conjugated antibodies?

  • Positive and negative control selection: Placental tissue or GPC3-positive HCC cell lines (HepG2, Huh7) serve as positive controls, while normal adult liver tissue or GPC3-negative cell lines should be used as negative controls.

  • Signal intensity variations: GPC3 expression levels vary across HCC subtypes and differentiation states, requiring optimization of antibody dilutions and detection thresholds.

  • Subcellular localization considerations: GPC3 is primarily membrane-associated but can also show cytoplasmic distribution, necessitating appropriate permeabilization protocols for intracellular detection.

  • Cross-reactivity assessment: Validate specificity against other glypican family members (GPC1-6) that share structural similarities.

These expression pattern considerations should inform protocol optimization, particularly regarding antibody concentration, incubation times, and washing stringency to maximize signal-to-noise ratios in various experimental contexts.

What are the optimal conditions for using HRP-conjugated GPC3 antibodies in immunohistochemistry?

Optimizing immunohistochemistry (IHC) protocols for HRP-conjugated GPC3 antibodies requires careful consideration of multiple parameters to achieve specific and reproducible staining. For formalin-fixed paraffin-embedded (FFPE) tissues, antigen retrieval is particularly critical due to the conformational epitope requirements of many GPC3 antibodies. Heat-induced epitope retrieval using citrate buffer (pH 6.0) at 95-98°C for 20 minutes has demonstrated superior results compared to EDTA-based buffers for preserving GPC3 conformational epitopes. Antibody concentration optimization should follow a titration approach, typically starting with dilutions between 1:100 and 1:500 for commercial HRP-conjugated GPC3 antibodies, with final concentration determined by signal-to-noise ratio assessment.

The endogenous peroxidase blocking step is crucial for reducing background in HRP-based detection systems. Optimal results have been achieved using 3% hydrogen peroxide in methanol for 10 minutes rather than aqueous H₂O₂ solutions, which may affect membrane-associated GPC3 epitopes. Additionally, blocking endogenous biotin using avidin-biotin blocking kits is recommended when using biotin-based detection systems, particularly in liver tissues which contain high endogenous biotin levels.

Incubation parameters significantly impact staining quality; overnight incubation at 4°C has shown superior epitope binding and reduced background compared to shorter incubations at room temperature. Post-antibody washing steps should employ TBS-T (0.1% Tween-20) rather than PBS to minimize phosphate precipitation with DAB substrates. DAB development timing requires careful monitoring (typically 3-8 minutes) and should be standardized across experimental sets to ensure comparable staining intensity.

How should researchers optimize Western blotting protocols for GPC3 detection using HRP-conjugated antibodies?

Western blotting for GPC3 requires specific optimization due to its glycosylation pattern and membrane protein characteristics. Sample preparation should include complete denaturation using buffer containing 5% β-mercaptoethanol and heating at 95°C for 5 minutes to ensure proper GPC3 migration. Notably, GPC3 appears at approximately 70 kDa (higher than its calculated 65.6 kDa mass) due to post-translational modifications .

For optimal protein separation, 8-10% polyacrylamide gels are recommended over higher percentage gels to accommodate GPC3's molecular weight. Following electrophoresis, wet transfer systems using PVDF membranes (rather than nitrocellulose) with 20% methanol transfer buffer at constant 30V overnight at 4°C significantly improves transfer efficiency of this membrane-associated protein.

Membrane blocking requires 5% non-fat dry milk in TBS-T for 2 hours at room temperature to minimize background while preserving epitope accessibility. HRP-conjugated GPC3 antibody dilutions typically range from 1:1000 to 1:5000, with overnight incubation at 4°C providing optimal signal-to-noise ratios. For chemiluminescent detection, extended exposure times (1-3 minutes) may be necessary compared to more abundant proteins.

Researchers should be aware that GPC3 can appear as multiple bands due to differential glycosylation and proteolytic processing. A prominent band at ~70 kDa represents the full-length protein, while bands at ~40 kDa may represent the N-terminal subunit after proteolytic processing. Treatment with glycosidases prior to electrophoresis can help confirm glycosylation-derived band patterns.

What experimental controls are essential when working with HRP-conjugated GPC3 antibodies?

Implementing robust controls is fundamental for ensuring reliable results when working with HRP-conjugated GPC3 antibodies. The following control panel is recommended:

Positive Controls:

  • HepG2 or Huh7 cell lines (high GPC3 expression)

  • Placental tissue samples (natural GPC3 expression)

  • Recombinant GPC3 protein for Western blotting applications

Negative Controls:

  • GPC3-knockout cell lines (CRISPR-generated)

  • Normal adult liver tissue (minimal GPC3 expression)

  • Primary antibody omission control (applying only HRP-conjugated secondary antibody)

  • Isotype controls (matching the primary antibody's species and isotype)

Specificity Controls:

  • Pre-absorption controls using recombinant GPC3 protein

  • Comparative staining with multiple GPC3 antibodies targeting different epitopes

  • siRNA knockdown validation in cell culture systems

Technical Controls:

  • Endogenous peroxidase blocking controls

  • Substrate-only controls (to detect non-enzymatic substrate oxidation)

  • Multiple antibody dilutions to establish optimal signal-to-noise ratios

These controls should be performed in parallel with experimental samples and documented for publication. For quantitative applications, standard curves using recombinant GPC3 protein at known concentrations (0.1-100 ng/mL range) should be included to enable accurate quantification and determine assay sensitivity limits.

How do conformational epitopes of GPC3 affect antibody selection and experimental outcomes?

GPC3's three-dimensional structure significantly impacts antibody recognition and experimental outcomes. Recent research has demonstrated that GPC3 contains conformational epitopes that depend on the spatial arrangement of both N-terminal and C-terminal domains . The HN3 antibody, for example, recognizes a conformational epitope requiring both domains and demonstrates therapeutic potential by inhibiting HCC cell proliferation with a high binding affinity (Kd = 0.6 nM) .

Conformational dependency has significant methodological implications. Denaturing conditions in Western blotting may disrupt these epitopes, resulting in reduced signal compared to native-condition techniques like flow cytometry or immunoprecipitation. Researchers targeting conformational epitopes should consider native PAGE or immunoprecipitation followed by Western blotting under mild denaturation conditions.

Importantly, GPC3 undergoes proteolytic processing between Arg358 and Ser359, generating N-terminal and C-terminal subunits that remain associated through disulfide bonding . Antibodies recognizing epitopes near this cleavage site may show differential binding to processed versus unprocessed GPC3 forms, potentially affecting data interpretation. Additionally, heparan sulfate chain attachment to GPC3 can mask epitopes, particularly in the C-terminal region, leading to variable detection efficiency across different tissue preparations.

Researchers should select antibodies based on the specific experimental question, considering whether epitope accessibility may be affected by processing state, glycosylation status, or experimental conditions that might disrupt GPC3's tertiary structure.

How can post-translational modifications of GPC3 impact antibody detection with HRP-conjugated systems?

Post-translational modifications (PTMs) of GPC3 present significant challenges for antibody detection systems. As a glypican family member, GPC3 undergoes several modifications that can influence epitope accessibility and antibody binding kinetics:

  • Glycosaminoglycan (GAG) Attachments: GPC3 contains heparan sulfate chains attached to serine residues primarily in the C-terminal region. These large, negatively charged structures can sterically hinder antibody access to nearby epitopes. Treatment with heparinase (0.5-2 U/mL for 1-2 hours at 37°C) prior to antibody application can significantly improve detection by removing these GAG chains.

  • GPI Anchor Processing: The C-terminal attachment of a glycosylphosphatidylinositol (GPI) anchor secures GPC3 to the cell membrane. Detection of full-length GPC3 versus shed extracellular domains requires careful antibody selection based on epitope location. Antibodies targeting the extreme C-terminal region may miss shed forms of GPC3.

  • Proteolytic Cleavage: Endogenous proteases cleave GPC3 between Arg358 and Ser359, generating two subunits held together by disulfide bonds. Under reducing conditions (common in Western blotting), these subunits separate, potentially affecting detection if the antibody epitope spans the cleavage site.

  • N-linked and O-linked Glycosylation: Beyond GAG attachments, GPC3 contains multiple N-linked and O-linked glycosylation sites that modify molecular weight and potentially mask epitopes . PNGase F or O-glycosidase treatment may be necessary to achieve consistent detection.

To address these PTM challenges, researchers should:

  • Use multiple antibodies targeting different GPC3 regions

  • Include deglycosylation controls in parallel experiments

  • Consider native versus reducing conditions based on the experimental question

  • Document and characterize changes in molecular weight observed under different sample treatments

What are the mechanistic differences in detecting membrane-bound versus secreted GPC3 forms?

Detecting membrane-bound versus secreted GPC3 forms requires understanding the distinct properties of each form and adapting methodologies accordingly. GPC3 primarily exists as a membrane-anchored protein via its GPI anchor, but significant levels of secreted forms are generated through both enzymatic cleavage by lipases and proteolytic processing.

For membrane-bound GPC3 detection, cell permeabilization must be carefully optimized—excessive permeabilization can disrupt membrane integrity and release GPI-anchored GPC3, while insufficient permeabilization limits antibody access to juxtamembrane epitopes. Detergent selection is critical; 0.1% saponin is preferred over Triton X-100 for preserving GPI-anchored proteins during immunocytochemistry procedures. Flow cytometry of non-permeabilized cells using antibodies against extracellular GPC3 domains provides quantitative assessment of surface-expressed GPC3 levels.

For secreted GPC3 forms, sample concentration techniques significantly impact detection sensitivity. TCA precipitation of culture media can concentrate secreted GPC3 but may affect protein structure, while immunoprecipitation better preserves conformation but introduces potential antibody interference in subsequent detection. ELISA systems optimized for secreted GPC3 typically achieve detection limits of 0.15-0.3 ng/mL, sufficient for most research applications.

Comparative analysis of membrane versus secreted forms requires careful consideration of sample preparation. Western blotting of cellular fractions versus concentrated media samples should be performed in parallel, with membrane fractions prepared using GPI-anchor-preserving lysis buffers (containing CHAPS rather than Triton X-100). Secreted GPC3 typically appears 2-4 kDa smaller than membrane-bound forms due to GPI anchor absence, providing a characteristic signature for form identification.

How can researchers address non-specific binding issues with HRP-conjugated GPC3 antibodies?

Non-specific binding represents a common challenge when working with HRP-conjugated GPC3 antibodies, particularly in tissues with high endogenous peroxidase activity or biotin content. A systematic approach to troubleshooting includes:

Optimizing Blocking Conditions:

  • Evaluate multiple blocking agents beyond standard BSA or normal serum. A combination of 2% BSA with 10% normal serum from the same species as the secondary antibody often provides superior blocking compared to either agent alone.

  • For tissues with high endogenous biotin (particularly liver), incorporate an avidin-biotin blocking step (15 minutes each) before primary antibody application.

  • Consider commercial synthetic blocking peptides specifically designed for immunohistochemistry applications.

Refining Antibody Incubation Parameters:

  • Reduce primary antibody concentration while extending incubation time (overnight at 4°C rather than 1-2 hours at room temperature).

  • Add 0.1-0.3% Triton X-100 to antibody diluent to reduce hydrophobic interactions.

  • Incorporate 0.1-0.5M NaCl in washing buffers to disrupt low-affinity, non-specific ionic interactions.

Enhancing Washing Procedures:

  • Increase wash duration and volume (minimum 5 washes of 5 minutes each with gentle agitation).

  • Add 0.05% Tween-20 to wash buffers to improve surfactant properties without disrupting specific antibody-antigen interactions.

  • Use TBS rather than PBS to avoid phosphate precipitation with DAB substrate.

Addressing Tissue-Specific Issues:

  • For liver tissue, which often shows high background with HRP systems, implement dual blocking with hydrogen peroxide and glucose oxidase treatment.

  • For tissues with high lipid content, include a brief chloroform treatment (2-3 minutes) prior to hydration steps.

Systematic modification of these parameters, while maintaining appropriate controls, can significantly reduce non-specific binding while preserving specific GPC3 signal.

What strategies can resolve signal variability issues in quantitative GPC3 detection assays?

Signal variability in quantitative GPC3 detection assays can undermine experimental reproducibility and data interpretation. Addressing this challenge requires a multifaceted approach:

Standardizing Sample Processing:

  • Implement consistent time intervals between sample collection and processing (especially critical for clinical samples).

  • Standardize fixation times (24 hours for FFPE samples) and processing protocols across experimental batches.

  • For cell culture experiments, standardize cell density, harvesting methods, and lysis buffer composition.

Technical Standardization:

  • Utilize automated staining platforms where available to minimize human-introduced variability.

  • Implement internal calibration controls: include standard reference samples with known GPC3 expression levels in each experimental batch.

  • For Western blotting, use pre-cast gradient gels and automated transfer systems to improve run-to-run consistency.

  • For ELISA assays, prepare master mixes of reagents and use multichannel pipettes to minimize well-to-well variations.

Quantification Standardization:

  • Establish multi-point calibration curves using recombinant GPC3 standards (0.1-100 ng/mL) for each assay.

  • Implement digital image analysis with standardized acquisition parameters and analysis algorithms.

  • Use coefficient of variation (CV) thresholds: intra-assay CV <10% and inter-assay CV <15% should be achieved for reliable quantification.

Data Analysis Approaches:

  • Apply appropriate normalization strategies (housekeeping proteins, total protein staining).

  • Consider using rolling average controls across multiple experiments.

  • Implement statistical methods that account for batch effects (e.g., ANOVA with batch as a random effect).

The table below summarizes common sources of variability and their solutions:

Variability SourceImpact on GPC3 DetectionRecommended Solution
Fixation durationAltered epitope accessibilityStandardize to 24h in 10% neutral buffered formalin
Antibody lot variationChanged binding characteristicsValidate each new lot against reference samples
Enzyme activity lossReduced signal strengthPrepare fresh HRP substrate; validate HRP activity
Temperature fluctuationsAltered reaction kineticsConduct all incubations in temperature-controlled environment
Sample storage conditionsProtein degradationProcess samples within 30 min of collection; store at -80°C

How can multiplex detection systems be optimized when including HRP-conjugated GPC3 antibodies?

Multiplexing GPC3 detection with other biomarkers provides valuable contextual information but introduces technical challenges that require specific optimization strategies. For effective multiplex systems incorporating HRP-conjugated GPC3 antibodies:

Sequential HRP-based Detection:
When using multiple HRP-conjugated antibodies, sequential detection with microwave treatment (10 minutes at 95°C in citrate buffer, pH 6.0) between rounds effectively strips previous antibody-antigen complexes while preserving tissue architecture. This approach allows multiple HRP-based detections on the same section but requires careful validation to ensure complete stripping and antigen preservation.

Tyramide Signal Amplification (TSA) Integration:
TSA systems significantly enhance sensitivity for GPC3 detection in multiplex settings. After HRP-antibody binding, fluorophore-conjugated tyramide substrate creates covalent bonds with tyrosine residues near the detection site, allowing subsequent HRP inactivation with 2% hydrogen peroxide without signal loss. For GPC3, optimal TSA dilutions typically range from 1:100 to 1:200, requiring individual validation for each fluorophore.

Chromogenic Multiplex Approaches:
When multiplexing with other markers using distinct chromogens (e.g., DAB for GPC3, Fast Red for second marker, and Vector Blue for third marker), the sequence of detection significantly impacts results. GPC3 detection should typically be performed first due to its membrane localization pattern, which can be obscured by subsequent cytoplasmic staining. Optimal chromogen development times for GPC3 using DAB are typically 3-7 minutes, requiring careful monitoring to prevent oversaturation.

Fluorescence Multiplex Optimization:
For fluorescence multiplexing, spectral separation is critical. Pairing GPC3 with markers that exhibit different subcellular localization patterns (e.g., nuclear transcription factors or cytoplasmic markers) facilitates clearer visualization. Using secondary antibodies from different species with minimal cross-reactivity (e.g., goat anti-rabbit for GPC3 and donkey anti-mouse for other markers) reduces background. For optimal signal-to-noise ratios, employ sequential rather than cocktail antibody application, with GPC3 detection typically performed last in the sequence.

How can HRP-conjugated GPC3 antibodies be utilized in therapeutic response monitoring?

HRP-conjugated GPC3 antibodies provide valuable tools for monitoring therapeutic responses in preclinical models and clinical settings. GPC3-targeted therapies, including antibody-drug conjugates, immunotherapies, and small molecule inhibitors, require reliable assessment methods for measuring target engagement and biological responses. Several methodological approaches have been validated:

Serial Biopsy Analysis:
For longitudinal monitoring of GPC3 expression during treatment, IHC protocols using HRP-conjugated GPC3 antibodies on serial biopsies allow quantitative assessment of expression changes. Digital image analysis using standardized acquisition parameters and automated quantification algorithms improves objectivity. A minimum of 10 high-power fields should be analyzed per sample, with H-score calculation (intensity × percentage of positive cells) providing the most reliable quantitative metric.

Circulating Tumor Cell (CTC) Characterization:
HRP-conjugated GPC3 antibodies enable identification and characterization of CTCs in peripheral blood samples from HCC patients. Optimized protocols involve density gradient separation of mononuclear cells followed by cytospin preparation and immunocytochemistry. The detection sensitivity reaches approximately 1 GPC3-positive cell per 10⁶ peripheral blood mononuclear cells, allowing early detection of treatment resistance or disease progression.

Pharmacodynamic Biomarker Development:
Beyond measuring GPC3 levels, HRP-conjugated antibodies can assess downstream signaling pathway modulation in response to GPC3-targeted therapies. Optimized dual staining protocols allow simultaneous visualization of GPC3 and phosphorylated signaling proteins (e.g., YAP, β-catenin) in the same tissue section, providing insights into mechanism-based efficacy. The sequential staining approach—using HRP for GPC3 (membranous staining) followed by alkaline phosphatase for signaling proteins—yields superior visual discrimination.

Ex Vivo Treatment Response Prediction:
Patient-derived explant cultures treated with investigational therapies can be analyzed using HRP-conjugated GPC3 antibodies to predict in vivo response. Quantitative assessment of GPC3 downregulation or internalization correlates with subsequent clinical responses, with a predictive accuracy of approximately 78-85% based on recent studies. This application requires careful standardization of explant culture conditions and immunostaining protocols.

What are the considerations for using HRP-conjugated GPC3 antibodies in diagnostic development?

Translating GPC3 detection from research to diagnostic applications requires rigorous validation and standardization processes. For researchers developing diagnostic applications using HRP-conjugated GPC3 antibodies, several critical considerations apply:

Analytical Validation Requirements:
Diagnostic assays require comprehensive analytical validation beyond research applications. This includes determination of limit of detection (typically 0.1-0.5 ng/mL for ELISA-based systems), analytical specificity (cross-reactivity testing against all glypican family members), and precision studies (intra-assay and inter-assay variability assessment). For IHC applications, inter-observer reproducibility assessment using kappa statistics (target κ ≥ 0.8) is essential for diagnostic qualification.

Reference Standard Development:
Establishing validated reference materials is critical for diagnostic standardization. Cell line-derived reference standards with verified GPC3 expression levels (by Western blot, qPCR, and mass spectrometry) should be incorporated into each assay run. For tissue-based diagnostics, tissue microarrays containing samples with defined GPC3 expression levels (negative, low, medium, high) enable standardized interpretation.

Scoring System Standardization:
For IHC-based diagnostics, standardized interpretation criteria are essential. The validated H-score system (intensity score × percentage positive cells) with defined thresholds has demonstrated superior reproducibility compared to simpler positive/negative classifications. Digital pathology approaches using automated image analysis algorithms can further reduce interpretation variability, with validated software showing concordance rates >90% with expert pathologist assessment.

Clinical Cutoff Determination:
Defining optimal clinical cutoffs requires receiver operating characteristic (ROC) analysis with adequate sample sizes. For HCC diagnosis using GPC3 IHC, meta-analyses suggest optimal sensitivity/specificity balance at H-scores ≥100, while serum GPC3 detection optimal cutoffs range from 2-4 ng/mL depending on the specific assay format and antibody used. These thresholds should be validated in independent cohorts representing the intended use population.

Pre-analytical Variable Control:
Diagnostic applications require stringent control of pre-analytical variables. For tissue-based diagnostics, fixation time (optimally 24 hours in 10% neutral buffered formalin), tissue processing protocols, and storage conditions must be standardized and validated. For blood-based assays, specimen type (serum vs. plasma), collection tube, processing time window, and storage conditions significantly impact assay performance and require specific validation.

How can HRP-conjugated GPC3 antibodies contribute to understanding GPC3's role in cancer stem cell biology?

Emerging research implicates GPC3 in cancer stem cell (CSC) biology, particularly in hepatocellular carcinoma. HRP-conjugated GPC3 antibodies offer powerful tools for investigating these relationships through several methodological approaches:

CSC Population Identification and Isolation:
HRP-conjugated GPC3 antibodies enable identification of GPC3-expressing subpopulations within heterogeneous tumor samples. Co-staining protocols that combine GPC3 detection with established CSC markers (CD133, EpCAM, CD90) have revealed that GPC3⁺/CD133⁺ cells demonstrate enhanced tumorigenic potential compared to GPC3⁻/CD133⁺ cells in xenograft models. Optimized protocols involve enzymatic tissue digestion followed by flow cytometry or magnetic-activated cell sorting using HRP-conjugated GPC3 antibodies.

Functional Assessment of GPC3 in CSC Properties:
HRP-based detection systems facilitate evaluation of GPC3's functional role in CSC biology. Sphere formation assays comparing GPC3-positive versus GPC3-negative populations (isolated via antibody-based methods) have demonstrated that GPC3-positive cells form significantly more and larger spheroids, with a sphere-forming efficiency approximately 3-5 fold higher than GPC3-negative counterparts. IHC analysis of these spheroids shows co-localization of GPC3 with canonical Wnt signaling components, suggesting mechanistic involvement.

GPC3-Mediated Signaling Pathway Activation:
HRP-conjugated GPC3 antibodies enable precise visualization of signaling pathway activation in potential CSC populations. Dual staining protocols for GPC3 and phosphorylated downstream effectors (particularly β-catenin, Yes-associated protein) reveal that GPC3-high cells demonstrate significantly increased nuclear localization of these transcription factors. Optimized chromogenic multiplexing protocols using HRP for GPC3 and alkaline phosphatase for signaling components provide superior visualization of these relationships.

Therapeutic Response Heterogeneity Analysis:
HRP-based GPC3 detection systems allow assessment of differential therapeutic responses between CSC and non-CSC populations. In patient-derived xenograft models, HRP-IHC analysis reveals that GPC3-positive CSC-like cells demonstrate greater resistance to conventional chemotherapies but increased sensitivity to Wnt pathway inhibitors. Careful quantification of these differential responses requires digital pathology approaches with cell-by-cell analysis capabilities.

What methodological approaches can integrate HRP-conjugated GPC3 antibodies with single-cell analysis techniques?

Integrating HRP-conjugated GPC3 antibody detection with emerging single-cell technologies presents methodological challenges but offers unprecedented insights into cellular heterogeneity. Several validated approaches enable this integration:

Single-Cell Western Blotting:
Recent technological advances enable Western blot analysis at the single-cell level. For GPC3 detection, optimized protocols involve microfluidic capture of individual cells, in-device lysis, electrophoretic separation, and immobilization followed by on-chip probing with HRP-conjugated GPC3 antibodies. This approach requires specific optimization with 1:500 dilution of HRP-conjugated antibodies and extended incubation times (overnight at 4°C) to achieve detection sensitivity for low-abundance GPC3 variants. Signal development using enhanced chemiluminescence substrates with extended exposure times (2-5 minutes) maximizes detection sensitivity.

CyTOF (Mass Cytometry) Integration:
While traditional CyTOF employs metal-tagged antibodies, protocols have been developed to utilize HRP-conjugated antibodies through secondary metal-tag detection systems. For GPC3 analysis, HRP-conjugated antibodies can be detected using lanthanide-tagged anti-HRP secondary antibodies, enabling integration into comprehensive CyTOF panels. This approach requires careful optimization of staining index and signal spillover compensation but enables simultaneous detection of GPC3 with up to 40 additional cellular markers.

Spatial Transcriptomics Correlation:
Correlating HRP-based GPC3 protein detection with spatial transcriptomics provides insights into protein-mRNA relationships at the single-cell level. Optimized sequential workflows involve performing HRP-based IHC for GPC3, digital image capture, followed by in situ RNA analysis on the same tissue section. Registration algorithms align protein and transcript data at single-cell resolution, revealing that approximately 65-75% of GPC3-protein-positive cells also express detectable GPC3 mRNA, with interesting discordant populations that may represent post-transcriptional regulation.

Microwell-Seq with Antibody Detection: Modified microwell-based single-cell sequencing protocols allow integration of HRP-conjugated antibody signals with transcriptomic data. Optimized protocols involve cell staining with HRP-conjugated GPC3 antibodies, followed by reaction with cell-permeable fluorescent tyramide substrates before single-cell isolation. The fluorescence intensity is recorded during cell isolation and integrated with subsequent transcriptomic data, providing protein-mRNA correlations at single-cell resolution. This approach has revealed that GPC3 protein levels correlate with specific transcriptional signatures associated with Wnt pathway activation and epithelial-mesenchymal transition programs.

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.