PEG10 Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to PEG10 Monoclonal Antibodies

PEG10 (Paternally Expressed Gene 10) is a retrotransposon-derived protein implicated in placental development, cancer progression, and apoptosis regulation. PEG10 monoclonal antibodies are critical tools for detecting and studying this protein in research and diagnostic settings. These antibodies are engineered to bind specifically to PEG10, enabling applications such as immunoprecipitation, western blotting, flow cytometry, and enzyme-linked immunosorbent assays (ELISAs). Below is a detailed analysis of their properties, applications, and research findings.

General Properties

ParameterDetails
Target ProteinPEG10 (UniProt ID: Q86TG7; Molecular Weight: ~80–100 kDa)
Host SpeciesMouse or Rabbit (depending on antibody)
ApplicationsELISA, Western Blot (WB), Immunoprecipitation (IP), Immunohistochemistry (IHC), Flow Cytometry (FC)
ImmunogensRecombinant human PEG10 protein fragments or synthetic peptides

ELISA Pairing for Quantitative Detection

  • MAB99751 (Capture) and MAB9975 (Detection) form an antibody pair for sandwich ELISA assays. This combination enables precise quantification of PEG10 in human samples .

  • Optimal Dilutions: Capture antibody (e.g., MAB99751) is typically used at 1:100–1:200, while detection antibodies (e.g., MAB9975) are diluted 1:1000–1:2000 .

Western Blotting and Protein Validation

  • ab215035 (Abcam) detects PEG10 at ~100 kDa in HepG2, HeLa, and HEK293 lysates, though the predicted molecular weight is 80 kDa .

  • M03240 (Boster Bio) validates PEG10 in HeLa lysates via WB, confirming specificity against recombinant PEG10 .

Flow Cytometry and Intracellular Staining

  • MAB99751 enables intracellular staining of PEG10 in fixed/permeabilized HepG2 cells, distinguishing it from isotype controls .

  • ab215035 is used for intracellular FC, labeling PEG10 in HepG2 cells with PE-conjugated secondary antibodies .

Role in Cancer Progression

PEG10 overexpression is linked to enhanced proliferation, metastasis, and poor prognosis in lung, breast, and liver cancers . Key findings include:

  • Lung Cancer (NSCLC):

    • PEG10 upregulates β-catenin, MMP-2, and MMP-9 while suppressing E-cadherin, promoting epithelial-mesenchymal transition (EMT) and cell invasion .

    • Wound Healing Assay: PEG10 knockdown in A549 cells reduces migration rate by ~40% over 48 hours .

    • Transwell Invasion Assay: PEG10 siRNA decreases invasive cell count by ~50% .

Cell LineAssayEffect of PEG10 KnockdownP-Value
A549Wound HealingReduced migration rate<0.05
A549Transwell InvasionDecreased invasive cell count<0.05

Apoptosis and Signaling Pathways

  • TGF-β Signaling: PEG10 interacts with TGF-β receptor ACVRL1, inhibiting TGF-β-mediated apoptosis .

  • Wnt/β-Catenin Pathway: PEG10 downregulation reduces β-catenin levels and increases E-cadherin expression, reversing EMT .

Clinical and Prognostic Implications

  • Biomarker Potential: High PEG10 expression correlates with advanced TNM stages and poor survival in lung cancer patients .

  • Therapeutic Target: Targeting PEG10 may suppress metastasis by inhibiting MMPs and restoring E-cadherin .

Product Specs

Form
Purified mouse monoclonal antibody in a buffer containing 0.1M Tris-Glycine (pH 7.4, 150 mM NaCl), supplemented with 0.2% sodium azide and 50% glycerol.
Lead Time
We typically dispatch orders for PEG10 Monoclonal Antibody within 1-3 business days of receipt. Delivery times may vary depending on the chosen shipping method and destination. For specific delivery timelines, please contact your local distributor.
Synonyms
EDR, HB-1, Mar2, MEF3L, Mart2, RGAG3

Q&A

What is PEG10 and why is it important in research?

PEG10 is a human retrotransposon-derived imprinted gene located at chromosome 7q21. The mRNA of PEG10 encodes two protein isoforms: the Gag-like protein (RF1 PEG10) coded by reading frame 1, and the Gag-Pol-like polyprotein (RF1/RF2 PEG10) coded by reading frames 1 and 2. These proteins are translated through a typical retroviral frameshift mechanism. PEG10 is critically important in research because it plays essential roles in placenta formation and adipocyte differentiation, with knockout of this gene causing embryonic lethality. Furthermore, its upregulated expression has been linked to oncogenesis and cancer progression in multiple malignancies, including hepatocellular carcinoma, bladder cancer, lung cancer, breast cancer, pancreatic cancer, and cutaneous T-cell lymphoma .

What are the structural characteristics of PEG10 proteins?

The PEG10 gene encodes two overlapping open reading frames (ORFs). The first reading frame produces the RF1 PEG10 protein with Gag-like characteristics, associated with invasion and drug resistance. The second protein, RF1/RF2 PEG10, is synthesized by a programmed -1 frameshift translation mechanism typical of retroviruses. The protease (PR) domain of RF2 PEG10 contains an -Asp-Ser-Gly- sequence, which corresponds to the consensus -Asp-Ser/Thr-Gly- active-site motif found in retroviral aspartic proteases. The substrate binding sites of PEG10 protease have been analyzed through homology modeling, revealing similarities with retroviral proteases like HIV-1, HIV-2, and EIAV (Equine Infectious Anemia Virus) proteases, particularly in the S1-S4 binding sites that determine substrate specificity .

How does a PEG10 monoclonal antibody differ from polyclonal antibodies in research applications?

When using PEG10 monoclonal antibodies in research, you achieve higher specificity for particular epitopes compared to polyclonal alternatives. Methodologically, this translates to more reproducible experimental outcomes when detecting PEG10 expression in different cellular contexts. For optimal results when conducting immunohistochemistry or Western blot analyses, use monoclonal antibodies targeting conserved regions of both RF1 and RF1/RF2 variants to ensure detection of all PEG10 isoforms. When investigating specific functions of individual PEG10 domains, select epitope-specific monoclonal antibodies that recognize distinct regions (gag domain versus protease domain) to differentiate the functional roles of each protein segment .

What are the recommended protocols for detecting PEG10 expression using monoclonal antibodies?

For detecting PEG10 expression in cancer cells or tissues, a multi-faceted approach yields the most reliable results. Start with Western blotting using 20-30μg of total protein lysate and a 1:1000 dilution of PEG10 monoclonal antibody, followed by overnight incubation at 4°C. For immunohistochemistry, optimize antigen retrieval by using citrate buffer (pH 6.0) with heat-induced epitope retrieval for 20 minutes. A dilution range of 1:100-1:200 typically produces optimal staining with minimal background. When performing immunofluorescence, include a blocking step with 5% normal serum matching the secondary antibody host species for 1 hour at room temperature to minimize non-specific binding. For flow cytometry applications, permeabilize cells with 0.1% Triton X-100 before antibody incubation to ensure access to intracellular PEG10 proteins. In all applications, validate specificity by including PEG10-knockout or siRNA-treated samples as negative controls .

How can researchers effectively validate PEG10 monoclonal antibody specificity?

Validating PEG10 monoclonal antibody specificity requires a combination of complementary approaches. First, perform side-by-side testing with at least two different PEG10 monoclonal antibodies targeting distinct epitopes to confirm consistent detection patterns. Second, implement genetic validation through siRNA or CRISPR-mediated knockdown/knockout of PEG10, which should result in corresponding diminished signal intensity in antibody-based detection assays. Third, conduct peptide competition assays using the specific immunogen peptide that was used to generate the antibody; pre-incubation with this peptide should abolish specific binding. Fourth, perform cross-validation with mRNA expression analysis (qRT-PCR or RNA-seq) to confirm that protein expression patterns correlate with transcript levels. Finally, include positive control cell lines with known high PEG10 expression (such as specific liver cancer or breast cancer cell lines) and negative control tissues where PEG10 should be minimally expressed .

What are the optimal experimental conditions for studying PEG10 protease activity?

To study PEG10 protease activity, researchers should design experiments that account for the unique characteristics of this retroviral-like protease. Methodologically, express both wild-type PEG10 (RF1/RF2) and a frameshift mutant form (fsRF1/RF2) in a suitable expression system such as HEK293T cells using optimized transfection protocols. Include a protease active-site mutant (D370A) as a crucial negative control. When conducting in vitro protease activity assays, use fluorogenic peptide substrates designed based on predicted cleavage site preferences, adjusting buffer conditions to pH 5.5-6.5 to match the optimal pH range for aspartic proteases. For monitoring autocatalytic processing, analyze protein samples at multiple time points (4, 8, 12, and 24 hours post-transfection) by Western blot using antibodies that can differentiate between processed and unprocessed forms. When assessing cellular effects of PEG10 protease activity, concurrently measure cell proliferation and viability using complementary assays (e.g., MTT/WST-1 and Annexin V/PI staining) to distinguish between effects on growth versus apoptosis inhibition .

How can researchers design experiments to investigate the regulatory mechanisms of PEG10 expression?

To investigate PEG10 regulatory mechanisms, implement a comprehensive experimental design that examines multiple levels of regulation. Begin with promoter analysis using luciferase reporter assays containing progressively truncated regions of the PEG10 promoter (approximately 2kb upstream of the transcription start site) to identify key regulatory elements. Follow this with chromatin immunoprecipitation (ChIP) assays targeting suspected transcription factors that might bind these regions. For epigenetic regulation studies, treat cells with DNA methyltransferase inhibitors (5-azacytidine) and histone deacetylase inhibitors (trichostatin A) at various concentrations (1-10μM) and timepoints (24-72h), then measure changes in PEG10 expression via qRT-PCR and Western blot. To investigate post-transcriptional regulation, perform RNA immunoprecipitation (RIP) assays with antibodies against RNA-binding proteins, followed by qRT-PCR for PEG10 mRNA. For translational control analysis, specifically examine the -1 frameshift mechanism by constructing dual-luciferase reporters containing the PEG10 frameshift signal between Renilla and firefly luciferase genes. Compare results across multiple cell types with varying PEG10 expression levels to identify tissue-specific regulatory mechanisms .

What approaches should be used to investigate PEG10's role in epithelial-mesenchymal transition (EMT) and drug resistance?

To investigate PEG10's role in EMT and drug resistance, implement a multi-faceted experimental approach beginning with manipulation of PEG10 expression levels. First, establish stable cell lines with PEG10 overexpression (both RF1 and RF1/RF2 variants) and knockdown/knockout using inducible systems that allow temporal control. For EMT analysis, examine morphological changes using phase-contrast microscopy and quantitatively assess a comprehensive panel of epithelial markers (E-cadherin, ZO-1, claudins) and mesenchymal markers (N-cadherin, vimentin, fibronectin) via immunoblotting, qRT-PCR, and immunofluorescence. Conduct functional EMT assays including cell migration (wound healing), invasion (transwell with Matrigel), and 3D spheroid formation with time-course imaging. For drug resistance studies, perform dose-response experiments with relevant therapeutic agents (e.g., CDK4/6 inhibitors for breast cancer models) in parental versus PEG10-modified cells, calculating IC50 values and resistance indices. Mechanistically investigate downstream targets by examining p21 and SIAH1 expression through Western blot and qRT-PCR, as these have been identified as mediators of PEG10-induced CDK4/6 inhibitor resistance. Validate findings using in vivo xenograft models treated with relevant therapeutics and monitor tumor growth, metastasis, and survival endpoints .

What are the most effective methods for studying the interaction between PEG10 and its binding partners?

To effectively study PEG10 protein interactions, employ a systematic approach combining complementary techniques. Begin with co-immunoprecipitation experiments using PEG10 monoclonal antibodies against different domains followed by mass spectrometry to identify novel binding partners without bias. Verify these interactions through reciprocal co-IP and Western blotting. For detailed binding characterization, use surface plasmon resonance (SPR) or isothermal titration calorimetry (ITC) with purified recombinant proteins to determine binding kinetics and thermodynamic parameters. To visualize protein interactions in their native cellular context, implement proximity ligation assays (PLA) or fluorescence resonance energy transfer (FRET) microscopy using fluorophore-tagged proteins. For mapping specific interaction domains, create a series of deletion mutants targeting different PEG10 regions and assess their binding capabilities. Establish the functional significance of key interactions by designing specific peptide inhibitors that competitively disrupt binding or by introducing targeted mutations that preserve protein structure while disrupting specific interaction interfaces. Complement these approaches with computational modeling to predict structural bases for observed interactions and guide experimental design .

How can PEG10-based endogenous virus-like particles (eVLPs) be engineered for cancer vaccine development?

Engineering PEG10-based eVLPs for cancer vaccines requires a systematic approach with careful attention to structural design and functional validation. Begin by genetically fusing selected tumor neoantigens to the C-terminal of the PEG10 gag domain, maintaining the natural self-assembly capabilities while incorporating antigenic peptides. Optimize the expression system using HEK293T cells and quantify productivity by measuring total protein yield (expect approximately 200-250μg from a 10cm dish of transfected cells compared to 70-80μg for control constructs). To enhance targeting to antigen-presenting cells, modify the eVLP surface with CpG-ODN, which interacts with dendritic cell DEC-205 receptors and stimulates TLR9 signaling. Purify the assembled eVLPs (designated as ePAC when CpG-modified) using ultracentrifugation at 28,000 rpm for 2 hours at 4°C through a 20% sucrose cushion. Validate neoantigen presentation by measuring uptake in bone marrow-derived dendritic cells and subsequent activation of neoantigen-specific T cells using interferon-gamma ELISPOT and intracellular cytokine staining assays. Test therapeutic efficacy in orthotopic tumor models, measuring not only tumor growth inhibition but also changes in tumor-infiltrating lymphocytes, neoantigen-specific T cell expansion, and combination effects with immune checkpoint inhibitors like anti-TIM-3 .

What strategies can researchers employ to target PEG10 for overcoming drug resistance in cancer therapy?

To target PEG10 for overcoming drug resistance, researchers should implement a multi-modal approach focusing on transcriptional and post-transcriptional inhibition strategies. First, design and synthesize PEG10-specific antisense oligonucleotides (ASOs) targeting conserved regions of PEG10 mRNA, screening multiple sequences for optimal knockdown efficiency (aim for >80% reduction in expression). Test these ASOs alone and in combination with relevant therapeutics (such as CDK4/6 inhibitors for breast cancer) in resistant cell lines, assessing synergistic effects through Combination Index calculations. For mechanistic understanding, analyze downstream effects on p21 and SIAH1 expression, as their suppression by PEG10 contributes to resistance mechanisms. Conduct in vivo efficacy studies using established PEG10-high xenograft models, administering ASOs (5-10mg/kg twice weekly) in combination with standard-of-care drugs, monitoring not only tumor volume but also pharmacodynamic markers in tumor biopsies. In parallel, develop small molecule inhibitors targeting the protease activity of PEG10 through structure-based drug design, screening initial candidates using in vitro protease assays before moving to cellular models. To establish clinical relevance, analyze PEG10 expression in patient samples before treatment and at progression, correlating expression levels with treatment outcomes and progression-free survival .

What considerations are important when developing PEG10 monoclonal antibodies for potential therapeutic applications?

When developing PEG10 monoclonal antibodies for therapeutic applications, researchers must address several critical considerations spanning antibody design, functional validation, and delivery optimization. For antibody engineering, focus on developing antibodies with high affinity (KD < 1nM) and specificity for functionally important domains of PEG10, particularly the regions involved in protein-protein interactions that drive oncogenic signaling. Test multiple antibody formats including conventional IgG, Fab fragments, and bispecific antibodies to identify optimal pharmacokinetic and tissue penetration properties. Conduct extensive cross-reactivity testing against homologous proteins to ensure target specificity and minimize off-target effects. For functional validation, assess both direct neutralization of PEG10 activity and potential for antibody-dependent cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC) in relevant cancer models. Evaluate potential immunogenicity through in silico T-cell epitope analysis and in vitro dendritic cell activation assays. For delivery optimization, explore various formulations including antibody-drug conjugates targeting PEG10-expressing cells, which may provide enhanced therapeutic index by delivering cytotoxic payloads specifically to cancer cells. Finally, develop companion diagnostic assays using the same antibody or complementary antibodies to identify patients with high PEG10 expression who would most likely benefit from PEG10-targeted therapies .

What are the current limitations in PEG10 monoclonal antibody research and how might they be addressed?

Current limitations in PEG10 monoclonal antibody research include issues with epitope specificity, cross-reactivity with related proteins, and inconsistent performance across different applications. To address these challenges, researchers should implement comprehensive validation protocols including parallel testing with multiple antibodies targeting different PEG10 epitopes, thorough examination in PEG10 knockout/knockdown systems, and cross-validation with orthogonal detection methods. The limited availability of well-characterized positive control samples presents another challenge; develop standardized reference materials with defined PEG10 expression levels to enable interlaboratory reproducibility. Additionally, current antibodies often cannot distinguish between the different PEG10 isoforms (RF1 vs. RF1/RF2); address this by developing isoform-specific antibodies targeting unique epitopes present only in one variant. Another significant limitation is the lack of antibodies optimized for less common applications such as ChIP-seq or proximity labeling; design application-specific validation protocols and modify existing antibodies with appropriate conjugates or tags to enhance performance in these specialized contexts. Finally, antibody batch variation impacts reproducibility; implement rigorous lot-testing procedures using standardized quantitative metrics before employing new antibody batches in critical experiments .

How can researchers integrate computational approaches with experimental methods to advance PEG10 research?

To effectively integrate computational approaches with experimental methods in PEG10 research, researchers should implement a comprehensive strategy spanning multiple levels of analysis. Begin with sequence-based predictions by performing phylogenetic analyses comparing PEG10 across species to identify evolutionarily conserved functional domains. Use this information to guide the design of domain-specific monoclonal antibodies. For structural insights, develop homology models of PEG10 protease domains based on related retroviral proteases, as shown in the research where substrate binding site comparisons revealed similarities with HIV proteases. Use these models to predict potential cleavage sites and design targeted inhibitors or substrate mimetics that can be experimentally validated. At the systems level, employ network analysis algorithms to integrate proteomics data from PEG10 interaction studies with transcriptomics data from PEG10 manipulation experiments, identifying hub genes that may represent key functional nodes in PEG10-mediated pathways. For translational applications, implement machine learning approaches to analyze clinical datasets, correlating PEG10 expression patterns with treatment outcomes and patient survival to stratify patients who might benefit from PEG10-targeted therapies. Finally, use molecular dynamics simulations to examine the structural consequences of mutations in PEG10 or to predict the binding modes of potential therapeutic antibodies, informing the design of higher-affinity variants with optimized properties .

What emerging technologies might transform our understanding of PEG10 function and therapeutic targeting?

Emerging technologies poised to transform PEG10 research span multiple disciplines and methodological approaches. Single-cell multi-omics integration combines single-cell RNA sequencing with proteomics and epigenetic profiling, enabling researchers to map PEG10 expression heterogeneity across different cell populations within tumors and correlate it with cellular states and therapeutic responses. Implement this approach in patient-derived samples to identify specific cellular contexts where PEG10 drives malignant phenotypes. CRISPR-based technologies offer unprecedented precision for functional studies; apply CRISPR activation/interference systems for temporal control of PEG10 expression and CRISPR base/prime editing for introducing specific mutations that affect particular domains without disrupting the entire protein. Structural biology advancements, particularly cryo-electron microscopy, can reveal the native conformation of PEG10 assemblies and virus-like particles, informing rational design of inhibitors and therapeutic antibodies. Advanced imaging techniques such as live-cell super-resolution microscopy combined with optogenetic tools allow real-time visualization and manipulation of PEG10 localization and interactions in living cells. For therapeutic development, novel antibody engineering platforms including nanobodies, intrabodies, and protac-antibody conjugates offer innovative approaches to target PEG10 with greater specificity and efficacy than conventional methods. Finally, organ-on-chip and patient-derived organoid technologies provide physiologically relevant systems to evaluate PEG10-targeted therapies before advancing to clinical studies .

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 2024 Thebiotek. All Rights Reserved.