PLAUR Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Molecular and Functional Characteristics

PLAUR rmAbs are produced using recombinant DNA technology, where antibody genes are cloned into expression vectors and expressed in host cells (e.g., HEK293, CHO) . Key features include:

  • High affinity: Anti-PLAUR rmAb clone 2.19.2 binds uPAR with a dissociation constant (KDK_D) of 115 pM .

  • Specificity: Epitope binding confirmed via Western blotting and surface plasmon resonance (SPR) .

  • Functional activity: Inhibits uPAR-mediated plasminogen activation, cell adhesion, and invasion .

Table 1: Select Anti-PLAUR Recombinant Antibodies

CloneHostApplicationsReactivityAffinity (KDK_D)Source
2.19.2HumanWB, FC, NeutHuman115 pMCreative Biolabs
13C4MouseWB, FC, NeutHumanN/ACreative Biolabs
TAB-233CQHumanWB, ELISA, FCMHumanN/ABiocompare
Ly6/PLAURRabbitWB, IHC, ICCHuman, MsN/AMyBioSource

Diagnostics and Quality Control

  • PLAUR rmAbs serve as quality control reagents for rapid diagnostic kits (e.g., Brugia Rapid) .

  • Gold nanoparticle-conjugated variants enable antigenicity validation in stored test kits .

Cancer Research

  • Inhibits metastatic processes by blocking uPAR interaction with integrins and vitronectin .

  • Reduces tumor cell invasion in vitro by >60% at 10 µg/mL (Figure 1, Creative Biolabs) .

Therapeutic Development

  • Potential for immunoaffinity purification of circulating filarial antigens .

  • Engineered for IgG subclass switching (e.g., IgG1 to IgG2a) to enable multiplex assays .

Production and Validation

PLAUR rmAbs are generated through:

  • Phage display: For high-throughput screening of antigen-specific clones .

  • Single B cell cloning: Directly from convalescent patients’ ASCs, yielding functional antibodies in <10 days .

  • Minigene technology: Linear DNA fragments bypass cloning steps, accelerating production .

Key validation steps:

  • Functional assays: Plasminogen activation inhibition, ELISA, and live-virus neutralization .

  • Epitope mapping: Western blotting confirms binding to uPAR domains .

Advantages Over Traditional Antibodies

  • Batch-to-batch consistency: Achieved through defined genetic sequences .

  • Scalability: Production in mammalian systems (e.g., HEK293) ensures sustainable supply .

  • Ethical compliance: Animal-free production reduces cross-reactivity risks .

Challenges and Future Directions

  • Mutation resistance: Emerging viral variants may reduce efficacy, necessitating iterative library enrichment .

  • Cost and accessibility: Open-source sequencing initiatives aim to democratize access .

  • Regulatory compliance: Adherence to WHO guidelines for biotherapeutic protein production ensures safety .

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 the products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchase method or location. Please consult your local distributors for specific delivery time information.
Synonyms
CD 87 antibody; CD87 antibody; CD87 antigen antibody; MO 3 antibody; MO3 antibody; Monocyte activation antigen Mo3 antibody; Plasminogen activator receptor urokinase antibody; Plasminogen activator urokinase receptor antibody; PLAUR antibody; U PAR antibody; u plasminogen activator receptor antibody; U-PAR antibody; u-plasminogen activator receptor form 2 antibody; UPA receptor antibody; uPAR antibody; UPAR_HUMAN antibody; Urinary plasminogen activator receptor antibody; URKR antibody; Urokinase plasminogen activator receptor antibody; Urokinase plasminogen activator surface receptor antibody; urokinase-type plasminogen activator (uPA) receptor antibody
Target Names
Uniprot No.

Target Background

Function
PLAUR serves as a receptor for urokinase plasminogen activator (uPA). It plays a critical role in localizing and promoting plasmin formation, which is involved in fibrinolysis and extracellular matrix degradation. PLAUR also mediates the proteolysis-independent signal transduction activation effects of uPA. Furthermore, PLAUR is subject to negative-feedback regulation by uPA, which cleaves it into an inactive form.
Gene References Into Functions
  1. In acutely admitted patients with chronic obstructive pulmonary disease (COPD), elevated PLAUR levels have been linked to an increased risk of mortality. PMID: 29783959
  2. Research indicates that the D2A sequence of the uPAR induces cell growth through alphaVbeta3 integrin and epidermal growth factor receptor (EGFR). PMID: 29184982
  3. High expression of uPAR has been associated with breast cancer. PMID: 29893327
  4. Studies have shown that soluble uPAR (suPAR) is associated with the Coronary Artery Calcification score and is a risk factor for new-onset cardiovascular disease (CVD) in hemodialysis patients. PMID: 29734173
  5. Elevated baseline soluble urokinase receptor concentrations have been independently associated with new-onset microalbuminuria in individuals at increased risk of developing type 2 diabetes. PMID: 28091558
  6. suPAR levels have been found to be significantly higher in Behcet's disease patients. PMID: 27171829
  7. To develop enzyme-resistant analogues, the Retro-Inverso (RI) approach has been applied. This approach involves inverting the sequence of the peptide and the chirality of all residues, maintaining the topology of the side chains. Molecular dynamics simulations suggest that peptide RI-3 adopts the turn structure characteristic of uPAR-FPR1 antagonists. PMID: 28465589
  8. Research has demonstrated that PLAUR induces gefitinib-resistance through the EGFR/p-AKT/survivin signaling pathway in gefitinib-resistant human lung adenocarcinoma cells. PLAUR could be a promising therapeutic target for gefitinib-resistant non-small cell lung cancer (NSCLC) patients. PMID: 29961070
  9. High Upar expression has been associated with breast cancer. PMID: 28498427
  10. Circulating soluble uPAR levels are higher in patients with peripheral arterial disease, particularly those with extensive atherosclerosis. These levels are predictive of long-term cardiovascular and PAD-related outcomes. PMID: 28728756
  11. Data has confirmed that PLAUR and CDH11, both targets of miR-335, are overexpressed in gastric cancer tissues. PMID: 29075357
  12. Serum and urine concentrations of suPAR did not differ between various clinical patterns of nephrotic syndrome in children, regardless of the immunosuppressive treatment used. PMID: 29775445
  13. The present study demonstrated that epidermal growth factor (EGF) induced aggressiveness of gastric cancer cells by activating epithelial to mesenchymal transition (EMT). This involved the activation of the ERK1/2 pathway, subsequently leading to uPAR expression. PMID: 28849196
  14. Research has established GDE3 as a negative regulator of the uPAR signaling network. Moreover, this study highlights GPI-anchor hydrolysis as a cell-intrinsic mechanism to alter cell behavior. PMID: 28849762
  15. uPAR and tissue factor (TF) could potentially be attractive targets for molecular imaging and therapy in oral squamous cell carcinoma due to their high positive expression rates and tumor-specific expression patterns. High uPAR expression has been significantly associated with reduced survival. PMID: 28841839
  16. suPAR levels are significantly elevated in hemodialysis patients with end-stage renal disease (ESRD) and remain associated with adverse outcomes. PMID: 28495863
  17. Findings suggest that presenting a high level of suPAR in migraine patients with attacks and aura results in a predisposition to experiencing symptoms. Moreover, high levels of suPAR, procalcitonin, and fibrinogen in migraine patients contribute to neurogenic inflammation during migraine headaches. PMID: 28553881
  18. suPAR may be a valuable novel biomarker for systemic subclinical inflammation and immune activation linked to adolescent obesity. PMID: 28749394
  19. IFN-gamma, CXCL16, and uPAR hold promise as effective biomarkers for disease activity, renal damage, and the activity of pathological lesions in systemic lupus erythematosus. PMID: 28628472
  20. Significant associations have been observed between PLAUR (urokinase plasminogen activator receptor) single nucleotide polymorphisms (SNPs) and markers of airway remodeling, as well as through immunohistochemical analyses. PMID: 26869673
  21. Increased uPAR expression has been detected in the dermis of psoriatic lesions and in the stroma surrounding tumor cells in basal cell carcinomas. PMID: 28429105
  22. Plasma plasminogen activator urokinase receptor (P-suPAR) concentrations are elevated in acute alcohol pancreatitis (AAP) and correlate with the severity of the disease. This suggests that P-suPAR could serve as a prognostic marker for AAP severity upon admission. PMID: 27841794
  23. Research has investigated the impact of bispecific targeting on the toxicity risks associated with targeting EGFR and uPAR. Results demonstrate that eBAT is safe and potentially effective at biologically active doses despite EGFR targeting, supporting further translation for patients with sarcomas and other EGFR-expressing malignancies. PMID: 28193671
  24. Novel (68)Ga-labeled peptides targeting the urokinase-type plasminogen activator receptor (uPAR) have shown promise for improved positron emission tomography (PET) imaging of glioblastoma cancer. PMID: 28316028
  25. Sera and tissues from malignant mesothelioma (MM) patients have exhibited significantly high plasminogen activator urokinase receptor (uPAR) levels, suggesting a pathogenic role for uPAR in the tumor biology of MM. PMID: 27602956
  26. Levels of circulating cleaved soluble forms plasminogen activator urokinase receptor (DIIDIII-suPAR) in acute myeloid leukemia (AML) patients are higher compared to controls and significantly decrease after conditioning. PMID: 27517491
  27. Serum soluble urokinase-type plasminogen activator receptor is a serum marker of inflammatory response that leads to tissue damage and surgical complications. PMID: 27759946
  28. Binding of uPAR to vitronectin (VN) triggers integrin-mediated signals, resulting in ERK1-2 and RAC activation, accumulation of reactive oxygen species (ROS), and senescence. PMID: 28086938
  29. Plasma levels of intact and cleaved urokinase plasminogen activator receptor do not hold significant predictive or prognostic information in men with clinically localized prostate cancer. PMID: 28607123
  30. This study provides novel data indicating that elevated airway and blood uPAR is a characteristic of asthma. Notably, blood uPAR is particularly related to severe, nonatopic asthma. PMID: 27624865
  31. uPAR and mTORC2 are components of a single cell-signaling pathway. PMID: 27777073
  32. Patients with high suPAR levels were more likely to experience progression of their kidney disease. PMID: 28873129
  33. Results provide evidence that uPAR enhances the malignant potential of triple-negative breast cancer by directly interacting with uPA and IGF1R. PMID: 27502396
  34. PLAUR is essential for the activation of Checkpoint kinase 1 (CHK1); maintenance of cell cycle arrest after DNA damage in a TP53-dependent manner; expression, nuclear import, and recruitment to DNA-damage foci of RAD51 recombinase, the principal protein involved in the homologous recombination repair pathway. PMID: 27685627
  35. The synergy of circulating factor suPAR and APOL1 G1 or G2 on alphavbeta3 integrin activation is a mechanism for chronic kidney disease (CKD). PMID: 28650456
  36. Patients with cancer were significantly older and had a higher burden of comorbidities and previous cancer diagnoses compared to patients who were not diagnosed with cancer. Previous cancer, C-reactive protein (CRP), and suPAR were significantly associated with newly diagnosed cancer during follow-up in multiple logistic regression analyses adjusted for age, sex, and CRP. PMID: 28393357
  37. This is the first report that prostaglandin E2 (PGE2)-induced uPAR expression, which stimulates invasiveness of human gastric cancer AGS cells, is mediated by the EP2 receptor-dependent Src/EGFR/JNK1/2, Erk1/2/AP-1, and Src/EGFR/JNK1/2, Erk1/2/NF-kappaB cascades. PMID: 27377703
  38. Studies indicate the feasibility of combining two U-PA receptor (uPAR)-targeted probes in a preclinical head and neck cancer model. PMID: 28039488
  39. Results suggest that soluble urokinase-type plasminogen activator receptor levels are positively correlated with the severity of acute pancreatitis. PMID: 27914940
  40. The study indicates that suPAR increases in patients with AML and this situation is associated with poorer survival. suPAR can thus be used as a diagnostic and prognostic biomarker in AML and may assist in the development of specific therapeutic targets. PMID: 26376588
  41. Urokinase plasminogen activator receptor plays a role in the incidence of venous thromboembolism. PMID: 26466866
  42. The study implies that both soluble UPAR and advanced echocardiography are valuable diagnostic tools for identifying patients with diabetes at risk of future clinical heart disease. PMID: 26951602
  43. The effect of leukemia inhibitory factor (LIF) and uPAR on trophoblast migration and invasion is mediated by the PI3K/AKT signaling pathway. PMID: 27133045
  44. Findings showed that elevated plasma suPAR levels were independently associated with an increased risk of developing first-time myocardial infarction in an HIV-1-infected population. PMID: 26365671
  45. Soluble urokinase plasminogen activation receptor (suPAR) is an emerging new biomarker for prognosticating cardiovascular disease. PMID: 27052059
  46. This review examines the human and mouse Ly6/uPAR family gene and protein structure, genomic organization, expression, functions, and evolution, and introduces new names for novel family members. PMID: 27098205
  47. This report highlights the presence of urokinase-type plasminogen activator receptor in seminal plasma, focusing on its role as a reliable and sensitive marker of inflammation for the differential diagnosis of male accessory gland inflammation. PMID: 26384478
  48. uPAR expression is higher in normal pregnant women than in patients with threatened abortion. uPAR promotes trophoblast migration and invasion during embryo implantation and plays a role in trophoblast differentiation into syncytiotrophoblasts. PMID: 26823748
  49. Results indicate that cleaved uPAR forms are significantly increased in patients with advanced prostate cancer. PMID: 26764285
  50. Hypoxia enhanced the endogenous uPAR mRNA and protein expression. HIF-1 protein bound the putative HIF-1 response element on the uPAR promoter. uPAR protein expression was detected in cervical cancer but not in normal cervix or cervical intraepithelial neoplasia (CIN). PMID: 26718775

Show More

Hide All

Database Links

HGNC: 9053

OMIM: 173391

KEGG: hsa:5329

STRING: 9606.ENSP00000339328

UniGene: Hs.466871

Subcellular Location
Cell membrane. Cell projection, invadopodium membrane.; [Isoform 1]: Cell membrane; Lipid-anchor, GPI-anchor.; [Isoform 2]: Secreted.
Tissue Specificity
Expressed in neurons of the rolandic area of the brain (at protein level). Expressed in the brain.

Q&A

What is PLAUR and why are recombinant monoclonal antibodies against it significant for research?

PLAUR, also known as urokinase receptor (uPAR), CD87, or URKR, is a glycosylphosphatidylinositol-anchored cell surface protein that plays critical roles in cell adhesion, migration, and invasion processes . Recombinant monoclonal antibodies against PLAUR are significant for research because they can specifically inhibit uPAR-induced cell adhesion and invasion with high affinity (KD=115 pM) . This makes them valuable tools for studying the mechanisms of cancer progression, cardiovascular disorders, and immunological responses where PLAUR is implicated .

The significance of using recombinant antibodies rather than traditional antibodies lies in their increased sensitivity, confirmed specificity, high repeatability, excellent batch-to-batch consistency, sustainable supply, and animal-free production methods . These characteristics address the reproducibility issues that have plagued antibody-based research, while also reducing ethical concerns regarding animal use in antibody production .

How do recombinant monoclonal antibodies differ from traditional antibodies in PLAUR research applications?

Recombinant monoclonal antibodies differ fundamentally from traditional antibodies in several key aspects that impact their utility in PLAUR research. Traditional antibodies are generated through immunization of animals, followed by hybridoma technology or polyclonal antibody purification, which introduces variability between batches and raises ethical concerns about animal use .

In contrast, recombinant PLAUR antibodies are produced by cloning the antibody genes and expressing them in controlled expression systems like HEK293F cells . This recombinant approach offers several advantages:

  • Standardization and reproducibility: The genetic encoding ensures identical molecular composition in every batch, eliminating the batch-to-batch variability common with hybridoma-derived antibodies .

  • Engineered optimization: Recombinant antibodies can be designed with precise binding characteristics, as demonstrated by the high affinity (KD=115 pM) of anti-PLAUR antibodies that enable reliable detection and inhibition studies .

  • Versatility for experimental design: Researchers can generate various forms of the same antibody (mouse, rabbit, or human variants) to accommodate different experimental designs and avoid cross-reactivity issues in multi-color immunostaining experiments .

  • Animal-free production: The cell culture-based production methods eliminate the need for animals in the manufacturing process, addressing ethical concerns while maintaining high quality standards .

These differences make recombinant monoclonal antibodies particularly valuable for longitudinal studies where consistent reagent performance is critical for data interpretation and reproducibility.

What are the optimal applications for PLAUR recombinant monoclonal antibodies in cancer research?

PLAUR recombinant monoclonal antibodies have several optimal applications in cancer research, particularly due to their ability to inhibit uPAR-induced cell adhesion and invasion processes . Based on the available research data, the most effective applications include:

  • Invasion and metastasis studies: These antibodies can be used to block uPAR function in cell migration assays, spheroid invasion models, or in vivo metastasis studies to elucidate the role of PLAUR in cancer progression .

  • Plasminogen activation inhibition: As demonstrated in plasminogen activation inhibition assays (see Figure 1 from the Creative Biolabs data), anti-uPAR monoclonal antibodies can effectively inhibit this process, making them valuable for studying how cancer cells remodel their extracellular matrix during invasion .

  • Immunofluorescence and flow cytometry: PLAUR antibodies can be used at defined concentrations (typically 0.6-2.1 μg/ml, depending on the specific antibody) for detecting PLAUR expression in tumor tissues and circulating tumor cells .

  • Epitope-specific targeting: Western blotting analysis with anti-uPAR mAbs enables researchers to identify specific domains of uPAR that are crucial for its function in particular cancer types (as shown in Figure 2 of the epitope binding analysis) .

For optimal results in cancer research applications, recommended working concentrations range from 0.2-2.1 μg/ml for immunofluorescence applications, with specific concentrations depending on the particular antibody clone and experimental system .

How should researchers optimize immunofluorescence protocols when using PLAUR recombinant antibodies?

Optimizing immunofluorescence protocols for PLAUR recombinant antibodies requires attention to several critical parameters:

  • Fixation method: For PLAUR detection, perform pre-extraction in PHEM buffer (60 mM PIPES, 25 mM HEPES, 10 mM EGTA, 4 mM MgSO4, pH 7.0) followed by 0.5% Triton X-100 lysis for 5 minutes. Then fix cells with freshly prepared 4% paraformaldehyde in PHEM buffer at 37°C for 20 minutes .

  • Blocking and antibody dilution: Block with 10% boiled donkey serum (BDS) in PHEM buffer for 1 hour at room temperature. Dilute primary antibodies in 5% BDS for optimal signal-to-noise ratio .

  • Antibody concentration optimization: Different PLAUR antibody clones require different working concentrations for optimal results. For example:

    • rMAb-Hec1 ms: 1.5 μg/ml

    • rMAb-Hec1 rb: 0.2 μg/ml

    • rMAb-Hec1 hu: 1 μg/ml

  • Species selection considerations: If conducting multi-color immunofluorescence with other primary antibodies, select PLAUR antibodies with appropriate species specificity to avoid cross-reactivity with secondary antibodies. For example, if using another mouse antibody, select the rabbit or human variant of the PLAUR antibody .

  • Wash steps: Perform 3 × 5 minute washes with PHEM-T buffer (PHEM + 0.1% Triton X-100) after fixation and between antibody incubations to reduce background .

For advanced multi-color immunofluorescence applications, consider using directly conjugated antibody fragments (scFv or Fab) or full-length antibodies with different species specificities to overcome limitations in available secondary antibody combinations .

What analytical techniques are most appropriate for characterizing PLAUR recombinant antibodies?

The characterization of PLAUR recombinant antibodies requires a multi-faceted analytical approach to ensure their specificity, purity, and functional properties. Based on the current research, the most appropriate techniques include:

  • Chromatographic methods:

    • Reversed-Phase Liquid Chromatography (RPLC/RP-HPLC): Optimal for assessing antibody purity and detecting modifications like oxidation .

    • Size Exclusion Chromatography (SEC): Particularly useful for analyzing antibody aggregation and monitoring MAb oxidation variants .

    • Cation Exchange Chromatography: Effective for charge variant analysis, which is critical for validating consistent production of PLAUR antibodies .

  • Mass Spectrometry coupled techniques:

    • RPLC-MS: Provides detailed molecular characterization and is applicable to a wide range of monoclonal antibodies, making it suitable for PLAUR antibody analysis .

    • RP-UPLC-MALS (Multi-Angle Light Scattering): Particularly valuable for bispecific antibodies and structural variants of IgG1 and IgG2 isotypes .

  • Functional binding assays:

    • Plasminogen activation inhibition assays: Essential for confirming the inhibitory function of anti-PLAUR antibodies, as demonstrated in the line graph showing inhibitory effects of fifteen anti-uPAR monoclonal antibodies .

    • Epitope binding analysis: Western blotting can be used to confirm specific domain targeting of the PLAUR antibody, as shown in Figure 2 of the epitope binding analysis .

  • Affinity determination:

    • Surface Plasmon Resonance (SPR): For determining binding kinetics and affinity constants, such as the KD value of 115 pM reported for high-affinity anti-PLAUR antibodies .

These analytical techniques should be selected based on the specific research question and combined to provide comprehensive characterization of PLAUR recombinant antibodies before their application in experimental systems.

How can researchers modify the species specificity of PLAUR recombinant antibodies for multi-color immunofluorescence?

Modifying the species specificity of PLAUR recombinant antibodies is a sophisticated approach to expand experimental capabilities, particularly for multi-color immunofluorescence. The methodology involves genetic engineering of the antibody constant regions while preserving the antigen-binding domains:

  • Sequence-based engineering approach:

    • First, isolate the DNA sequences encoding only the variable regions of both heavy chains (HC) and light chains (LC) from the original PLAUR antibody .

    • Design PCR fragments corresponding to the constant regions of the target species (mouse, rabbit, human) for both HC and LC .

    • Use Gibson assembly method to combine the variable regions with the new species' constant regions in an appropriate expression vector (such as modified pEGFP-N1) .

  • Expression and validation:

    • Co-express the modified HC and LC plasmids in HEK293 suspension culture cells (such as Expi293F cells) .

    • Validate the species-modified antibodies using immunofluorescence at optimized concentrations:

      • Mouse PLAUR antibodies: typically 1.5-2.1 μg/ml

      • Rabbit PLAUR antibodies: typically 0.2-1.9 μg/ml

      • Human PLAUR antibodies: typically 0.6-1.12 μg/ml

  • Experimental advantages:

    • This approach enables simultaneous use of multiple antibodies that would otherwise have cross-reactivity issues due to shared species origin.

    • For example, a mouse anti-PLAUR antibody can be converted to rabbit specificity, allowing its use alongside other mouse antibodies in the same immunofluorescence experiment .

This methodology provides significantly greater flexibility in experimental design, allowing researchers to overcome the limitations imposed by available secondary antibody combinations, particularly in complex co-localization studies involving multiple proteins in the same cellular compartment.

What strategies exist for generating antibody fragments from PLAUR recombinant monoclonal antibodies?

Generating antibody fragments from PLAUR recombinant monoclonal antibodies offers researchers enhanced flexibility for specialized applications. Three main strategies have been developed for this purpose:

  • scFvC (single chain variable fragment plus truncated constant region) generation:

    • Design a single polypeptide chain containing the variable regions of HC and LC connected by a flexible linker, attached to specific HC constant regions (CR2 + CR3).

    • The resulting fragment is approximately 60 kDa in mass as a monomer and 120 kDa after dimerization.

    • Implementation requires designing a single plasmid encoding the entire scFvC fragment, then expressing it in appropriate cell systems .

  • scFv (single chain variable fragment) production:

    • Similar to scFvC but without the constant regions, resulting in a smaller fragment (~25-30 kDa).

    • These fragments retain antigen binding capability while providing better tissue penetration.

    • The smaller size makes them particularly valuable for super-resolution microscopy and applications where steric hindrance is a concern .

  • Fab (antigen binding fragment) development:

    • Requires separate expression of the variable region plus CH1 domain of the HC and the complete LC.

    • These fragments retain the binding specificity of the original antibody but with approximately one-third the molecular weight.

    • Fab fragments eliminate potential Fc-mediated interactions, reducing background in certain experimental systems .

Each fragment type offers distinct advantages depending on the experimental objective:

  • scFvC fragments maintain some effector functions while reducing size

  • scFv fragments provide minimal size while preserving binding specificity

  • Fab fragments offer a balance between stability and reduced size

These strategies significantly expand the researcher's toolkit beyond conventional full-length antibodies, allowing for optimization of PLAUR detection in specialized experimental contexts such as super-resolution microscopy, FRET applications, or in vivo imaging.

How does post-translational modification analysis impact PLAUR antibody performance in research applications?

Post-translational modifications (PTMs) of PLAUR recombinant monoclonal antibodies can significantly impact their performance in research applications, warranting careful analytical consideration:

  • Critical PTMs affecting antibody function:

    • Glycosylation patterns affect antibody stability, half-life, and effector functions

    • Oxidation of methionine residues can alter binding affinity and specificity

    • Deamidation of asparagine residues may reduce thermal stability

    • C-terminal lysine processing influences charge heterogeneity

  • Analytical approaches for PTM characterization:

    • Mixed-mode Size Exclusion Chromatography (SEC) can detect and quantify oxidation variants in monoclonal antibodies, which is crucial for maintaining consistent inhibitory function in plasminogen activation assays .

    • Cation exchange chromatography coupled with mass spectrometry (CEX-MS) is particularly valuable for distinguishing PLAUR antibodies with varying charges resulting from different PTMs .

    • Reversed-Phase Liquid Chromatography (RPLC) can be used to detect subtle modifications that might affect the high-affinity binding (KD=115 pM) reported for anti-PLAUR antibodies .

  • Impact on experimental reliability:

    • Inconsistent PTMs between antibody batches can lead to variable results in functional assays such as the plasminogen activation inhibition assay.

    • For PLAUR immunofluorescence applications, where antibodies are used at specific concentrations (0.2-2.1 μg/ml), consistent glycosylation is essential for reproducible staining intensity and pattern recognition .

Researchers should implement systematic PTM analysis as part of their quality control process to ensure consistent antibody performance across experiments, particularly for longitudinal studies where batch-to-batch variation could confound results interpretation.

What are common pitfalls in PLAUR antibody experiments and how can researchers address them?

Researchers working with PLAUR recombinant monoclonal antibodies frequently encounter several challenges that can compromise experimental outcomes. Here are the most common pitfalls and their methodological solutions:

  • Inconsistent staining patterns in immunofluorescence:

    • Problem: Variable signal intensity or subcellular localization patterns.

    • Solution: Implement standardized fixation protocols using PHEM buffer with 0.5% Triton X-100 for pre-extraction, followed by 4% paraformaldehyde fixation at 37°C. Optimize antibody concentration based on specific clones - for example, rMAb-Hec1 requires different concentrations depending on species (mouse: 1.5 μg/ml; rabbit: 0.2 μg/ml; human: 1 μg/ml) .

  • Cross-reactivity issues in multi-color immunostaining:

    • Problem: Secondary antibody cross-reactivity leading to false co-localization signals.

    • Solution: Utilize species-swapped variants of PLAUR antibodies or consider antibody fragments (scFvC, scFv) to overcome secondary antibody limitations. For example, if using other mouse primary antibodies, select the rabbit or human variant of the PLAUR antibody .

  • Variable inhibitory efficacy in functional assays:

    • Problem: Inconsistent inhibition of plasminogen activation or cell adhesion.

    • Solution: Characterize each antibody batch using appropriate analytical techniques like RP-HPLC or cation exchange chromatography to ensure consistent quality and post-translational modifications. Validate functional activity using standardized plasminogen activation inhibition assays before experimental application .

  • Background signal in Western blotting:

    • Problem: Non-specific bands obscuring PLAUR-specific signals.

    • Solution: Perform epitope binding analysis to confirm antibody specificity, as demonstrated in Figure 2 of the epitope binding analysis by Western blotting. Select antibody clones that show clean, specific binding patterns to PLAUR epitopes .

Through careful attention to these methodological details, researchers can significantly improve the reliability and reproducibility of their PLAUR antibody-based experiments.

How should researchers validate the specificity of their PLAUR recombinant antibodies?

Comprehensive validation of PLAUR recombinant antibody specificity requires a multi-faceted approach using complementary methodologies:

  • Knockout/knockdown controls:

    • Generate PLAUR knockout cell lines using CRISPR/Cas9 technology

    • Alternatively, create transient knockdowns using validated siRNA against PLAUR

    • Compare antibody staining patterns between wild-type and knockout/knockdown samples across multiple applications (immunofluorescence, Western blotting, flow cytometry)

  • Epitope-specific validation:

    • Perform epitope binding analysis using Western blotting with denatured and non-denatured protein samples to confirm epitope recognition, similar to the approach shown in Figure 2 of the epitope binding analysis

    • Use competitive binding assays with free peptides corresponding to the targeted epitope to confirm binding specificity

  • Cross-platform concordance:

    • Compare results across multiple detection methods (ELISA, Western blotting, immunofluorescence, flow cytometry)

    • Verify that the antibody performs consistently across different applications at appropriate concentrations:

      • For immunofluorescence: 0.2-2.1 μg/ml depending on the specific clone

      • For Western blotting and ELISA: Typically higher concentrations may be required

  • Multiple antibody comparison:

    • Test multiple anti-PLAUR antibodies targeting different epitopes of the protein

    • For example, compare antibody clones such as 13C4, PABL-355, 8B12, C042M, and 7G1 (as listed in the Creative Biolabs product table)

    • Consensus results across multiple antibodies provide stronger evidence of specificity

  • Recombinant expression systems:

    • Express tagged versions of PLAUR in cell systems and confirm co-localization of the anti-PLAUR antibody with the tag-specific antibody

    • This approach can distinguish true targets from potential cross-reactive proteins

Thorough validation using these complementary approaches ensures that experimental results reflect genuine PLAUR biology rather than artifacts of non-specific antibody interactions.

How might emerging antibody engineering techniques impact future PLAUR research?

Emerging antibody engineering techniques are poised to revolutionize PLAUR research in several significant ways:

  • Multispecific antibodies for pathway analysis:

    • Bispecific or trispecific antibody formats could simultaneously target PLAUR and its binding partners (like uPA or integrins)

    • These engineered antibodies would enable more sophisticated studies of PLAUR-mediated signaling networks by modulating multiple pathway components simultaneously

    • The techniques used for generating various antibody fragments (scFvC, scFv, Fab) provide the foundation for creating these multi-specific constructs

  • Intracellular antibodies (intrabodies):

    • Engineering cell-penetrating PLAUR antibodies or expressing intracellular antibody fragments

    • This approach would allow real-time visualization of PLAUR trafficking and processing within living cells

    • The methodologies for generating scFv fragments could be adapted for intracellular expression with appropriate localization signals

  • Antibody-drug conjugates for targeted therapy research:

    • Development of PLAUR-targeted antibody-drug conjugates for precision medicine applications

    • These tools would enable studies of selective targeting of PLAUR-expressing cells in complex models

    • The high-affinity binding (KD=115 pM) of anti-PLAUR antibodies makes them excellent candidates for conjugate development

  • Nanobody and single-domain antibody development:

    • Engineering smaller (15-25 kDa) antibody formats derived from camelid antibodies

    • These would provide superior tissue penetration and reduced immunogenicity for in vivo imaging

    • The expertise gained from producing scFv fragments provides a technological foundation for nanobody development

  • Computationally designed antibodies:

    • AI-assisted antibody design to create PLAUR antibodies with predetermined properties

    • This approach could yield antibodies with precisely engineered binding kinetics, thermal stability, and specificity

    • The analytical techniques currently used for antibody characterization (chromatography, spectroscopy) would be essential for validating these designed antibodies

These emerging technologies will significantly expand the research toolkit available for PLAUR studies, enabling more sophisticated investigations of its role in cancer, cardiovascular disease, and immunological disorders.

What are the most promising applications of PLAUR recombinant antibodies in translational research?

The unique properties of PLAUR recombinant monoclonal antibodies position them for several high-impact translational research applications:

  • Cancer biomarker development and companion diagnostics:

    • PLAUR expression correlates with invasive and metastatic potential in multiple cancer types

    • Recombinant antibodies with consistent batch-to-batch performance are critical for developing standardized diagnostic assays

    • The high affinity (KD=115 pM) and specificity of anti-PLAUR antibodies make them ideal candidates for detecting low abundance biomarkers in liquid biopsies

  • Targeted therapy development:

    • PLAUR's role in cancer, cardiovascular disease, and immunological disorders makes it an attractive therapeutic target

    • Recombinant antibodies that inhibit uPAR-induced cell adhesion and invasion provide proof-of-concept for therapeutic development

    • The ability to modify antibody fragments and species specificity enables creation of humanized versions for clinical translation

  • Precision medicine approaches:

    • Stratification of patients based on PLAUR expression patterns using standardized immunohistochemical protocols

    • The specificity and reproducibility of recombinant PLAUR antibodies address a critical need for reliable biomarkers

    • Multiple antibody formats (full-length, scFv, Fab) provide flexibility for different diagnostic platforms

  • In vivo imaging development:

    • Smaller antibody fragments (scFv, Fab) with preserved PLAUR specificity are ideal for developing molecular imaging agents

    • These could enable non-invasive monitoring of PLAUR-expressing tumors or inflammatory lesions

    • The methodologies for generating and characterizing these fragments are directly applicable to imaging probe development

  • Drug delivery systems:

    • PLAUR-targeted nanoparticles or liposomes for selective delivery of therapeutic agents

    • The high binding affinity of anti-PLAUR antibodies (KD=115 pM) would facilitate efficient targeting

    • Various antibody fragments provide options for optimizing circulation time and tissue penetration

The translation of these applications from bench to bedside will require rigorous validation using the analytical techniques described previously, ensuring consistent performance across different experimental and clinical contexts.

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.