PDPN Antibody

Podoplanin, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Overview of PDPN Antibody

Podoplanin (PDPN) antibodies are monoclonal or polyclonal antibodies targeting PDPN, a transmembrane glycoprotein overexpressed in cancer cells, cancer-associated fibroblasts (CAFs), and inflammatory macrophages . PDPN promotes tumor progression through mechanisms like epithelial-mesenchymal transition (EMT), metastasis, and immunosuppression . Antibodies against PDPN are used for diagnostic, prognostic, and therapeutic purposes in cancers such as glioblastoma, squamous cell carcinoma, mesothelioma, and melanoma .

Mechanisms of Action

PDPN antibodies exert effects through multiple pathways:

  • Disruption of PDPN-CLEC-2 interaction: Blocks platelet aggregation and tumor cell extravasation, reducing metastasis (e.g., NZ-1, MS-1 antibodies) .

  • Antibody-dependent cellular cytotoxicity (ADCC): Humanized antibodies like humLpMab-23-f induce NK cell-mediated tumor cell lysis .

  • Complement-dependent cytotoxicity (CDC): Activates the complement system to lyse PDPN-positive cells .

  • Inhibition of immunosuppressive signaling: Targets PDPN-positive CAFs to reverse TGF-β-mediated immunosuppression .

Diagnostic Applications

PDPN antibodies are widely used in immunohistochemistry (IHC) to detect lymphatic invasion and tumor margins .

AntibodyCloneApplicationSpecificity
D2-40Mouse IgG3Lymphatic endothelial tumor diagnosisPDPN on lymphatic endothelial cells
NZ-1Rat IgG2aGlioma, mesothelioma, lung cancerPLAG domain of PDPN
5B3Mouse IgG2bIHC and ELISA for tumor detectionRecombinant PDPN fragment
PMab-117Mouse IgG2aCancer-specific PDPN detectionTumor cells, not normal podocytes

Preclinical Findings

  • NZ-8 and NZ-12: Reduced tumor growth in xenograft models of mesothelioma and lung cancer via ADCC/CDC .

  • PMab-117-mG2a: Suppressed PDPN-positive glioblastoma (LN319) and lung cancer (PC-10) xenografts by 48–64% .

  • Anti-PDPN + anti-CTLA-4: Synergistically enhanced NK cell infiltration and antitumor immunity in mesothelioma models .

Clinical Trials

  • Phase I/II in canines: Anti-PDPN antibody P38Bf showed stable disease in malignant melanoma with grade 1/2 adverse effects (e.g., vomiting, elevated CRP) .

Research Advancements

StudyKey FindingReference
Humanized antibodiesDefucosylated humLpMab-23-f enhanced ADCC against glioblastoma and lung cancer
Cancer-specific mAbsPMab-117-mG2a avoided reactivity with normal kidney/lung cells, reducing toxicity
Combination therapyAnti-PDPN + anti-CTLA-4 increased NK cell infiltration and tumor necrosis

Challenges and Future Directions

  • Specificity: Non-cancer-specific antibodies (e.g., NZ-1) bind normal tissues, necessitating CasMabs like PMab-117 .

  • Combination therapies: Pairing PDPN antibodies with immune checkpoint inhibitors (e.g., anti-CTLA-4) improves efficacy .

  • Clinical translation: Human trials are pending despite success in canine and xenograft models .

Product Specs

Introduction
Podoplanin, a small mucin-like type-1 transmembrane protein, is expressed in various specialized cell types. As a type-I integral membrane glycoprotein, it's found in diverse human tissues. Its mucin-type character contributes to its physiological function. In other species, the homologous protein acts as a differentiation antigen and influenza-virus receptor. During mouse development, PDPN is expressed in lymphatic progenitor cells and later in lymphatic endothelial cells, making it a specific marker for these cells. Overexpressing podoplanin significantly increases endothelial cell adhesion, migration, and tube formation, while inhibiting it decreases cell adhesion in human dermal lymphatic endothelial cells. Therefore, it serves as a specific marker for lymphatic endothelium in histopathology. Notably, podoplanin expression is elevated in most human colon, rectum, and small intestine tumors. AGGRUS, potentially, can differentiate seminomas (which generally overexpress the protein) from embryonal carcinoma in testicular germ cell tumors.
Physical Appearance
The product is a clear and colorless solution that has been sterilized by filtration.
Formulation
The antibody is supplied at a concentration of 1mg/ml in a solution of PBS at pH 7.4 with 0.1% sodium azide.
Storage Procedures
For short-term storage (up to 1 month), the antibody can be stored at 4°C. For extended storage, it is recommended to store the antibody at -20°C. Repeated freezing and thawing of the antibody should be avoided.
Stability / Shelf Life
The antibody is stable for 12 months when stored at -20°C and for 1 month when stored at 4°C.
Applications
The PDPN antibody has undergone testing using both ELISA and Western blot analysis to confirm its specificity and reactivity. However, as specific experimental conditions may vary, it is recommended to titrate the antibody for each application to determine the optimal working concentration. For Western blot analysis, a dilution range of 1:500 to 1:1000 is suggested, with an initial dilution of 1:500 being a suitable starting point.
Synonyms
Podoplanin, Glycoprotein 36, PA2.26 antigen, T1A, GP36, GP40, Gp38, OTS8, T1A2, HT1A-1, PA2.26, T1-alpha, PDPN.
Purification Method
PDPN antibody was purified from mouse ascitic fluids by protein-G affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
P5E2AT.
Immunogen
Anti-human PDPN mAb is derived from hybridization of mouse FO myeloma cells with spleen cells from BALB/c mice immunized with recombinant human PDPN amino acids 1-206 purified from E. coli.
Ig Subclass
Mouse IgG2b heavy chain and κ light chain.

Q&A

What is PDPN and why is it a significant target for antibody development?

Podoplanin (PDPN) is a 38 kDa O-linked transmembrane sialoglycoprotein consisting of 162 amino acids that has emerged as an important target for antibody development . PDPN exhibits a specific expression pattern that makes it valuable for both diagnostic and therapeutic applications. It is highly expressed in lymphatic endothelial cells but not in vascular endothelial cells, making it a specific marker for lymphatic vessels .

Moreover, PDPN overexpression is associated with poor clinical outcomes in various tumors and plays a role in malignant tumor progression by promoting invasiveness and metastasis . This dual characteristic—specific expression in lymphatic vessels and upregulation in aggressive tumors—makes PDPN an attractive target both for diagnostic applications in pathology and for therapeutic intervention through monoclonal antibody development.

How is PDPN expression distributed in normal tissues versus tumor tissues?

PDPN shows a distinct distribution pattern between normal and tumor tissues, which has significant implications for antibody development:

Normal Tissue Expression:

  • Lymphatic endothelial cells (primary expression site)

  • Follicular dendritic cells

  • Reticular cells

  • Mesothelial cells

  • Testicular germ cells

  • Ovarian cells

  • Kidney podocytes

Tumor Tissue Expression:

  • Lymphangiomas

  • Kaposi sarcomas

  • Seminomas

  • Epithelioid mesotheliomas

  • Hemangioblastomas

  • Various carcinomas with lymphatic invasion

This differential expression pattern enables the diagnosis of lymphatic endothelial cell-derived tumors and assessment of lymphatic invasion and metastasis in other tumor tissues . The expression profile has guided the development of cancer-specific antibodies (CasMabs) like PMab-117, which selectively target PDPN in tumor tissues while showing minimal reactivity with normal PDPN-expressing cells, such as kidney podocytes and normal epithelial cells .

What are the key domains of PDPN that serve as targets for antibody development?

The platelet aggregation-stimulating (PLAG) domains of PDPN represent primary targets for antibody development, each with distinct characteristics:

PLAG Domain Structure and Targeting:

  • PLAG3 domain: Targeted by antibodies such as MS-1 mAb, which recognizes both monkey and human podoplanin PLAG3 domains

  • PLAG4 domain: Targeted by antibodies like PG4D2, which specifically recognizes the perimeter structure from Arg 79 to Leu 83 (79-RIEDL-83) in human podoplanin

Species Variations in PLAG Domains:

  • Human PLAG4 domain contains Arg 79

  • Cynomolgus monkey PLAG4 domain has His 79 instead of Arg 79, affecting cross-reactivity of some antibodies

Researchers have employed various strategies to target these domains effectively:

  • Expression of the extracellular part of PDPN (ePDPN) as fusion proteins

  • Creation of tandemly connected repeats of specific PDPN domains (e.g., amino acids 76-89 connected 21 times)

  • Generation of domain-specific deletion mutants (ΔPLAG3, ΔPLAG4) to map epitope specificity

Understanding these structural domains is crucial for developing antibodies with specific binding properties and desired functional characteristics for both research and clinical applications.

What methodologies are employed for anti-PDPN monoclonal antibody production?

Several methodologies have been developed for producing anti-PDPN monoclonal antibodies, each with distinct advantages for different research applications:

A. Recombinant Protein Expression in E. coli:

  • Synthesis of genes encoding the extracellular part of PDPN (ePDPN)

  • Expression of fusion proteins (ePDPN-His and GST-ePDPN)

  • Purification of fusion proteins for immunization

B. Cell-Based Immunization:

  • Immunization with PDPN-overexpressed glioblastoma cells (e.g., LN229/PDPN)

  • Selection of hybridomas based on reactivity patterns to PDPN-positive versus negative cell lines

C. Domain-Specific Peptide Approach:

  • Creation of tandemly connected repeats of specific PDPN domains

  • Expression as GST-tagged peptides

  • Purification for use as immunogens

D. Hybridoma Technology Workflow:

  • Immunization of mice or rats with the selected PDPN antigen

  • Cell fusion procedure to generate hybridomas

  • Screening of positive clones by ELISA with recombinant PDPN

  • Subcloning by limiting dilution (typically three rounds)

  • Antibody purification from ascites using:

    • Ammonium sulfate precipitation

    • Protein G affinity chromatography

This methodological diversity enables researchers to develop antibodies with specific binding characteristics and functional properties tailored to different experimental requirements and potential clinical applications.

How can researchers evaluate the specificity of anti-PDPN antibodies?

Evaluating antibody specificity is crucial for ensuring reliable results in both diagnostic and therapeutic applications. Based on the research literature, several complementary approaches are recommended:

A. Flow Cytometry Analysis:

  • Compare reactivity against PDPN-positive cell lines (e.g., PC-10, LN319) versus PDPN-negative or knockout lines

  • Assess binding to normal cells expressing PDPN (e.g., kidney podocytes) versus tumor cells

  • Determine binding affinity (KD) through titration experiments

B. ELISA Testing:

  • Screen hybridoma supernatants using recombinant PDPN extracellular domain

  • Compare binding to wild-type versus mutated PDPN constructs

C. Western Blot Evaluation:

  • Test recognition of denatured PDPN protein in cell lysates

  • Compare signal intensity between PDPN-positive and negative samples

D. Immunohistochemistry Assessment:

  • Evaluate staining patterns in tissue sections

  • Compare with established antibodies like D2-40

  • Analyze differential staining between tumor and normal tissues

E. Functional Inhibition Assays:

  • Test ability to inhibit PDPN-induced platelet aggregation

  • Evaluate blocking of PDPN interaction with its receptor CLEC-2

F. Domain Mapping:

  • Test binding to cells expressing PDPN mutants with specific domain deletions (ΔPLAG3, ΔPLAG4)

  • Identify critical amino acid residues for antibody recognition

These methodologies collectively provide a comprehensive assessment of antibody specificity and are essential for characterizing novel anti-PDPN antibodies before application in research or clinical settings.

What functional properties distinguish cancer-specific from non-cancer-specific anti-PDPN antibodies?

Cancer-specific monoclonal antibodies (CasMabs) against PDPN exhibit distinctive functional properties compared to non-cancer-specific antibodies, with important implications for therapeutic development:

Differential Binding Properties:

PropertyCancer-Specific Antibodies (e.g., PMab-117)Non-Cancer-Specific Antibodies (e.g., NZ-1)
Tumor cell reactivityHighHigh
Normal cell reactivityLow/negligibleHigh
Binding affinity (KD)Lower (1.9 × 10⁻⁷ M)Higher (5.0 × 10⁻⁹ M)
Recognition patternCancer-specific modificationsCore PDPN structure
Safety profileReduced off-target effectsPotential normal tissue reactivity

Cancer-specific antibodies like PMab-117 selectively bind to PDPN on cancer cells while showing minimal reactivity with PDPN expressed in normal cells such as kidney podocytes and normal epithelial cells from various tissues . This selectivity is likely due to recognition of cancer-specific modifications (glycosylation patterns or conformational epitopes) present in tumor-associated PDPN but absent in normal tissues.

In contrast, non-cancer-specific antibodies like NZ-1 exhibit high reactivity to PDPN in both tumor and normal cells . While this universal binding may be advantageous for certain diagnostic applications, it presents potential safety concerns for therapeutic use due to possible adverse effects on normal PDPN-expressing tissues.

The cancer-specific binding profile makes CasMabs promising candidates for targeted cancer therapy with potentially reduced off-target effects, particularly important since PDPN plays essential roles in normal cells like kidney podocytes .

How can the cross-reactivity of anti-PDPN antibodies between human and non-human primates be evaluated?

Evaluating cross-reactivity between human and non-human primate PDPN is essential for preclinical safety studies and translational research. The following methodological approaches provide a systematic framework:

A. Sequence and Structural Analysis:

  • Compare amino acid sequences of PDPN between species, focusing on epitope regions

  • Identify key substitutions that might affect antibody binding

  • Example: PLAG4 domain in human has Arg 79, while cynomolgus monkey has His 79

B. Cell-Based Binding Assays:

  • Generate stable cell lines expressing human or monkey PDPN (e.g., CHO/hPDPN, CHO/mkyPDPN)

  • Perform flow cytometry to quantitatively compare antibody binding

  • Example finding: PG4D2 shows significantly lower reactivity to monkey PDPN compared to human PDPN

C. Functional Inhibition Studies:

  • Assess the ability of antibodies to inhibit PDPN-CLEC-2 interaction across species

  • Evaluate inhibition of platelet aggregation induced by human versus monkey PDPN

  • Example result: PG4D2 suppresses monkey PDPN-induced platelet aggregation but with lower efficacy than for human PDPN

D. Domain-Specific Mutant Analysis:

  • Generate PLAG domain deletion mutants for both human and monkey PDPN:

    • Human: hPDPN-ΔPLAG3, hPDPN-ΔPLAG4

    • Monkey: mkyPDPN-ΔPLAG3, mkyPDPN-ΔPLAG4, mkyPDPN-ΔPLAG3+4

  • Compare antibody reactivity against these mutants to map epitope conservation

E. Tissue Cross-Reactivity Studies:

  • Test antibody binding to tissue sections from humans and non-human primates

  • Compare staining patterns and intensity in corresponding tissues

This comprehensive approach enables researchers to predict the translatability of anti-PDPN antibodies from preclinical models to clinical applications and helps identify potential safety concerns before human studies.

What methods are used to assess the therapeutic potential of anti-PDPN antibodies?

Assessing the therapeutic potential of anti-PDPN antibodies requires multiple complementary approaches to evaluate both efficacy and safety. The following methodologies provide a framework for comprehensive evaluation:

A. Antibody-Dependent Cellular Cytotoxicity (ADCC) Assays:

  • Co-culture PDPN-positive tumor cells with effector cells (e.g., splenocytes)

  • Treat with anti-PDPN antibodies at various concentrations

  • Measure target cell lysis to quantify ADCC activity

  • Example findings for PMab-117-mG2a:

    • 17.3% cytotoxicity against LN229/PDPN cells

    • 42.1% cytotoxicity against PC-10 cells

    • 23.9% cytotoxicity against LN319 cells

B. Xenograft Model Studies:

  • Inoculate immunodeficient mice with PDPN-positive human tumor cells

  • Administer anti-PDPN antibodies (e.g., intraperitoneal injection on days 1, 8, 16)

  • Monitor tumor volume throughout the study

  • Measure final tumor weight at endpoint

  • Assess body weight to evaluate potential toxicity

Tumor Growth Inhibition Results with PMab-117-mG2a:

Xenograft ModelTumor Weight ReductionStatistical Significance
LN229/PDPN64%p < 0.01
PC-1055%p < 0.01
LN31948%p < 0.01

No significant body weight loss was observed in treated animals, suggesting minimal toxicity .

C. Binding Kinetics Analysis:

  • Determine antibody affinity constants (KD) by flow cytometry

  • Evaluate on-rate and off-rate kinetics

  • Correlate binding properties with therapeutic efficacy

D. Mechanism of Action Studies:

  • Investigate effects on tumor cell invasion and migration

  • Assess impact on PDPN-mediated signaling pathways

  • Evaluate combination approaches with other therapeutic agents

These methodologies collectively provide a comprehensive assessment of both the therapeutic potential and safety profile of anti-PDPN antibodies, guiding their development toward clinical applications.

How do different isotypes of anti-PDPN antibodies compare in their functional properties?

Different antibody isotypes demonstrate distinct functional properties that significantly impact their potential applications in both research and therapy:

Isotype Comparison Table:

PropertyIgM (e.g., PMab-117)IgG2a (e.g., PMab-117-mG2a, NZ-1)Other IgG Subclasses
Size~900 kDa (pentamer)~150 kDa~150 kDa
ADCC activityLowHighVariable (IgG1>IgG4)
Complement activationHighModerateVariable (IgG3>IgG1>IgG2>IgG4)
Tissue penetrationLimitedGoodGood
Half-lifeShortLongVariable
Example in studiesOriginal PMab-117Converted PMab-117-mG2a, NZ-1-

Key Functional Differences:

  • Effector Function Enhancement Through Isotype Conversion:

    • PMab-117 was initially isolated as a rat IgM antibody

    • When converted to mouse IgG2a (PMab-117-mG2a), it gained enhanced ADCC activity

    • The isotype conversion maintained cancer specificity while improving effector functions

  • Species-Dependent Considerations:

    • Rat-derived antibodies (PMab-117, NZ-1) are commonly used in preclinical research

    • Mouse IgG2a versions (PMab-117-mG2a) generated for enhanced ADCC in mouse models

    • The species origin affects interaction with Fc receptors on effector cells

  • Application-Specific Isotype Selection:

    • For diagnostic applications (IHC): binding specificity is prioritized over effector functions

    • For therapeutic applications: isotypes with strong ADCC/CDC activity (IgG1, IgG2a) are preferred

    • For studying PDPN biology without immune activation: IgG4 or Fab fragments may be optimal

These isotype-dependent properties significantly influence the behavior of anti-PDPN antibodies in different experimental contexts and must be carefully considered when selecting antibodies for specific applications in research or therapy development.

What challenges exist in developing anti-PDPN antibodies for immunohistochemistry applications?

Developing effective anti-PDPN antibodies for immunohistochemistry (IHC) presents several technical challenges that researchers must address:

A. Antigen Source Limitations:

  • Traditional methods used dysgerminoma tissue as an antigen source (e.g., for D2-40 antibody)

  • This approach faces challenges including:

    • Weak immune response due to low concentration of PDPN protein

    • Generation of non-specific antibodies due to complex tissue composition

    • Limited availability of suitable carcinoma in situ or cancer cell lines

B. Expression System Considerations:

  • Establishing stable eukaryotic cell lines expressing PDPN is challenging due to:

    • Risk of contamination during extended cell culture

    • Variability in expression levels

    • Potential alterations in post-translational modifications

C. Specificity and Sensitivity Balance:

  • Antibodies must distinguish PDPN from structurally similar proteins

  • They must recognize PDPN across different fixation and tissue preparation methods

  • The balance between detecting all PDPN-positive cells (sensitivity) while avoiding false positives (specificity) is critical

D. Epitope Preservation Issues:

  • Formalin fixation can alter protein structure and mask epitopes

  • Heat-induced epitope retrieval methods may affect antibody recognition

  • Different epitopes show variable sensitivity to processing methods

E. Reproducibility Concerns:

  • Batch-to-batch variation in antibody production affects staining consistency

  • Differences in epitope recognition between antibody clones can lead to discrepant results

  • Standardization across laboratories remains challenging

Innovative Solutions:

  • Expressing PDPN antigen in E. coli as a rapid, cost-effective method

  • Producing well-characterized fusion proteins (e.g., GST-ePDPN) for immunization

  • Implementing rigorous screening with multiple cell lines and tissue types

  • Comprehensive epitope mapping to select antibodies recognizing preserved regions

These challenges highlight the need for systematic approach to antibody development and validation for IHC applications in both research and clinical diagnostics.

How can researchers optimize anti-PDPN antibodies for therapeutic applications?

Optimizing anti-PDPN antibodies for therapeutic applications involves several strategic approaches to enhance their efficacy and safety profiles:

A. Engineering Cancer Specificity:

  • Develop cancer-specific antibodies (CasMabs) that selectively target tumor-associated PDPN

  • Screen for clones with minimal reactivity to normal PDPN-expressing tissues

  • Examples include PMab-117, LpMab-2, and LpMab-23, selected for cancer-specific binding

B. Isotype Optimization for Enhanced Effector Functions:

  • Convert promising antibodies to isotypes with optimal effector functions

  • Example: PMab-117 (IgM) → PMab-117-mG2a (mouse IgG2a) enhanced ADCC activity

  • Select isotypes based on the desired mechanism of action (ADCC, CDC, blocking)

C. Epitope Selection Strategy:

  • Target functional domains of PDPN critical for tumor progression

  • Focus on epitopes with cancer-specific modifications or accessibility

  • Consider targeting PLAG domains involved in platelet aggregation and metastasis

D. Functional Screening Cascade:

Screening LevelMethodsCriteria
Initial screeningFlow cytometryBinding to tumor cells but not normal cells
Secondary screeningADCC assays≥20% cytotoxicity at therapeutic concentrations
Tertiary screeningXenograft models≥40% tumor growth inhibition
Safety assessmentCross-reactivity studiesMinimal binding to vital normal tissues

E. Antibody Format Diversification:

  • Develop alternative formats beyond conventional antibodies:

    • Bispecific antibodies targeting PDPN and immune effector cells

    • Antibody-drug conjugates for targeted delivery of cytotoxic agents

    • Single-chain variable fragments for improved tumor penetration

F. Preclinical Validation Strategy:

  • Test in multiple xenograft models with varying PDPN expression levels

  • Evaluate in models that recapitulate the tumor microenvironment

  • Conduct comprehensive toxicity studies in relevant species

These optimization strategies provide a framework for developing anti-PDPN antibodies with improved therapeutic potential while minimizing risks, potentially leading to effective clinical applications in cancer treatment.

What experimental controls are essential when evaluating anti-PDPN antibodies?

Proper experimental controls are critical for the rigorous evaluation of anti-PDPN antibodies. The following control system ensures reliable and interpretable results:

A. Cell Line Control Panel:

Control TypeExamplesPurpose
PDPN-positive cellsPC-10, LN319, LN229/PDPNPositive control for antibody binding
PDPN-negative cellsLN229Negative control to assess background
PDPN-knockout cellsPDPN-knockout LN319 (BINDS-55)Control for antibody specificity
Normal PDPN-expressing cells293FT, PODO/TERT256Assess binding to physiological PDPN

These cell controls help confirm antibody specificity for the target protein .

B. Domain-Specific Controls:

  • Cells expressing PDPN mutants with specific domain deletions:

    • ΔPLAG3 (deletion of PLAG3 domain)

    • ΔPLAG4 (deletion of PLAG4 domain)

    • ΔPLAG3+4 (deletion of both domains)

  • These controls help map the epitope specificity of antibodies

C. Antibody Controls:

  • Isotype-matched control antibodies (e.g., control mouse IgG2a for PMab-117-mG2a)

  • Well-characterized anti-PDPN antibodies (e.g., NZ-1, D2-40) as benchmark controls

  • Pre-immune serum controls for polyclonal antibody evaluation

D. Functional Assay Controls:

  • For ADCC assays:

    • Target cells without effector cells (spontaneous lysis control)

    • Effector cells without antibodies (background control)

    • Positive control antibody with known ADCC activity

  • For xenograft studies:

    • Vehicle control groups with matched conditions

    • Isotype control antibody treatment groups

E. Cross-Species Controls:

  • Cells expressing PDPN from different species (human vs. monkey)

  • Tissue sections from different species

  • These controls help assess cross-reactivity for preclinical translation

F. Technical Controls for IHC:

  • Antigen retrieval controls (with and without treatment)

  • Absorption controls (pre-incubation of antibody with recombinant PDPN)

  • Secondary antibody-only controls to assess non-specific binding

Implementing this comprehensive control system ensures that experimental results from anti-PDPN antibody evaluation are specific, reproducible, and biologically relevant, establishing a solid foundation for both research applications and clinical development.

Product Science Overview

Introduction

Podoplanin is a type I transmembrane glycoprotein that is widely expressed in various tissues and cell types. It is known by several other names, including gp36, Aggrus, PA2.26 antigen, and T1 alpha. Podoplanin plays a crucial role in the development of the lymphatic system, cell migration, and tumor progression. The mouse anti-human podoplanin antibody is a monoclonal antibody that specifically targets human podoplanin, making it a valuable tool in research and diagnostic applications.

Structure and Function of Podoplanin

Podoplanin is a 140 amino acid protein with a molecular weight of approximately 36 kDa. It is characterized by its single-pass transmembrane domain and an extracellular domain that is heavily glycosylated. The extracellular domain contains three platelet aggregation-stimulating (PLAG) domains, which are responsible for its interaction with the C-type lectin-like receptor 2 (CLEC-2) on platelets . This interaction induces platelet aggregation and is crucial for maintaining the integrity of the lymphatic system.

Podoplanin is involved in various biological processes, including:

  • Lymphangiogenesis: Podoplanin is strongly expressed on lymphatic endothelial cells and is essential for the formation and maintenance of the lymphatic vasculature .
  • Cell Migration and Invasion: Podoplanin promotes cell migration and invasion by reorganizing the actin cytoskeleton and enhancing cellular adhesion .
  • Tumor Progression: Podoplanin expression is upregulated in several types of tumors, where it contributes to tumor cell migration, invasion, and metastasis .
Mouse Anti-Human Podoplanin Antibody

The mouse anti-human podoplanin antibody is a monoclonal antibody that specifically binds to human podoplanin. It is produced by immunizing mice with the human podoplanin ectodomain expressed in Escherichia coli. The antibody is then purified from the mouse serum using affinity chromatography.

Applications

The mouse anti-human podoplanin antibody is widely used in various research and diagnostic applications, including:

  • Immunohistochemistry (IHC): This antibody is used to detect podoplanin expression in tissue sections, allowing researchers to study its distribution and localization in different tissues .
  • Western Blotting (WB): The antibody is used to detect podoplanin in protein extracts from cells and tissues, providing information about its expression levels .
  • Flow Cytometry (FC): The antibody is used to analyze podoplanin expression on the surface of cells, enabling the identification and characterization of podoplanin-expressing cell populations .
  • Enzyme-Linked Immunosorbent Assay (ELISA): The antibody is used in ELISA to quantify podoplanin levels in biological samples .
Specificity and Validation

The mouse anti-human podoplanin antibody has been extensively validated for its specificity and performance. It has been shown to specifically bind to human podoplanin without cross-reacting with other proteins. The antibody’s specificity has been confirmed through various techniques, including relative expression analysis and knockdown experiments .

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.