Podoplanin (PDPN) Human, also known as Aggrus or T1-alpha, is a recombinant protein produced in Escherichia coli (E. coli) that corresponds to a 130-amino acid fragment of the human podoplanin protein . PDPN is a mucin-type transmembrane glycoprotein widely expressed in lymphatic endothelial cells, alveolar type I epithelial cells, and renal podocytes . It serves as a critical marker for lymphatic vessel endothelium and plays roles in development, immune regulation, and cancer progression .
PDPN mediates diverse physiological and pathological processes:
Lymphatic System: Critical for blood-lymphatic vessel separation during embryogenesis via CLEC-2 interaction .
Lung Development: Required for alveolar type I cell maturation .
Facilitates dendritic cell (DC) migration to lymph nodes by interacting with CLEC-2 on DCs .
Expressed on inflammatory macrophages, promoting platelet aggregation and fungal defense .
Metastasis: Overexpression in tumors (e.g., gliomas, squamous cell carcinomas) enhances motility and invasion via RhoA/ROCK signaling .
Diagnostic Marker: Distinguishes seminomas from embryonal carcinomas .
Antibody-Drug Conjugates (ADCs): Targeting PDPN-expressing tumors .
CAR-T Therapy: Engineered T cells with PDPN-specific chimeric antigen receptors .
Liquid Biopsy: Detecting PDPN in circulating tumor cells for early cancer diagnosis .
PDPN knockdown reduces glioma cell proliferation and invasion by 40–60% .
PDPN-CLEC-2 interaction stabilizes lymphatic vessels, preventing blood mixing .
The recombinant PDPN Human (PRO-626) is formulated in 20 mM Tris-HCl (pH 7.5) and 0.1 M NaCl, with stability maintained at 4°C for 2–4 weeks or -20°C for long-term storage . Key notes:
Recombinant Human PDPN, produced in E.Coli, is a single, non-glycosylated polypeptide chain. It consists of 130 amino acids (99-207 a.a), resulting in a molecular mass of 13.4kDa. The PDPN is fused to a 21 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
MGSSHHHHHH SSGLVPRGSH MASTGQPEDD TETTGLEGGV AMPGAEDDVV TPGTSEDRYK SGLTTLVATS VNSVTGIRIE DLPTSESTVH AQEQSPSATA SNVATSHSTE KVDGDTQTTV EKDGLSTVTL.
PDPN is a mucin-type transmembrane sialoglycoprotein with a molecular weight of approximately 36-43 kDa that is encoded by the PDPN gene in humans . Structurally, it is a type-I integral membrane, heavily O-glycosylated glycoprotein with diverse distribution in human tissues . The protein is relatively well conserved between species, with homologues present in humans, mice, rats, dogs, and hamsters .
To study PDPN's structure:
Recombinant protein expression systems can be utilized to produce the extracellular domain (ePDPN) for structural analysis
X-ray crystallography and NMR spectroscopy provide detailed structural information
Glycosylation patterns can be analyzed through mass spectrometry and glycoprotein-specific staining techniques
While the specific functions of PDPN are still being elucidated, several important roles have been identified:
PDPN plays a critical role in the proper formation of linkages between the cardiovascular and lymphatic systems during development
It mediates effects on cell migration and adhesion through interaction with various binding partners
During development, it facilitates blood and lymphatic vessel separation by binding to CLEC1B, triggering platelet activation and/or aggregation
In lymph nodes, PDPN controls fibroblastic reticular cells (FRCs) adhesion to the extracellular matrix and regulates actomyosin contraction
In neural tissue, PDPN is involved in processes including development and angiogenesis
For experimental investigation of these functions, gene knockout/knockdown models, co-culture systems with PDPN-expressing and PDPN-deficient cells, and interaction assays with identified binding partners are commonly employed.
PDPN shows a diverse distribution pattern across human tissues. Current expression data indicates:
High expression in lymphatic endothelial cells (serving as a specific lymphatic marker)
Expression in kidney podocytes (hence the name "podoplanin")
Found in other cell types including follicular dendritic cells, reticular cells, mesothelial cells, testicular germ cells, and ovarian cells
Methodologically, tissue expression patterns can be analyzed through:
Immunohistochemistry using specific anti-PDPN antibodies like D2-40 or the 5B3 monoclonal antibody
RNA sequencing data from databases like The Human Protein Atlas
Single-cell RNA sequencing for cell-type specific expression profiling
In situ hybridization for localization of PDPN mRNA
Several validated methods for PDPN detection include:
Immunohistochemistry (IHC): Using monoclonal antibodies such as D2-40 or 5B3 for tissue sections
Flow cytometry: For detecting PDPN on live cells, with antibodies like humLpMab-23 demonstrating high affinity (KD values between 4.7-5.4 × 10−9 M)
Western blotting: For protein detection in cell or tissue lysates
Immunofluorescence microscopy: For co-localization studies, particularly useful as PDPN has been shown to co-localize with nestin
When selecting detection methods, researchers should consider that PDPN is heavily glycosylated, which may affect antibody binding efficiency. The 5B3 monoclonal antibody has demonstrated excellent specificity in ELISA, western blot, and immunohistochemistry experiments, with an affinity constant of 2.94 × 108 L/mol .
PDPN interacts with several proteins to mediate its diverse functions:
CLEC-2 (C-type lectin 2): Expressed on platelets and hematopoietic cells, this interaction is crucial for blood/lymphatic vessel separation during embryonic development
CD9: Interaction attenuates platelet aggregation induced by PDPN
ERM proteins (Ezrin, Radixin, Moesin): Promotes epithelial-mesenchymal transition (EMT) by triggering RHOA activation
CD44: Promotes directional cell migration in epithelial and tumor cells
LGALS8 (Galectin-8): May participate in connecting lymphatic endothelium to surrounding extracellular matrix
To study these interactions experimentally:
Co-immunoprecipitation assays
Proximity ligation assays
FRET/BRET analysis for real-time interaction monitoring
Yeast two-hybrid screening to identify novel binding partners
Surface plasmon resonance for binding kinetics analysis
PDPN has been extensively studied in cancer research with several mechanisms identified:
PDPN is often upregulated in various cancers including squamous cell carcinomas, malignant mesothelioma, and brain tumors
It serves as a specific lymphatic vessel marker, with lymphangiogenesis levels correlating with poor prognosis in cancer patients
In cancer-associated fibroblasts (CAFs), PDPN upregulation has been associated with poor prognosis
In squamous cell carcinomas, PDPN plays a key role in cancer cell invasiveness by controlling invadopodia formation and promoting efficient extracellular matrix (ECM) degradation
It modulates RHOC activity to activate ROCK1/ROCK2 and LIMK1/LIMK2 pathways while inactivating CFL1, leading to invadopodia stability and maturation
Methodological approaches to study these mechanisms include:
3D invasion assays with PDPN-expressing versus knockdown cells
ECM degradation assays using fluorescently labeled matrix proteins
In vivo metastasis models with PDPN-manipulated cancer cells
Analysis of patient samples for correlation between PDPN expression patterns and clinical outcomes
Co-culture models of cancer cells with PDPN-positive CAFs
Research has revealed complex interactions between PDPN-expressing fibroblasts and tumor progression:
Podoplanin-positive cancer-associated fibroblasts (CAFs) positively correlate with tumor size, grade of malignancy, lymph node metastasis, lymphovascular invasion, and poor patient outcomes in breast cancer
Ectopic expression of podoplanin significantly increases the migration capabilities of fibroblasts (as demonstrated in MSU1.1 and Hs 578Bst fibroblast cell lines)
PDPN expression in fibroblasts affects the formation of pseudo tubes by endothelial cells, with podoplanin-rich fibroblasts resulting in endothelial cell capillary-like networks characterized by significantly lower numbers of nodes and meshes
Rather than directly affecting cancer cell migration and invasion, PDPN may facilitate fibroblast movement into tumor stroma, creating a favorable microenvironment for tumor progression by increasing CAF numbers
Experimental approaches to investigate these interactions include:
Co-culture systems with PDPN-expressing fibroblasts and cancer cells
3D organoid models incorporating multiple cell types
Conditional knockout models targeting PDPN specifically in fibroblasts
In vitro tube formation assays with endothelial cells and PDPN-manipulated fibroblasts
RNA-seq analysis of PDPN-positive versus negative fibroblasts to identify secreted factors
Given PDPN's important role as a lymphatic marker, several specialized approaches are used:
Immunohistochemical analysis using anti-PDPN antibodies for lymphatic vessel density quantification in tissues
3D lymphatic vessel formation assays using lymphatic endothelial cells with PDPN manipulation
PDPN knockout/knockdown studies in lymphatic endothelial cells to assess functional impacts
In vivo lymphangiogenesis models with fluorescent labeling of lymphatic vessels
Co-culture systems to study interactions between PDPN-expressing cells and lymphatic endothelium
Researchers should be aware that PDPN's diagnostic utility stems from its specificity as a lymphatic vessel marker, with lymphangiogenesis levels correlating with poor prognosis in cancer patients . For accurate assessment, multiple PDPN-positive vessel quantification methods should be employed, preferably using automated image analysis systems to reduce observer bias.
Development of anti-PDPN antibodies follows several strategies:
Generation of recombinant extracellular PDPN (ePDPN) fusion proteins in expression systems like E. coli, as demonstrated with ePDPN-His and GST-ePDPN fusion proteins
Immunization protocols using purified fusion proteins with adjuvants like QuickAntibody-Mouse5W
Hybridoma technology for monoclonal antibody production, as exemplified by the 5B3 cell line generating anti-PDPN mAb
Humanization of mouse antibodies by fusing the variable domain CDRs with human immunoglobulin constant domains, as shown with humLpMab-23
Engineering of defucosylated antibodies (like humLpMab-23-f) to enhance antibody-dependent cellular cytotoxicity (ADCC)
These antibodies have multiple research applications:
Diagnostic immunohistochemistry for identifying lymphatic vessels and PDPN-expressing tumors
Flow cytometry for cell sorting and quantification of PDPN-positive populations
Therapeutic targeting of PDPN-expressing cancer cells
Blocking experiments to study PDPN's functional roles
In vivo imaging of PDPN-expressing tissues
PDPN's involvement in neurological diseases is an emerging research area:
PDPN is involved in several physiological and pathological processes in the brain, including development and angiogenesis
Neurological disorders constitute a major cause of disability and death worldwide (16.8% of total deaths), with a 36% increase in associated deaths over the past 25 years
Vasculopathy, inflammation, and immune abnormalities play important roles in neurological diseases, with PDPN potentially involved in these processes
PDPN expression has been detected in neural tissue in both mouse and human samples
Methodological approaches for investigating PDPN in neurological contexts include:
Immunohistochemical analysis of PDPN expression in normal versus diseased neural tissue
Primary neural cell cultures with PDPN manipulation
In vitro blood-brain barrier models to study PDPN's vascular effects
Conditional knockout models targeting PDPN in specific neural cell populations
Cerebrospinal fluid analysis for PDPN as a potential biomarker
PDPN influences cell migration through several mechanisms:
In fibroblasts, ectopic expression of podoplanin significantly increases migration capability, as demonstrated in MSU1.1 and Hs 578Bst fibroblast cell lines
Through MSN or EZR (ezrin) interaction, PDPN promotes epithelial-mesenchymal transition (EMT) leading to EZR phosphorylation and triggering RHOA activation, resulting in increased cell migration and invasiveness
Interaction with CD44 promotes directional cell migration in epithelial and tumor cells
In keratinocytes, PDPN induces changes in cell morphology, including elongated shape, numerous membrane protrusions, major reorganization of the actin cytoskeleton, increased motility, and decreased cell adhesion
PDPN controls invadopodia stability and maturation through modulation of RHOC activity, activating ROCK1/ROCK2 and LIMK1/LIMK2 while inactivating CFL1
Experimental techniques to study these pathways include:
Live cell imaging with fluorescently labeled cytoskeletal components
RHOA/RHOC activity assays in PDPN-manipulated cells
Phosphorylation analysis of EZR and other downstream targets
2D and 3D migration assays with pathway inhibitors
Quantitative analysis of membrane protrusion dynamics
Several specialized techniques help elucidate PDPN's role in ECM interactions:
Invadopodia formation assays using fluorescently labeled ECM components to visualize degradation
Atomic force microscopy to measure cell-ECM adhesion forces in PDPN-expressing versus control cells
Traction force microscopy to assess mechanical forces exerted by cells on the ECM
ECM remodeling assays measuring collagen contraction by PDPN-expressing fibroblasts
Co-immunoprecipitation studies to identify PDPN interactions with ECM receptors like integrins
These approaches are particularly relevant given that PDPN modulates invadopodia stability and maturation leading to efficient ECM degradation in tumor cells , and controls fibroblastic reticular cells (FRCs) adhesion to the ECM . Additionally, through binding with LGALS8 (Galectin-8), PDPN may participate in connecting lymphatic endothelium to the surrounding ECM .
Researchers can employ various models to investigate tissue-specific PDPN functions:
In vitro models:
Primary cell cultures from different PDPN-expressing tissues (lymphatic endothelium, neural cells, fibroblasts)
Cell lines with endogenous or manipulated PDPN expression
3D organoid models incorporating multiple cell types
Co-culture systems (e.g., fibroblasts with endothelial or cancer cells)
In vivo models:
Global PDPN knockout mice (note: PDPN is required for normal lung cell proliferation and alveolus formation at birth)
Conditional tissue-specific PDPN knockout models
PDPN reporter mice for lineage tracing studies
Xenograft models using PDPN-manipulated human cells
Clinical samples:
Tissue microarrays from various PDPN-expressing normal and pathological tissues
Patient-derived organoids
Single-cell analysis of tissues to identify PDPN-expressing cell populations
When selecting appropriate models, researchers should consider tissue-specific PDPN functions, such as its role in lymphatic vessels, neural tissue, cancer-associated fibroblasts, and various epithelial tissues.
Podoplanin, also known as PDPN, is a small mucin-type transmembrane glycoprotein. It is highly conserved across species, including humans, mice, rats, dogs, and hamsters . Podoplanin is involved in various physiological and pathological processes, making it a significant molecule in biomedical research.
Podoplanin is a 36- to 43-kDa protein that is extensively expressed in different tissues and cells. These include lymphatic endothelial cells, type I alveolar cells, osteocytes, choroid plexus epithelial cells, glial cells, and stromal reticular cells in lymphoid organs . The protein’s structure includes a short cytoplasmic tail that interacts with proteins of the ezrin/radixin/moesin family, which function as cross-linkers between actin filaments and the plasma membrane .
Podoplanin plays a crucial role in several biological processes:
Podoplanin is widely used as a marker for lymphatic endothelial cells and fibroblastic reticular cells of lymphoid organs. It is also used to identify lymphatics in the skin and tumor microenvironment . The protein’s ability to aggregate and activate CLEC-2-expressing platelets makes it a significant player in thrombosis and inflammation .