PEDF Human, His

Pigment Epithelium-Derived Factor Human Recombinant, His Tag
Shipped with Ice Packs
In Stock

Description

Molecular Identity of PEDF Human, His

PEDF Human, His is a recombinant protein produced in Escherichia coli with a 20-amino acid histidine (His) tag fused to its N-terminus . Key characteristics include:

  • Amino acid sequence: 420 residues (positions 20–418 of native PEDF + His tag) .

  • Molecular mass: 46.7 kDa .

  • Post-translational modifications: Non-glycosylated (unlike native PEDF, which is glycosylated at Asn285) .

  • Variant: Contains the M72T natural polymorphism .

Production and Purification

PEDF Human, His is synthesized using proprietary chromatographic techniques :

ParameterDetails
Expression systemE. coli
Purification methodImmobilized metal affinity chromatography (IMAC) via His tag
Formulation20 mM Tris-HCl (pH 8.0), 0.1 M NaCl, 20% glycerol
Purity>90% (SDS-PAGE)
StabilityStable at 4°C for 2–4 weeks; long-term storage at -20°C with carrier protein

Anti-Angiogenic Activity

  • Inhibits endothelial cell proliferation by inducing apoptosis .

  • Suppresses VEGF and FGF-2 signaling, critical for pathological angiogenesis .

Neurotrophic Effects

  • Promotes neurite outgrowth in retinoblastoma cells (Y-79, Weri) .

  • Enhances expression of neuronal markers (e.g., neuron-specific enolase) .

Anti-Tumor Mechanisms

  • Prostate cancer:

    • Reduces metastasis by inhibiting stromal vasculature and inducing tumor cell apoptosis .

    • Upregulates caspase-3 activation in prostate carcinoma cells .

  • Breast cancer:

    • Overexpression decreases brain metastasis in xenograft models .

Research Applications

PEDF Human, His is utilized in:

  • In vitro assays: Binding studies with PEDF-R, its cognate receptor .

  • Preclinical models:

    • Xenograft tumor studies (e.g., prostate, breast cancer) .

    • Neuroprotection assays in retinal and neuronal cells .

Comparative Analysis with Native PEDF

FeatureNative PEDFPEDF Human, His
GlycosylationYes (Asn285) No
Molecular mass~50 kDa 46.7 kDa
Expression systemMammalian cells (natural) E. coli
Functional domainsRetained (24–57, 58–101) Retained

Clinical Implications

  • Ocular diseases: Potential therapy for age-related macular degeneration (AMD) and diabetic retinopathy .

  • Cancer: Phase I/II trials explore PEDF peptides for anti-angiogenic therapy .

  • Bone disorders: Low PEDF levels correlate with osteogenesis imperfecta type VI .

Table 1: Functional Peptide Regions in PEDF

DomainResiduesActivityReference
Neurotrophic region58–101Neuronal differentiation
Anti-angiogenic region24–57Endothelial apoptosis induction
Heparin-binding siteHelices D–FGlycosaminoglycan interaction

Table 2: Expression in Disease States

ConditionPEDF LevelsConsequenceReference
Prostate cancerDecreasedIncreased metastasis risk
MelanomaReducedTumor progression
Osteogenesis imperfectaUndetectableBone fragility

Product Specs

Introduction
Pigment epithelium-derived factor (PEDF), also known as SerpinF1, is a neurotrophic protein with anti-angiogenic properties. It is a member of the serpin family but lacks serine protease inhibitory activity. PEDF plays a crucial role in neuronal differentiation, tumor suppression, and the pathogenesis of diabetic retinopathy. It counteracts the effects of vascular endothelial growth factor (VEGF) and is implicated in various ocular diseases.
Description
Recombinant human PEDF, containing a natural M72T variant, was expressed in E. coli. This non-glycosylated protein consists of 420 amino acids (20-418 a.a.), with a 20 amino acid His Tag at the N-terminus, resulting in a molecular weight of 46.7 kDa. Purification was achieved using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The PEDF protein is supplied in a solution containing 20mM Tris-HCl buffer (pH 8.0), 0.1M NaCl, and 20% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, freeze at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity of PEDF is greater than 90% as determined by SDS-PAGE analysis.
Synonyms
Pigment epithelium-derived factor, PEDF, Serpin-F1, SerpinF1, EPC-1, EPC1, PIG35.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MQNPASPPEE GSPDPDSTGA LVEEEDPFFK VPVNKLAAAV SNFGYDLYRV RSSMSPTTNV LLSPLSVATA LSALSLGAEQ RTESIIHRAL YYDLISSPDI HGTYKELLDT VTAPQKNLKS ASRIVFEKKL RIKSSFVAPL EKSYGTRPRV LTGNPRLDLQ EINNWVQAQM KGKLARSTKE IPDEISILLL GVAHFKGQWV TKFDSRKTSL EDFYLDEERT VRVPMMSDPK AVLRYGLDSD LSCKIAQLPL TGSMSIIFFL PLKVTQNLTL IEESLTSEFI HDIDRELKTV QAVLTVPKLK LSYEGEVTKS LQEMKLQSLF DSPDFSKITG KPIKLTQVEH RAGFEWNEDG AGTTPSPGLQ PAHLTFPLDY HLNQPFIFVL RDTDTGALLF IGKILDPRGP.

Q&A

What is the molecular structure of human PEDF and how does it relate to function?

Human PEDF is a 50 kDa glycoprotein belonging to the serpin superfamily. The crystal structure of glycosylated human PEDF has been solved to 2.85 Å resolution, revealing several critical features that explain its multifunctional nature . The structure shows that PEDF possesses a striking asymmetric charge distribution that is likely functionally significant, with basic residues concentrated on helices D, E, and F, as well as on strands 1, 2, and 3 of β-sheet A . This asymmetric charge distribution creates distinct binding surfaces for various interaction partners.

The structure comprises well-ordered regions with the exception of 15 residues at the N-terminus (residues 1-15) and 8 residues in the reactive center loop (residues 353-360) . Understanding this structure provides crucial insights for designing experiments to probe specific domains and their roles in PEDF's diverse biological activities.

Which receptors mediate human PEDF signaling and how can they be studied?

PEDF interacts with multiple cell-surface receptors, including PEDF receptor (PEDFR; encoded by PNPLA2), laminin receptor, F1 ATPase/synthase, and low-density lipoprotein receptor-related protein 6 (LRP6) . These diverse receptor interactions explain the pleiotropic effects of PEDF across different tissues and cell types.

For studying these interactions, researchers should consider:

  • Receptor-specific blocking antibodies to isolate individual receptor contributions

  • siRNA knockdown approaches targeting specific receptors

  • Competitive binding assays using truncated PEDF variants

  • Proximity ligation assays to visualize PEDF-receptor interactions in situ

The solvent-accessible surface formed by helices C and D and the loop connecting them (loop 90) appears to be important for receptor binding and neurotrophic activity . When designing receptor interaction studies, focus on these regions rather than the entire peptide fragments previously used in some studies.

What are the key functional domains of human PEDF that researchers should be aware of?

Several key functional domains have been identified in human PEDF:

  • Neurotrophic domain: Located in the exposed regions of helices C and D and loop 90, contrary to earlier studies that suggested larger regions were necessary

  • Heparin-binding domain: Involves basic amino acids Lys146, Lys147, and Arg149

  • Hyaluronan-binding domain: Comprised of Lys189, Lys191, Arg194, and Lys197

  • Collagen-binding domain: Contains acidic amino acids Asp256, Asp258, and Asp300

  • Anti-angiogenic domain: While not fully mapped in the provided sources, this activity appears to be separable from the neurotrophic function

When designing experiments targeting specific PEDF functions, researchers should carefully consider which domains they wish to engage or modify, as mutations in different regions can selectively affect particular biological activities.

How does the unusual charge distribution of PEDF contribute to its biological functions?

The crystal structure of human PEDF reveals a remarkably asymmetric charge distribution that likely underlies its diverse functionality . The basic residues are concentrated on helices D, E, and F, on strands 1, 2, and 3 of β-sheet A, and the loop 170 . This creates a positively charged surface that serves as a binding site for heparin and proteoglycans, which is significantly larger than what was previously predicted through homology modeling .

This charge asymmetry likely facilitates:

  • Binding to extracellular matrix components

  • Tissue-specific localization

  • Regulation of bioavailability

  • Receptor specificity in different cellular contexts

To investigate these functions experimentally, researchers should consider:

  • Site-directed mutagenesis of key charged residues

  • Binding assays under varying ionic strength conditions

  • Molecular dynamics simulations to predict conformational changes upon binding

  • In vivo studies with charge-modified PEDF variants to assess tissue distribution

What experimental approaches can resolve conflicting data on PEDF's role in different cancer types?

To resolve conflicting data, researchers should:

  • Use multiple cancer cell lines within the same tissue type

  • Compare 2D versus 3D culture systems

  • Employ both genetic (overexpression/knockdown) and recombinant protein approaches

  • Distinguish between direct anti-tumor effects and indirect effects via angiogenesis inhibition

  • Assess dose-dependent responses across a wide concentration range

  • Investigate context-dependent effects (normoxia vs. hypoxia, inflammatory vs. non-inflammatory environments)

Of particular interest is PEDF's dual role in brain metastases from breast cancer, where it not only decreases metastatic potential but also protects neurons from tumor-induced damage . This demonstrates how PEDF can simultaneously affect both tumor cells and the surrounding microenvironment.

How can researchers differentiate between PEDF's anti-angiogenic and direct anti-tumor effects?

Distinguishing between PEDF's anti-angiogenic properties and its direct effects on tumor cells requires carefully designed experiments:

  • In vitro segregation:

    • Conduct parallel studies on endothelial cells (for anti-angiogenic effects) and tumor cells (for direct effects)

    • Use endothelial-free tumor spheroid models to isolate direct effects

    • Employ conditioned media experiments to identify secreted factors

  • In vivo approaches:

    • Use tumor models with varied vascularization dependencies

    • Combine PEDF treatment with specific angiogenesis inhibitors

    • Perform temporal studies (early vs. late intervention)

    • Measure both microvessel density and tumor cell apoptosis/proliferation

  • Molecular segregation:

    • Utilize PEDF variants with mutations in domains specific to each function

    • Apply receptor-specific blocking strategies

    • Monitor distinct downstream signaling pathways

Studies have shown that PEDF can independently exhibit antimigratory activity on breast tumor cells and neuroprotective effects on neurons, highlighting its multifunctional nature beyond angiogenesis inhibition .

What are the optimal conditions for expressing and purifying biologically active His-tagged human PEDF?

For optimal expression and purification of His-tagged human PEDF:

Expression Systems:

  • Mammalian expression systems (HEK293, CHO cells) are preferred for proper glycosylation

  • Insect cell systems (Sf9, High Five) offer a compromise between yield and post-translational modifications

  • Bacterial systems may be used for structural studies but might lack essential modifications

Purification Strategy:

  • Immobilized metal affinity chromatography (IMAC) using Ni-NTA or Co-based resins

  • Ion exchange chromatography as a secondary purification step

  • Size exclusion chromatography for final polishing

  • Consider on-column refolding if using bacterial expression systems

Critical Considerations:

  • Include protease inhibitors throughout purification to prevent degradation

  • Avoid harsh elution conditions that may affect protein structure

  • Verify glycosylation status, as human PEDF is known to be glycosylated

  • Consider including low concentrations of glycosaminoglycans in storage buffers to stabilize the protein

What methods are recommended for quantifying PEDF in biological samples?

For accurate quantification of PEDF in biological samples:

ELISA-based Methods:
Commercial ELISA kits are available for human PEDF quantification with the following specifications:

  • Sensitivity: 1.28 ng/mL

  • Detection range: 3.13-200 ng/mL

  • Sample types: serum, plasma, tissue homogenates, cell lysates, cell culture supernates, and other biological fluids

The sandwich ELISA approach involves:

  • Antibody-coated plate binds PEDF

  • Biotin-conjugated detection antibody binds to captured PEDF

  • Avidin-HRP conjugate binds to biotin

  • TMB substrate provides colorimetric readout

  • Concentration determined by comparison to standard curve

Alternative Quantification Methods:

  • Western blotting with densitometry for semi-quantitative analysis

  • Mass spectrometry for absolute quantification and isoform discrimination

  • Proximity ligation assays for in situ quantification

  • Radioimmunoassay for high sensitivity detection

When selecting a quantification method, consider sample type, expected concentration range, and whether total or active PEDF is being measured.

How can the biological activity of purified His-tagged PEDF be verified?

To verify the biological activity of purified His-tagged PEDF, researchers should employ functional assays corresponding to PEDF's known activities:

Anti-angiogenic Activity:

  • Endothelial cell tube formation assays

  • Endothelial cell migration assays

  • Chick chorioallantoic membrane (CAM) assay

  • Matrigel plug assay in mice

Neurotrophic Activity:

  • Neuronal survival assays using primary neurons or neuroblastoma cells

  • Neurite outgrowth assays

  • Protection against glutamate toxicity

  • Assessment of neuronal differentiation markers

Anti-tumor Activity:

  • Cancer cell proliferation assays

  • Apoptosis assays (caspase activation, Annexin V staining)

  • Cell migration and invasion assays

  • Colony formation assays

Receptor Binding:

  • Surface plasmon resonance with purified receptors

  • Cell-based binding assays with receptor-expressing cells

  • Competitive binding assays with known ligands

Researchers should include appropriate positive controls (commercially available PEDF) and negative controls (heat-inactivated PEDF, irrelevant proteins) in all assays.

How can researchers address inconsistent results when studying PEDF's effects on different cell types?

Inconsistent results when studying PEDF across different cell types may stem from several factors:

Potential Sources of Variation:

  • Receptor expression profiles differ between cell types

  • Concentration-dependent effects (PEDF may have opposing effects at different concentrations)

  • Context-dependent signaling (microenvironment influences)

  • Posttranslational modifications of PEDF

  • Presence of co-factors or binding partners

Methodological Solutions:

  • Characterize receptor expression in each cell type before experiments

  • Use dose-response curves rather than single concentrations

  • Standardize experimental conditions (serum levels, cell density, passage number)

  • Verify protein quality before each experiment (activity assays, analytical SEC)

  • Consider the influence of matrix components in your experimental system

PEDF exhibits cell type-specific effects that may be contradictory in different contexts. For example, while it inhibits endothelial cell proliferation, it can promote neuronal survival and differentiation . Understanding the receptor landscape and signaling pathways in each cell type is crucial for interpreting results.

What strategies should be employed when PEDF activity diminishes during storage or handling?

PEDF activity can diminish during storage or handling due to several factors:

Common Causes of Activity Loss:

  • Protein aggregation

  • Proteolytic degradation

  • Oxidation of critical residues

  • Loss of essential co-factors

  • Deglycosylation

Prevention Strategies:

  • Storage recommendations:

    • Store at -80°C in small single-use aliquots

    • Include 10-20% glycerol in storage buffer

    • Add low concentrations of carrier proteins (BSA)

    • Consider adding reducing agents if cysteine residues are present

  • Handling protocols:

    • Minimize freeze-thaw cycles

    • Keep on ice during experiments

    • Use low-binding tubes and pipette tips

    • Filter sterilize rather than heat sterilize

  • Quality control:

    • Regularly verify protein integrity by SDS-PAGE

    • Monitor activity using simple functional assays

    • Check for aggregation using dynamic light scattering

    • Verify glycosylation status periodically

The recombinant human PEDF used in structural studies was confirmed to be glycosylated , suggesting that glycosylation might be important for stability and function.

How might the asymmetric charge distribution of PEDF inform the development of targeted therapeutics?

The striking asymmetric charge distribution revealed in the crystal structure of PEDF offers significant opportunities for developing targeted therapeutics . This unique feature creates distinct binding surfaces that could be selectively targeted or mimicked:

Therapeutic Strategies Based on Charge Distribution:

  • Development of peptide mimetics that replicate the basic patch for anti-angiogenic applications

  • Design of small molecules that disrupt specific charge-based interactions

  • Creation of engineered PEDF variants with enhanced charge asymmetry for improved activity

  • Generation of antibodies targeting specific charged surfaces for diagnostic or therapeutic applications

A structure-guided approach could lead to therapeutics that selectively modulate specific PEDF functions while leaving others intact, potentially reducing side effects. The crystal structure provides the necessary foundation for such detailed structure-function analyses that could lead to novel therapeutics against uncontrolled angiogenesis .

What emerging techniques might advance our understanding of PEDF's multiple biological roles?

Several cutting-edge techniques show promise for elucidating PEDF's complex biology:

  • Cryo-electron microscopy: For visualizing PEDF-receptor complexes at near-atomic resolution

  • Single-cell transcriptomics: To identify cell type-specific responses to PEDF treatment

  • CRISPR-based genetic screens: To discover novel components of PEDF signaling pathways

  • Intravital microscopy: For real-time visualization of PEDF effects in living tissues

  • Proteomics approaches: To identify the PEDF interactome under different conditions

  • Tissue-specific conditional knockouts: To dissect the role of PEDF in specific tissues

  • Organ-on-chip technology: To study PEDF in physiologically relevant microenvironments

These advanced techniques could help resolve whether PEDF's various biological roles (neurotrophic, anti-angiogenic, antitumorigenic) involve different regions of the protein and whether they engage the same or different receptors, which remains an open question .

Product Science Overview

Introduction

Pigment Epithelium-Derived Factor (PEDF), also known as serpin F1 (SERPINF1), is a multifunctional secreted protein with significant roles in anti-angiogenic, anti-tumorigenic, and neurotrophic functions . This protein has garnered attention for its potential therapeutic applications in conditions such as choroidal neovascularization, heart disease, and cancer .

Discovery and Structure

PEDF was first discovered by Joyce Tombran-Tink and Lincoln Johnson in the late 1980s while studying human retinal cell development . The protein is encoded by the SERPINF1 gene located on chromosome 17 in humans . PEDF belongs to the serine protease inhibitors (serpin) superfamily, although it is a non-inhibitory member .

Function and Mechanism

PEDF is a potent inhibitor of angiogenesis, the process of forming new blood vessels, which is crucial in limiting tumor growth and progression . It achieves this by binding to vascular endothelial growth factor receptors (VEGFR-1 and VEGFR-2), promoting their internalization and/or degradation, thereby inhibiting endothelial cell proliferation and migration . Additionally, PEDF induces apoptosis in endothelial cells through various pathways .

Recombinant PEDF (Human, His Tag)

Recombinant PEDF is produced using human cells and is often tagged with a 6x-His tag for purification purposes . This recombinant form retains the biological activity of the native protein, making it valuable for research and therapeutic applications . The His tag facilitates easy purification and detection of the protein in experimental settings.

Applications and Research

Research on PEDF has shown its potential in treating diseases characterized by abnormal blood vessel growth, such as diabetic retinopathy and certain cancers . Its neurotrophic properties also make it a candidate for neurodegenerative disease therapies . Ongoing studies aim to further elucidate its mechanisms and optimize its therapeutic use.

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.