PEDF Human

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

Description

Gene Organization

The SERPINF1 gene is located on human chromosome 17p13.1, spanning ~15.6 kb with 8 exons and 7 introns. Its promoter region contains binding sites for transcription factors like HNF4 and USF, regulating tissue-specific expression . PEDF is evolutionarily conserved in vertebrates, with homologs absent in invertebrates like fruit flies and worms .

Protein Architecture

PEDF consists of 418 amino acids, including a 19-residue N-terminal secretion signal, a 34-mer anti-angiogenic domain (residues 24–57), and a 44-mer neurotrophic domain (residues 58–101) . Key structural features include:

  • Asymmetric charge distribution: One side is highly basic, the other acidic, enabling interactions with heparin and cell surface receptors .

  • Reactive center loop (RCL): Unlike other serpins, PEDF’s RCL lacks protease inhibitory activity .

  • Glycosylation: A single N-linked glycosylation site at residue 285 enhances secretion and stability .

Table 1: PEDF Gene and Protein Features

FeatureDescription
Gene locationChromosome 17p13.1 (15.6 kb)
Exons8 exons, 7 introns
Protein size50 kDa, 418 amino acids
Key domainsAnti-angiogenic (24–57), Neurotrophic (58–101), RCL (365–390)
GlycosylationSingle site (residue 285)

Anti-Angiogenic Activity

PEDF inhibits endothelial cell migration and neovascularization by:

  • Inducing apoptosis: Activates the FAS/FASL and p38 MAPK pathways to promote endothelial cell death .

  • Blocking VEGF signaling: Interferes with VEGFR-1/2 by enhancing γ-secretase activity, cleaving the receptors .

  • Upregulating anti-angiogenic factors: Stimulates thrombospondin and PPAR-γ, which suppress angiogenesis .

Neurotrophic and Neuroprotective Effects

PEDF promotes neuronal differentiation and survival by:

  • Inducing neuronal markers: Increases neuron-specific enolase and neurofilament expression in retinoblastoma cells .

  • Suppressing apoptosis: Blocks glutamate- and peroxide-induced neuronal death via NF-κB and ERK5 pathways .

Anti-Tumorigenic and Tumor-Promoting Roles

PEDF exhibits dual roles depending on context:

  • Tumor suppression: Inhibits cancer cell proliferation, induces apoptosis, and reduces metastasis via FAS/FASL .

  • Tumor promotion: Overexpression observed in glioma stem cells and hepatocellular carcinoma, linked to stem cell self-renewal .

Clinical Relevance

  • Cancer: PEDF expression inversely correlates with melanoma progression but is elevated in esophageal and hepatocellular carcinomas, suggesting context-dependent roles .

  • Diabetes and Liver Disease: PEDF deficiency leads to hepatic steatosis, while elevated circulating PEDF in obesity may contribute to insulin resistance .

  • Exercise and Senescence: Recent studies show PEDF suppresses cellular senescence in skeletal muscle and lung tissues, mitigating age-related pathologies like COPD .

Applications in Disease Models

DiseaseMechanism of ActionClinical Outcome Potential
Choroidal neovascularizationInhibits VEGF-driven angiogenesisTreatment for macular degeneration
CancerInduces apoptosis, inhibits metastasisTarget for anti-metastatic therapies
Diabetic retinopathyProtects neurons from oxidative stressNeuroprotective adjuvant therapy
Pulmonary emphysemaReduces senescence markers, preserves lung functionSenolytic therapy candidate

Challenges and Considerations

  • Dual roles in cancer: PEDF’s tumor-suppressive effects must be balanced against its promotion of stem cell survival in certain cancers .

  • Delivery strategies: Systemic administration risks off-target effects; localized delivery (e.g., intravitreal) may optimize efficacy .

Product Specs

Introduction
PEDF, Pigment Epithelium-Derived Factor, is a multifaceted protein known for its neurotrophic, anti-angiogenic, and anti-tumorigenic properties. Found abundantly throughout the body, PEDF acts as a natural defense mechanism against uncontrolled blood vessel growth (angiogenesis), a hallmark of many diseases. It achieves this by prompting the self-destruction of proliferating endothelial cells, the building blocks of blood vessels, and by hindering the action of pro-angiogenic factors like VEGF and FGF-2. Furthermore, PEDF exhibits neuroprotective qualities, shielding neurons from damage caused by oxidative stress and excitotoxicity. Its interaction with a specific cell surface receptor, PEDF-R, is key to its multifaceted biological activities. This unique receptor-ligand partnership distinguishes PEDF within the serpin family and offers valuable insights into its mechanisms of action. The remarkable attributes of PEDF have sparked considerable interest in its therapeutic potential for conditions like age-related macular degeneration (AMD) and diabetic retinopathy, where abnormal angiogenesis plays a central role. Moreover, PEDF holds promise as a potential weapon against various cancers, further underscoring its significance in biomedical research.
Description
Recombinant Human PEDF, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 400 amino acids with a molecular weight of 44.5 kDa. It undergoes purification using proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White lyophilized powder.
Formulation
The protein solution, concentrated to 1 mg/ml and sterile filtered, was lyophilized with a buffer solution of 20mM sodium phosphate and 150mM NaCl, at a pH of 7.4.
Solubility
To reconstitute the lyophilized PEDF, it is recommended to dissolve it in sterile 18M-cm H₂O at a concentration not less than 100 µg/ml. This solution can then be further diluted into other aqueous solutions as needed.
Stability
Lyophilized PEDF, though stable at room temperature for up to 3 weeks, should ideally be stored desiccated at a temperature below -18°C. After reconstitution, it is advisable to store Human PEDF at 4°C for a period of 2-7 days. For long-term storage, freezing below -18°C is recommended. To enhance stability during long-term storage, consider adding a carrier protein such as 0.1% HSA or BSA. It is crucial to avoid repeated freeze-thaw cycles to maintain protein integrity and activity.
Purity
The purity of the recombinant Human PEDF is determined to be greater than 95% as assessed by SDS-PAGE analysis.
Synonyms
Pigment epithelium-derived factor,PEDF, Serpin-F1, SerpinF1, EPC-1, EPC1, PIG35.
Source
Escherichia Coli.
Amino Acid Sequence
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 PEDF and how was it discovered?

PEDF was first isolated from medium conditioned by human fetal retinal pigment epithelial cells in the late 1980s by researchers Joyce Tombran-Tink and Lincoln Johnson . They initially observed that the retinal pigmented epithelium (RPE) produced a factor promoting differentiation of primitive retinal cells into cells with neuronal phenotypes. After isolating proteins unique to RPE cells and testing them individually for neurotrophic function, they identified a neurotrophic protein around 50 kilodaltons, temporarily named RPE-54 before being officially termed pigment epithelium-derived factor . Subsequent sequencing revealed PEDF as a previously uncharacterized member of the serpin (serine protease inhibitor) family .

What is the genomic structure of PEDF and where is it expressed?

In humans, PEDF is encoded by the SERPINF1 gene located on chromosome 17p13.1 . The human PEDF gene spans approximately 15.6kb with an mRNA transcript of around 1.5kb . The gene structure consists of 8 exons and 7 introns with a 200bp promoter region containing putative binding sites for the transcription factors HNF4, CHOP, and USF . PEDF is widely expressed in human tissues, with particularly strong expression in healthy skin and melanocytes, while showing diminished expression in certain cancerous tissues like malignant melanoma . Researchers should consider tissue-specific expression patterns when designing experimental protocols for PEDF studies.

How should researchers detect and quantify PEDF in experimental samples?

Multiple methodological approaches can be employed to detect PEDF in research settings. Immunohistochemistry using monoclonal antibodies specific to human PEDF (typically at 1/200 dilution) is effective for tissue sections following formalin fixation and paraffin embedding . Western blotting using anti-PEDF monoclonal antibodies offers quantitative protein assessment, with GAPDH commonly used as a loading control for normalization . For mRNA expression analysis, semi-quantitative RT-PCR with PEDF-specific primers normalized to GAPDH provides reliable results . Researchers should consider using a biotin-streptavidin method with DAB as a chromogen for immunoperoxidase staining when working with formalin-fixed, paraffin-embedded tissue sections .

What are the key receptors mediating PEDF's biological effects?

PEDF interacts with several cell-surface receptors that mediate its diverse biological activities. The primary receptors include PEDF receptor (PEDFR; encoded by PNPLA2, also known as desnutrin, ATGL, or iPLA2ζ), laminin receptor, F1 ATPase/synthase, and low-density lipoprotein receptor-related protein 6 (LRP6) . Additionally, PEDF demonstrates binding affinity for extracellular matrix components including heparin, heparan sulphate, hyaluronan, and collagens . The amino acids crucial for these interactions have been mapped on human PEDF: basic amino acids (Lys146, Lys147, and Arg149) for heparin binding; other basic residues (Lys189, Lys191, Arg194, and Lys197) for hyaluronan binding; and acidic amino acids (Asp256, Asp258, and Asp300) for collagen binding . Understanding these receptor interactions is essential for designing experiments examining PEDF's tissue-specific functions.

Advanced Research Questions

Several experimental approaches have proven effective for studying PEDF's antitumorigenic properties. In vitro, researchers should consider measuring PEDF's effects on cell proliferation, migration, invasion, and apoptosis induction using established cancer cell lines alongside normal cell counterparts . For example, studies comparing normal human melanocytes with the A375 melanoma cell line have revealed significantly reduced PEDF expression at both protein and mRNA levels in the malignant cells .

For in vivo studies, researchers can employ gene delivery methods including viral vectors (direct administration or via infected mesenchymal stem cells), microparticles or nanoparticles of various compositions, or implanted micro-osmotic pumps . Overexpression of PEDF in melanoma cells has been shown to greatly inhibit subcutaneous tumor formation and completely prevent lung and liver metastasis . When designing such experiments, researchers should include appropriate controls and consider combination approaches with other agents, such as differentiation-inducing factors like IL-6 or with radiotherapy, which have shown promising results .

How should researchers reconcile contradictory findings regarding PEDF in hepatocellular carcinoma?

To resolve these contradictions, researchers should implement multi-level experimental designs that:

  • Simultaneously measure PEDF at protein and mRNA levels

  • Compare tissue and serum levels within the same subjects

  • Account for disease progression stages and treatment status

  • Consider liver-specific microenvironmental factors that might uniquely modulate PEDF

  • Analyze PEDF isoforms and post-translational modifications that might affect function

This apparent discrepancy may reflect tissue-specific regulatory mechanisms unique to the liver microenvironment that warrant further investigation through carefully designed comparative studies.

What are the optimal methodologies for studying PEDF's effects on tumor microenvironment?

Investigating PEDF's impact on the tumor microenvironment requires integrative approaches addressing both direct effects on cancer cells and indirect effects on stromal components. For comprehensive analysis, researchers should employ multi-modal methodologies including:

  • Co-culture systems incorporating cancer cells with endothelial cells, immune cells, and fibroblasts to study cell-cell interactions under PEDF influence

  • Three-dimensional organoid cultures that better recapitulate tissue architecture compared to traditional monolayer cultures

  • In vivo models with cell-specific PEDF expression or deletion to distinguish direct versus microenvironment-mediated effects

  • Multiplex immunofluorescence or immunohistochemistry to simultaneously visualize PEDF along with markers of angiogenesis, immune infiltration, and extracellular matrix remodeling

  • Single-cell RNA sequencing to capture cell-type specific responses to PEDF within heterogeneous tumor environments

What strategies exist for PEDF-based therapeutic development?

Current efforts to optimize PEDF for cancer treatment focus on two primary strategies. The first involves developing protein or peptide therapeutics with enhanced antitumorigenic activity, including PEDF phosphomimetics, PEDF variant forms, or discrete PEDF-derived peptides that recapitulate the anticancer activity of full-length PEDF . The second approach focuses on establishing efficient delivery methods utilizing viral vectors (either directly or via virally infected human mesenchymal stem cells), various micro/nanoparticle compositions, or implanted micro-osmotic pumps .

Researchers investigating therapeutic applications should consider combination protocols using PEDF alongside differentiation-inducing agents such as IL-6 or with radiotherapy, which have shown promising results in experimental models . For example, overexpression of PEDF by melanoma cells significantly inhibits subcutaneous tumor formation and completely prevents lung and liver metastasis in experimental models . Similarly, PEDF overexpression has been shown to decrease angiogenesis and inhibit the growth of human malignant melanoma cells in vivo .

How should researchers evaluate PEDF's dual role in metastasis suppression and normal tissue protection?

PEDF demonstrates remarkable dual functionality as both a metastasis suppressor and a protector of normal tissues, particularly evident in the context of brain metastases. Researchers investigating this dual role should implement experimental designs that simultaneously assess both functions within the same model systems. In brain metastases of breast cancer origin, PEDF is downregulated compared to primary breast tumors . Studies have shown that PEDF overexpression decreases metastasis of human and mouse breast cancer cells to the brain in a rapid and angiogenesis-independent manner, while simultaneously protecting neurons from tumor-induced damage .

To effectively study this dual functionality, researchers should:

  • Develop co-culture systems of cancer cells with normal cells of the target organ

  • Employ in vivo models that allow simultaneous assessment of metastasis inhibition and normal tissue protection

  • Implement tissue-specific conditional expression systems to modulate PEDF levels

  • Utilize advanced imaging techniques to track both cancer cell behavior and normal tissue integrity in response to PEDF

  • Develop quantitative assays that can measure both anti-migratory effects on cancer cells and pro-survival effects on normal cells

Understanding this dual role could inform the development of more targeted therapeutic approaches that maximize PEDF's beneficial effects while minimizing potential side effects.

What are the critical controls for PEDF expression studies?

When investigating PEDF expression, researchers must implement rigorous controls to ensure reliable and reproducible results. For protein expression studies, appropriate loading controls such as GAPDH should be used for Western blot normalization, as demonstrated in studies comparing PEDF expression between normal human melanocytes and A375 melanoma cells . For immunohistochemistry, researchers should include positive controls (tissues known to express high PEDF levels, such as healthy skin) and negative controls (antibody omission or isotype controls) .

When analyzing PEDF mRNA expression via RT-PCR or other nucleic acid quantification methods, appropriate housekeeping genes should be selected for normalization, with GAPDH commonly used as demonstrated in comparative studies between melanocytes and melanoma cells . Multiple housekeeping genes may provide more robust normalization. Additionally, researchers should consider the heterogeneity of tissue samples, particularly in tumor studies, where microdissection techniques may help isolate specific cell populations for more precise expression analysis.

How should researchers differentiate between PEDF's multiple biological functions in experimental design?

PEDF exhibits multiple biological activities including anti-angiogenic, antitumorigenic, antimetastatic, and neurotrophic functions. Differentiating between these activities requires thoughtful experimental design. Researchers should consider employing:

  • Structure-function studies using specific PEDF domains or peptides known to mediate distinct functions

  • Targeted mutation of PEDF amino acid residues critical for specific interactions

  • Selective receptor blocking antibodies or small molecules to inhibit particular pathways

  • Cell type-specific assays that isolate individual functions (e.g., endothelial tube formation for angiogenesis, neuronal differentiation assays for neurotrophic effects)

  • Temporal analysis to distinguish between immediate versus delayed effects that might reflect different mechanisms

For example, the ability of PEDF to simultaneously induce growth arrest, promote tumor cell differentiation to less-malignant phenotypes, and protect normal neuronal cells requires sophisticated experimental approaches to disentangle these overlapping functions .

What emerging technologies might enhance PEDF research?

Several cutting-edge technologies show promise for advancing PEDF research. CRISPR-Cas9 gene editing can create precise PEDF knockout or knock-in models to study function in specific contexts. Single-cell RNA sequencing could reveal cell-specific responses to PEDF within heterogeneous tumor microenvironments. Advanced imaging techniques including intravital microscopy might allow real-time visualization of PEDF effects on angiogenesis and tumor cell behavior in living organisms.

Proteomics approaches could identify novel PEDF-interacting proteins and post-translational modifications that regulate activity. Three-dimensional bioprinting of tissues incorporating PEDF might provide more physiologically relevant models for studying its complex functions. Additionally, computational approaches including machine learning could help predict optimal PEDF-derived peptides for specific therapeutic applications and identify patient populations most likely to benefit from PEDF-based interventions.

What are the most promising clinical applications for PEDF research?

Based on current evidence, several clinical applications show particular promise for PEDF research. In oncology, PEDF has demonstrated potential as both a biomarker and therapeutic agent across multiple tumor types, with particularly strong evidence in melanoma, retinoblastoma, and brain metastases from breast cancer . The inverse correlation between PEDF expression and malignancy in most cancers suggests potential prognostic value .

Beyond cancer, PEDF shows promise for retinal diseases due to its protective effects on retinal neurons . Research indicates PEDF offers greater protection to the retina, and methods for delivering therapeutically active PEDF have potential clinical advantages for longer-term treatments of retinal diseases . Additionally, PEDF may have applications in cardiovascular disease, as it inhibits angiotensin II-induced T cell proliferation by blocking autocrine production of IL-2 via suppression of NADPH oxidase-mediated reactive oxygen species generation, potentially becoming a novel therapeutic target for atherosclerosis .

Product Science Overview

Discovery and Identification

PEDF was first discovered in the late 1980s by researchers Joyce Tombran-Tink and Lincoln Johnson. They were studying human retinal cell development and identified PEDF as a secreted factor produced by the retinal pigmented epithelium (RPE), a layer of cells that supports the retina .

Structure and Function

PEDF is a 50 kDa secreted glycoprotein that belongs to the serpin (serine protease inhibitor) family, specifically classified as serpin F1. Despite being a serpin, PEDF does not exhibit protease inhibitory activity. Instead, it is known for its anti-angiogenic, anti-tumorigenic, and neurotrophic functions .

Biological Activities
  1. Anti-Angiogenic Activity: PEDF is a potent inhibitor of angiogenesis, the process of new blood vessel formation. This property makes it a promising therapeutic candidate for diseases characterized by abnormal blood vessel growth, such as age-related macular degeneration (AMD) and certain types of cancer .
  2. Neurotrophic Effects: PEDF promotes the survival and differentiation of neurons. It has been shown to protect retinal cells from degeneration and is being explored for its potential in treating neurodegenerative diseases .
  3. Anti-Tumorigenic Properties: PEDF exhibits anti-tumorigenic effects by inhibiting the growth and metastasis of various cancer cells. Its ability to induce apoptosis (programmed cell death) in tumor cells further enhances its therapeutic potential .
Recombinant PEDF

Recombinant PEDF refers to the protein produced through recombinant DNA technology, which allows for the large-scale production of this protein for research and therapeutic purposes. Human recombinant PEDF retains the biological activities of the naturally occurring protein and is used in various experimental and clinical studies .

Therapeutic Potential

Due to its multifunctional nature, PEDF is being investigated for its therapeutic potential in a wide range of conditions, including:

  • Ophthalmic Diseases: PEDF’s anti-angiogenic properties make it a candidate for treating diseases like AMD and diabetic retinopathy.
  • Cancer: Its ability to inhibit tumor growth and metastasis is being explored in cancer therapy.
  • Neurodegenerative Diseases: PEDF’s neuroprotective effects are being studied for potential applications in diseases such as Alzheimer’s and Parkinson’s .

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.