PDGF A Human

Platelet-Derived Growth Factor A Human Recombinant
Shipped with Ice Packs
In Stock

Description

Platelet-Derived Growth Factor A Human Recombinant short chain produced in E.Coli is a non-glycosylated, polypeptide chain containing 110 amino acids fragment (87-196) and having a total Mw of 17.02kDa, with an amino-terminal hexahistidine tag.
PDGF-A is purified by proprietary chromatographic techniques.

Product Specs

Introduction
The term 'PDGF' refers to a family of disulphide bond-linked dimeric isoforms that act as autocrine and paracrine growth factors and are produced by a variety of cell types other than platelets. They act as potent mitogens for almost all mesenchymally-derived cells. Aberrant expression is involved in certain cancers, fibroproliferative disorders and atherosclerosis. The protein also contributes to wound healing and neural regeneration. There are four members of the PDGF family – PDGF A, PDGF B, PDGF C and PDGF D. Two distinct types of PDGF-A exist – a short form that is soluble and a long form that is retained by the extracellular matrix.
Description
Platelet-Derived Growth Factor A Human Recombinant short chain produced in E.Coli is a non-glycosylated, polypeptide chain containing 110 amino acids fragment (87-196) and having a total Mw of 17.02kDa, with an amino-terminal hexahistidine tag. PDGF-A is purified by proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered clear solution.
Formulation
Platelet-derived Growth Factor A is supplied in 25mM Na-Acetate, pH-4.8, 1mM EDTA and 50% glycerol.
Stability
Store at 4°C if entire vial will be used within 2-4 weeks. Store, frozen at -20°C for longer periods of time. Please avoid freeze thaw cycles.
Purity
Greater than 95.0% as determined by SDS-PAGE.
Synonyms
Glioma-derived growth factor, GDGF, Osteosarcoma-derived Growth Factor, ODGF, PDGF-A, PDGF-1.
Source
Escherichia Coli.

Q&A

What is the molecular structure of human PDGF-A and how does it differ from other PDGF family members?

Human PDGF-A is a secreted growth factor belonging to the PDGF/VEGF family. It functions primarily by forming homodimers (PDGF-AA) or heterodimers with PDGF-B (PDGF-AB) . Unlike PDGF-C and PDGF-D, which require proteolytic processing to become active, PDGF-AA is secreted in an active form capable of immediate receptor binding .

Methodological approach: To study PDGF-A structure, researchers should employ:

  • X-ray crystallography or cryo-EM for detailed structural analysis

  • Size-exclusion chromatography to confirm dimerization status

  • Western blotting under non-reducing and reducing conditions to distinguish between monomeric and dimeric forms

  • Recombinant expression systems using constructs of amino acids Ser87-Thr211 of the human PDGF-A sequence to obtain functional protein

Which receptors does PDGF-A activate and through what signaling pathways?

PDGF-AA primarily binds to and activates PDGFR-α homodimers, while heterodimeric PDGF-AB can activate both PDGFR-α homodimers and PDGFR-α/β heterodimers . Upon binding, these receptor tyrosine kinases initiate downstream signaling cascades involving ERK and Akt pathways that regulate cell proliferation, survival, and migration .

Methodological approach: To investigate PDGF-A receptor binding and signaling:

  • Perform receptor binding assays using labeled PDGF-AA

  • Analyze receptor phosphorylation by immunoprecipitation followed by phospho-tyrosine immunoblotting

  • Use pathway-specific inhibitors (e.g., ERK or Akt inhibitors) to dissect distinct aspects of PDGF-A-induced cellular responses

  • Employ receptor-specific antibodies or siRNA knockdown to confirm receptor specificity

How can researchers effectively distinguish between PDGF-A-specific and other PDGF isoform effects in experimental systems?

Distinguishing PDGF-A-specific effects from those of other PDGF isoforms presents a significant challenge in research due to overlap in receptor binding and downstream signaling pathways.

Methodological approach:

  • Use isoform-specific neutralizing antibodies, such as those against human PDGF-AA (e.g., AB-221-NA), which can selectively inhibit PDGF-AA activity

  • Employ recombinant PDGF-AA protein in rescue experiments after PDGF knockdown

  • Design siRNA or CRISPR-Cas9 approaches targeting unique regions of PDGF-A mRNA

  • Utilize receptor-selective compounds that preferentially block PDGFR-α versus PDGFR-β

  • Analyze cell types that predominantly express PDGFR-α but not PDGFR-β to isolate PDGF-AA effects

What are the most reliable methods for detecting and quantifying human PDGF-A in tissue samples and biological fluids?

Accurate detection and quantification of PDGF-A is essential for understanding its role in various physiological and pathological conditions.

Methodological approach:

  • ELISA: Use validated human PDGF-AA-specific ELISA kits for quantitative analysis in serum, plasma, or tissue lysates

  • Immunohistochemistry: Apply validated antibodies like AB-221-NA for tissue localization, with proper antigen retrieval (heat-induced epitope retrieval using basic pH buffers)

  • Western blotting: Employ reducing conditions to detect monomeric PDGF-A (approximately 14.4 kDa) or non-reducing conditions for dimeric PDGF-AA (approximately 28.9 kDa)

  • qRT-PCR: Design primers specific to human PDGF-A mRNA, distinguishing it from other PDGF isoforms

  • Mass spectrometry: For unambiguous identification and absolute quantification in complex biological samples

What are the optimal conditions for working with recombinant human PDGF-AA in cell culture experiments?

The proper handling and application of recombinant PDGF-AA is critical for obtaining reliable and reproducible results in cell culture studies.

Methodological approach:

  • Reconstitution: Reconstitute lyophilized PDGF-AA in sterile 4 mM HCl at a concentration of 0.2 mg/mL to maintain stability

  • Storage: Prepare aliquots to avoid freeze-thaw cycles and store at -80°C; for short-term storage, keep at 2-8°C for up to 1 month

  • Working concentrations: Typically, 25-200 ng/mL for proliferation assays in responsive cell lines

  • Vehicle controls: Include appropriate vehicle controls (e.g., 4 mM HCl diluted to the same extent as in PDGF-AA treatments)

  • Serum conditions: Perform experiments under reduced serum conditions (0.5-2% FBS) to minimize interference from serum-derived growth factors

  • Time course: Design experiments to capture both early (minutes to hours) and late (hours to days) responses to PDGF-AA stimulation

How should researchers design experiments to investigate PDGF-A's role in angiogenesis and vascular development?

PDGF-A plays significant roles in vascular biology, particularly in relation to pericyte function and blood-brain barrier maintenance.

Methodological approach:

  • In vitro models:

    • Endothelial cell tube formation assays with PDGF-AA supplementation

    • Co-culture systems with endothelial cells and pericytes or smooth muscle cells

    • Transwell migration assays to assess chemotactic responses

    • 3D spheroid sprouting assays to evaluate complex angiogenic responses

  • In vivo models:

    • Matrigel plug assays with PDGF-AA

    • Transgenic mouse models with cell-type specific PDGF-A overexpression or deletion

    • Zebrafish models for real-time visualization of vascular development

    • Analyze vascular density, pericyte coverage, and barrier function using techniques like immunofluorescence, Evans blue extravasation, or intravital microscopy

What experimental approaches are most effective for studying PDGF-A's role in cancer progression and angiogenesis?

PDGF-A signaling has been implicated in multiple aspects of cancer biology, including tumor cell proliferation, angiogenesis, and stromal interactions.

Methodological approach:

  • Patient sample analysis: Evaluate PDGF-A expression in tumor tissues versus matched normal tissues using immunohistochemistry

  • Cancer cell lines: Assess effects of PDGF-A modulation (overexpression, knockdown, or neutralization) on proliferation, migration, and invasion

  • Animal models: Develop xenograft models with PDGF-A-overexpressing or PDGF-A-knockout tumor cells

  • Targeting strategies:

    • Test PDGF-A neutralizing antibodies in preclinical models

    • Evaluate receptor tyrosine kinase inhibitors that target PDGFR-α

    • Investigate combination approaches targeting multiple angiogenic factors

How can researchers effectively study PDGF-A's functions in fibrotic diseases and potential therapeutic interventions?

PDGF signaling pathways have been implicated in fibrotic diseases, with evidence suggesting that targeting these pathways may have therapeutic potential.

Methodological approach:

  • Disease models: Utilize established models of fibrosis in relevant organs (kidney, liver, lung, skin)

  • Intervention strategies:

    • Anti-PDGF-A antibodies administered at different disease stages

    • PDGFR-α inhibitors to block downstream signaling

    • Combined targeting of multiple pro-fibrotic pathways

  • Assessment methods:

    • Histological analysis of fibrotic tissue (H&E, Masson's trichrome, Sirius Red)

    • Quantification of extracellular matrix proteins (collagens, fibronectin)

    • Evaluation of myofibroblast activation (α-SMA expression)

    • Functional measurements specific to the organ system being studied

What are common technical challenges when working with human PDGF-A and how can they be addressed?

Working with PDGF-A presents several technical challenges that researchers should anticipate and address to ensure reliable results.

Methodological approach:

  • Protein stability issues:

    • Use carrier-free formulations for applications where BSA might interfere

    • Add 0.1% BSA to diluted solutions to prevent protein adsorption to tubes and plates

    • Minimize freeze-thaw cycles by preparing single-use aliquots

    • Store reconstituted protein at -80°C for long-term storage

  • Specificity concerns:

    • Validate antibody specificity using positive and negative controls

    • Perform blocking experiments with recombinant PDGF-A

    • Include isotype controls in immunostaining experiments

  • Activity measurement:

    • Use established responsive cell lines like NR6R-3T3 for proliferation assays

    • Include positive controls (known active PDGF-AA) in each experiment

    • Monitor response using multiple endpoints (proliferation, signaling pathway activation)

How should researchers interpret and reconcile contradictory findings regarding PDGF-A expression or function in the literature?

The PDGF field contains some apparently contradictory findings, which may result from methodological differences, context-dependent effects, or varying isoform specificity.

Methodological approach:

  • Critically evaluate:

    • Detection methods used (antibody specificity, mRNA vs. protein)

    • Experimental models (cell types, species differences, in vitro vs. in vivo)

    • Disease context and stage (acute vs. chronic, early vs. late)

    • Co-expression of other growth factors that may compensate or interact

  • Reproducibility assessment:

    • Replicate key experiments using multiple methodologies

    • Validate findings across different cell lines or animal models

    • Consider sex differences and genetic background effects in animal studies

    • Perform meta-analyses of published data when sufficient literature exists

Table 1: Comparison of PDGF Isoforms, Receptors, and Functions

PDGF IsoformReceptor SpecificityActivation MechanismKey Biological FunctionsCellular Sources
PDGF-AAPDGFR-α homodimersSecreted in active formMesenchymal cell proliferation, embryonic developmentPlatelets, macrophages, epithelial cells, tumor cells
PDGF-BBPDGFR-α and PDGFR-β (both homo- and heterodimers)Secreted in active formPericyte recruitment, vascular stability, wound healingPlatelets, endothelial cells, smooth muscle cells
PDGF-ABPDGFR-α homodimers, PDGFR-α/β heterodimersSecreted in active formOverlapping with AA and BBPlatelets, macrophages
PDGF-CCPDGFR-α homodimers, PDGFR-α/β heterodimersRequires proteolytic removal of CUB domainEmbryonic development, tissue remodelingEpithelial cells, fibroblasts
PDGF-DDPDGFR-β homodimers, PDGFR-α/β heterodimersRequires proteolytic removal of CUB domain by matriptaseVascular function, wound healing, renal processesFibroblasts, smooth muscle cells

How does PDGF-A activity compare with other PDGF family members in specific pathological contexts?

Understanding the distinct roles of different PDGF isoforms in pathological processes is crucial for developing targeted therapeutic strategies.

Methodological approach:

  • Expression profiling:

    • Comprehensive analysis of all PDGF ligands and receptors in the same samples

    • Correlation of expression patterns with disease progression markers

    • Cell-type specific expression analysis using single-cell approaches

  • Functional comparisons:

    • Side-by-side testing of different PDGF isoforms at equimolar concentrations

    • Selective inhibition studies using isoform-specific neutralizing antibodies

    • Receptor knockout or knockdown studies to define receptor dependency

  • Disease models with selective targeting:

    • Compare effects of targeting individual PDGF isoforms in the same disease model

    • Evaluate combination approaches targeting multiple PDGF isoforms

    • Analyze downstream signaling pathway activation by different PDGF isoforms

What are the most promising approaches for studying PDGF-A's role in the blood-brain barrier function and neurovascular unit?

Recent research has highlighted PDGF-B:PDGFRβ signaling in brain pericytes as critical for blood-brain barrier (BBB) maintenance, with implications for neurodegenerative diseases like Alzheimer's.

Methodological approach:

  • In vitro BBB models:

    • Transwell co-culture systems with brain endothelial cells, pericytes, and astrocytes

    • Measurement of barrier integrity using TEER (transendothelial electrical resistance) and permeability assays

    • Manipulation of PDGF-A levels and assessment of effects on tight junction proteins

  • In vivo approaches:

    • Two-photon microscopy to visualize BBB integrity in real-time

    • Mouse models with cell-type specific PDGF-A deletion or overexpression

    • Assessment of cerebrovascular function in disease models following PDGF-A modulation

  • Clinical correlations:

    • Analysis of CSF and plasma PDGF-A levels in patients with BBB dysfunction

    • Correlation of PDGF-A levels with neuroimaging markers of BBB integrity

    • Investigation of genetic variants in PDGF-A and their association with neurovascular diseases

How can advanced 'omics and systems biology approaches enhance our understanding of PDGF-A signaling networks?

Modern 'omics technologies offer unprecedented opportunities to understand PDGF-A signaling in a comprehensive, systems-level context.

Methodological approach:

  • Multi-omics integration:

    • Transcriptomics to identify PDGF-A-responsive genes

    • Proteomics to map phosphorylation cascades downstream of PDGFR-α

    • Metabolomics to uncover metabolic changes induced by PDGF-A signaling

    • Integration of multiple data types using computational approaches

  • Network analysis:

    • Construction of PDGF-A signaling networks based on experimental data

    • Identification of key network nodes and potential therapeutic targets

    • Comparison of network perturbations across different disease contexts

  • Spatial analysis:

    • Spatial transcriptomics to map PDGF-A signaling in complex tissues

    • Single-cell approaches to define cell-type specific responses

    • Analysis of cell-cell communication networks mediated by PDGF-A

Product Science Overview

Biological Properties and Functions

PDGF-A is integrally involved in several critical biological processes, including:

  • Embryonic Development: PDGF-A is essential for the development of various tissues and organs during embryogenesis .
  • Angiogenesis: It promotes the formation of new blood vessels, which is vital for tissue growth and repair .
  • Organogenesis: PDGF-A plays a role in the formation and development of organs .
  • Fibroblast Proliferation and Migration: It induces the proliferation and migration of fibroblasts, which are crucial for wound healing and tissue repair .
Mechanism of Action

PDGF-A exerts its effects by binding to the PDGF receptor alpha (PDGFR-α), a receptor tyrosine kinase. This binding activates several intracellular signaling pathways, including:

  • Ras-MAPK Pathway: Involved in cell proliferation and differentiation .
  • PI3K/AKT Pathway: Plays a role in cell survival and growth .
  • PLCγ Pathway: Involved in various cellular processes, including cell motility and metabolism .
Production and Purification

Recombinant human PDGF-A is typically produced in Escherichia coli and undergoes several purification steps to ensure high purity and bioactivity. The recombinant protein is often stored in a lyophilized state and reconstituted with carrier proteins to maintain stability .

Clinical Applications

Recombinant PDGF-A has shown promise in various clinical applications, particularly in regenerative medicine. It is used to promote the healing of periodontal and orthopedic bone defects, as well as in dermal wound healing . The use of recombinant PDGF-A in combination with bone grafts has demonstrated significant clinical benefits in tissue regeneration .

Regulatory Mechanisms

The expression and activity of PDGF-A are tightly regulated at multiple levels, including:

  • Transcriptional Regulation: Various transcription factors and signaling molecules regulate the expression of PDGF-A genes .
  • Post-translational Modifications: PDGF-A undergoes proteolytic cleavage to form the active protein, and its activity can be modulated by glycosylation and other modifications .
  • Extracellular Matrix Binding: PDGF-A can bind to components of the extracellular matrix, which influences its availability and activity in the tissue microenvironment .
Dysregulation and Disease

Dysregulation of PDGF-A expression or signaling is often associated with various pathological conditions, including cancer and fibrotic disorders . Overexpression of PDGF-A can lead to uncontrolled cell proliferation and migration, contributing to tumor growth and metastasis .

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.