PDGF BB Mouse

Platelet-Derived Growth Factor BB Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Bone Development and Remodeling

PDGF-BB stimulates osteoblast differentiation and collagen gel contraction, critical for skeletal integrity . Elevated PDGF-BB from preosteoclasts in transgenic mice (Pdgfb cTG) accelerates age-related bone loss, while its knockout (Pdgfb cKO) attenuates osteoporosis but paradoxically reduces baseline bone mass .

Vascular and Blood-Brain Barrier (BBB) Regulation

  • Angiogenesis: PDGF-BB promotes pericyte recruitment to endothelial cells, stabilizing microvasculature .

  • Pathological Calcification: Aged male mice with high serum PDGF-BB (>2× baseline) develop thalamic vascular calcification, mediated by PDGFRβ/ERK/RUNX2 signaling in pericytes .

  • BBB Breakdown: Persistent PDGF-BB exposure upregulates MMP14 in pericytes, shedding PDGFRβ and increasing BBB permeability .

Brain Calcification

Aged mice exhibit thalamic calcification linked to elevated PDGF-BB (2,153.1 ± 69.7 pg/mL in serum) . Pdgfb cTG mice (serum PDGF-BB >30 ng/mL) replicate this phenotype, while Pdgfb cKO mice show no calcification .

Metabolic and Osteogenic Crosstalk

High-fat diet (HFD)-challenged mice with elevated PDGF-BB demonstrate accelerated bone loss and hippocampal BBB dysfunction, reversible via PDGF-BB inhibition .

Recombinant PDGF-BB

  • Purity: ≥95% by SDS-PAGE and HPLC .

  • Bioactivity: Induces dose-dependent proliferation in NIH/3T3 fibroblasts (EC₅₀: 1–10 ng/mL) .

Intra-/Inter-Assay Precision (ELISA):

Assay TypeSamplenMean (pg/mL)CV%
IntraAssay1202,153.13.2
InterAssay1241,958.91.9

Therapeutic Implications

PDGF-BB’s dual role in tissue repair and pathology highlights its therapeutic potential:

  • Bone Regeneration: Local PDGF-BB delivery enhances fracture healing by coupling osteogenesis and angiogenesis .

  • Neurological Disorders: Targeting PDGF-BB/PDGFRβ signaling may mitigate age-related brain calcification and BBB leakage .

Product Specs

Introduction
Platelet-derived growth factor BB (PDGF-BB) belongs to the platelet-derived growth factor family, known for stimulating the growth of mesenchymal cells. These factors are characterized by a conserved eight-cysteine motif. PDGF-BB exists as a homodimer or forms a heterodimer with the platelet-derived growth factor alpha polypeptide (PDGF-AB), linked by disulfide bonds. Genetic alterations in PDGF-BB are linked to meningioma. Additionally, chromosomal translocations involving the PDGF-BB gene on chromosome 22 and the COL1A1 gene on chromosome 7 are associated with a skin tumor, dermatofibrosarcoma protuberans, characterized by uncontrolled growth factor expression. Two splice variants of the PDGF-BB gene have been identified.
Description
Recombinant Mouse Platelet-Derived Growth Factor BB, produced in E.coli, is a non-glycosylated homodimeric polypeptide comprising two chains of 110 amino acids each (total 220 amino acids). It has a molecular weight of 24.7 kDa. The protein is purified using proprietary chromatographic techniques.
Physical Appearance
White, lyophilized (freeze-dried) powder, sterile-filtered.
Formulation
The protein was lyophilized from a solution of 10mM Sodium Citrate, pH 3.0.
Solubility
To reconstitute the lyophilized Platelet-derived Growth Factor-BB, it is recommended to dissolve it in sterile 100mM acetic acid containing 0.1% BSA to a concentration of at least 100 µg/ml. This solution can be further diluted in other aqueous solutions.
Stability
Lyophilized Platelet-derived Growth Factor BB, though stable at room temperature for 3 weeks, should be stored in a dry environment below -18°C. After reconstitution, PDGF-BB should be stored at 4°C for 2-7 days. For long-term storage, it is recommended to add a carrier protein (0.1% HSA or BSA) and store below -18°C. Avoid repeated freeze-thaw cycles.
Purity
The purity is determined to be greater than 95.0% using the following methods:
(a) Reverse-phase High-Performance Liquid Chromatography (RP-HPLC) analysis.
(b) Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis (SDS-PAGE) analysis.
Biological Activity
The biological activity is assessed by measuring the dose-dependent stimulation of human umbilical vein endothelial cell (HUVEC) proliferation. This is determined within a concentration range of less than 1.6 ng/ml.
Protein Content
Protein quantification is performed using two independent methods:
1. UV spectroscopy at 280 nm. An absorbance value of 0.48 is used as the extinction coefficient for a 0.1% (1 mg/ml) solution. This value is calculated using the PC GENE computer analysis program for protein sequences (IntelliGenetics).
2. RP-HPLC analysis, employing a standard solution of PDGF-BB as a reference standard.
Synonyms
Glioma-derived growth factor, GDGF, Osteosarcoma-derived Growth Factor, ODGF, SIS, SSV, PDGF2, c-sis, FLJ12858, PDGF-BB, PDGF B-chain, Platelet-derived growth factor beta polypeptide, Becaplermin.
Source
Escherichia Coli.
Amino Acid Sequence

MSLGSLAAAE PAVIAECKTR TEVFQISRNL IDRTNANFLV WPPCVEVQRC SGCCNNRNVQ CRASQVQMRP VQVRKIEIVR KKPIFKKATV TLEDHLACKC ETIVTPRPVT

Q&A

What is PDGF-BB and how does it function in mouse models?

PDGF-BB is the homodimeric protein formed by two B chains of platelet-derived growth factor. In mouse research, PDGF-BB functions as a major growth factor found in bone matrix and has been shown to increase bone formation after intravenous administration . It's important to distinguish between PDGFB (the gene encoding the human platelet-derived growth factor B chain) and PDGF-BB (the resulting homodimeric protein) .

PDGF-BB exerts its effects through binding to PDGF receptors, particularly PDGFR-β, activating downstream signaling pathways that regulate cell proliferation, migration, and differentiation. In vitro studies confirm that PDGF-BB stimulates fibroblast proliferation in a dose-dependent manner, with proliferation significantly reduced when cells are blocked with an antibody to the PDGF beta receptor . Interestingly, PDGF-BB shows a trimodal dose-response curve in cell proliferation assays, with peak proliferation at 7.5 ng/ml, 50 ng/ml, and 7.5 μg/ml .

Beyond bone formation, PDGF-BB has been investigated for its potential role in wound healing, though results from diabetic mouse models show conflicting efficacy depending on experimental conditions .

How can PDGF-BB levels be accurately measured in mouse samples?

PDGF-BB can be measured in mouse samples using several methodological approaches:

  • Enzyme-Linked Immunosorbent Assay (ELISA): Commercial ELISA kits offer high sensitivity for PDGF-BB detection in mouse samples. For example, the Abcam Mouse PDGF BB ELISA Kit (ab224879) offers a sensitivity of 1.9 pg/ml with a detection range of 9.375-600 pg/ml . This method is suitable for multiple sample types including serum, plasma (citrate, EDTA, or heparin), and cell culture supernatants.

  • Western blotting: For qualitative or semi-quantitative assessment of PDGF-BB protein expression in tissue lysates.

  • Immunohistochemistry: To visualize PDGF-BB distribution in tissue sections and assess localization patterns.

  • qRT-PCR: For quantification of PDGFB gene expression at the mRNA level.

When measuring PDGF-BB in mouse samples, several methodological considerations are important:

  • Sample collection timing relative to the experimental intervention

  • Appropriate sample processing and storage (typically at -80°C with protease inhibitors)

  • Inclusion of proper controls to account for background signal and matrix effects

  • Validation of antibody specificity, particularly to distinguish between different PDGF isoforms

What are the effects of PDGF-BB on bone formation in mouse models?

PDGF-BB significantly influences bone formation in mouse models through multiple mechanisms:

  • Mesenchymal stem cell (MSC) stimulation: PDGF-BB released from transplanted PDGFB-overexpressing Sca1+ cells stimulates MSC proliferation and recruits MSCs to hematopoietic stem cell niches at endocortical and trabecular surfaces .

  • Promotion of angiogenesis: PDGF-BB enhances blood vessel formation, which is critical for bone development and repair .

  • Cross-talk with BMP signaling: PDGF-BB treatment increases Bmp2 and Bmp4 production, with evidence suggesting interaction between PDGF-BB and BMP2 signaling pathways. The fact that PDGF-BB's bone formation effects require BMPs may explain why it specifically increases bone formation in bone tissue but not in other tissues .

  • Dual regulation of bone remodeling: While PDGF-BB enhances bone formation, it also stimulates bone resorption by increasing expression of Csf1 and Tnfsf11, elevating the number of TRAP+ osteoclasts at the bone surface, and raising serum CTX-1 levels . This increased bone remodeling may improve bone quality and mechanical performance.

In stem cell gene therapy approaches, mouse hematopoietic stem cells engineered to express PDGF-BB under the phosphoglycerate kinase (PGK) promoter demonstrated remarkable effects: a 45% increase in bone strength, 20-fold increase in trabecular connectivity, and substantial reduction in cortical porosity .

Why do PDGF-BB effects on wound healing in diabetic mice show inconsistent results?

The conflicting results regarding PDGF-BB efficacy in diabetic wound healing can be attributed to several methodological and biological factors:

  • Different wound healing mechanisms: As noted in research studies, skin contracture is the primary wound closure mechanism in rodents, unlike humans where re-epithelialization predominates . This fundamental difference complicates the interpretation of PDGF-BB effects across species.

  • Experimental model variations: The use of splinted versus non-splinted wound models significantly impacts results. Splinted wounds, which prevent contraction and force healing through re-epithelialization (more similar to human wound healing), show different PDGF-BB responses than non-splinted wounds .

  • Variable experimental parameters: Published reports show inconsistent efficacy depending on:

    • Wound size: Smaller wounds (as in some studies) versus larger wounds (15mm×15mm in other studies)

    • Study endpoints: 11 days versus 21 days

    • PDGF-BB dosage: Different concentrations yield varying results

    • Delivery vehicle: Different formulations affect bioavailability

  • Cell-specific responses: While PDGF-BB stimulates fibroblast proliferation in vitro, it does not show significant effects on keratinocyte (HaCaT) proliferation . This differential response may explain why PDGF-BB improves granulation tissue formation without necessarily accelerating re-epithelialization in some studies.

  • Complex diabetic pathophysiology: The db/db mouse model represents type 2 diabetes with multiple metabolic abnormalities that may influence tissue responses to growth factors .

A controlled study using splinted wounds in db/db mice found that daily application of 3 μg PDGF-BB for 10 days failed to accelerate wound healing compared to vehicle control, despite confirmed bioactivity in vitro .

How can PDGF-BB-based stem cell gene therapy be optimized for bone regeneration?

Optimizing PDGF-BB-based stem cell gene therapy for bone regeneration requires careful consideration of several parameters:

  • Promoter selection: Using a relatively weak physiologic promoter (phosphoglycerate kinase or PGK promoter) produces significant increases in endosteal/trabecular bone formation without elevating circulating PDGF-BB levels, avoiding adverse effects such as osteomalacia while achieving substantial improvements in bone strength .

  • Cell selection and targeting: Hematopoietic stem cells (HSCs), particularly Sca1+ cells, can home to HSC niches at endocortical and trabecular surfaces, providing targeted delivery of PDGF-BB to bone marrow where it can stimulate mesenchymal stem cell proliferation and recruitment .

  • Expression level calibration: Optimal PDGF-BB expression must balance bone formation with potential bone resorption effects. The research indicates that PDGF-BB directly stimulates bone resorption by increasing osteoclast formation . While this contributes to bone remodeling and may improve bone quality, excessive resorption could counteract anabolic effects.

  • Consideration of signaling interactions: The research suggests cross-talk between PDGF-BB and BMP signaling pathways, with PDGF-BB-induced nodule formation enhanced in the presence of BMP2 . This interaction may be leveraged to enhance therapeutic efficacy.

  • Safety profile assessment: The stem cell gene therapy approach appears potentially safer than intravascular injection of PDGF-BB because it achieves optimal levels in bone marrow without affecting baseline circulation levels .

Studies demonstrate that transplantation of mouse HSCs engineered to express PDGF-BB by the PGK promoter results in "rapid, robust, and persistent formation of mineralized bone, leading to a considerable increase in bone strength, which is attributed to large increase in de novo trabecular formation and trabecular connectivity, and a significant reduction in cortical porosity" .

What methodological considerations are crucial when designing PDGF-BB experiments in mouse models?

Several methodological considerations are essential for robust PDGF-BB experiments in mouse models:

  • Selection of appropriate wound models: For wound healing studies, researchers must consider that skin contracture is the primary mechanism in rodents, unlike re-epithelialization in humans . Using silicone splints to inhibit dermal contraction increases the relevance to human wound healing .

  • Dosage determination: In vitro studies reveal a trimodal dose-response relationship for PDGF-BB, with peaks at 7.5 ng/ml, 50 ng/ml, and 7.5 μg/ml . This complex response pattern necessitates testing multiple concentrations rather than assuming linear dose-response relationships.

  • Appropriate controls: Proper controls are essential, including vehicle controls matching all components of the PDGF-BB formulation except the active protein . For example, studies have used 5% PEG in phosphate-buffered solution as a vehicle control for PDGF-BB delivery .

  • Delivery method standardization: Different delivery approaches (topical application, injection, gene therapy) significantly impact bioavailability and efficacy. Some studies apply 30 μl of 0.01% PDGF-BB (3 μg) daily , while others use gene therapy approaches for sustained expression .

  • Timing and duration considerations: Treatment duration and experimental endpoint significantly influence outcomes. Studies report different results with 11-day versus 21-day endpoints .

  • Comprehensive assessment metrics: Multiple parameters should be evaluated beyond wound closure or bone formation, including histological analysis, molecular markers, functional measures (like bone strength), and cellular responses .

  • Animal model selection: Different mouse models (wild-type, diabetic db/db, etc.) respond differently to PDGF-BB. The db/db mouse, representing type 2 diabetes, is particularly valuable for studying impaired wound healing .

  • In vitro validation: Confirming PDGF-BB bioactivity through in vitro assays before in vivo application helps distinguish between delivery failures and true biological non-response .

How does PDGF-BB interact with other signaling pathways in mouse models?

PDGF-BB interacts with several signaling pathways in mouse models, creating complex regulatory networks:

  • BMP signaling cross-talk: Research demonstrates interaction between PDGF-BB and bone morphogenetic protein (BMP) signaling. PDGF-BB treatment increases Bmp2 and Bmp4 production and elevates levels of phosphorylated Smad1/5 (pSmad1/5) in treated femurs . The observation that PDGF-BB-induced nodule formation increases in the presence of BMP2 suggests synergistic pathway interaction .

  • Osteoclastogenesis regulation: PDGF-BB stimulates bone resorption by increasing expression of Csf1 (Colony stimulating factor 1) and Tnfsf11 (RANKL), key factors in osteoclast differentiation and activation . This demonstrates interaction with pathways controlling bone remodeling.

  • Angiogenesis promotion: PDGF-BB promotes angiogenesis, suggesting interaction with vascular endothelial growth factor (VEGF) signaling and other pathways regulating blood vessel formation .

  • Receptor-mediated signaling: PDGF-BB acts primarily through PDGFR-β, which is detected on osteoclasts and other cell types . The cell proliferation response to PDGF-BB is significantly reduced when cells are blocked with an antibody to the PDGF beta receptor, confirming the specificity of this signaling pathway .

  • Context-dependent interactions: The finding that PDGF-BB increases bone formation specifically in bone tissue but not in other tissues suggests that the tissue microenvironment, particularly the presence of BMPs and other bone-specific factors, may determine PDGF-BB signaling outcomes .

Understanding these pathway interactions is crucial for developing effective therapeutic strategies and may explain the tissue-specific and context-dependent effects of PDGF-BB observed in different experimental settings.

What are the advantages and limitations of different PDGF-BB delivery methods in mouse models?

Different PDGF-BB delivery methods offer distinct advantages and limitations in mouse models:

Topical Application

Advantages:

  • Direct delivery to target tissues (e.g., wounds)

  • Precise dosage control (e.g., 3 μg PDGF-BB in 30 μl vehicle)

  • Minimal systemic exposure

Limitations:

  • Limited tissue penetration

  • Requires frequent reapplication (daily in many protocols)

  • Efficacy may be compromised in proteolytic environments

  • Results show inconsistency across different wound models

Stem Cell-Based Gene Therapy

Advantages:

  • Targeted delivery to specific tissues (e.g., bone marrow)

  • Sustained local PDGF-BB production

  • Achieves therapeutic effects without elevating circulating levels

  • Allows fine-tuning of expression through promoter selection

  • Can achieve 45% increase in bone strength with significant improvement in trabecular connectivity

Limitations:

  • More complex implementation

  • Potential variability in transduction efficiency and cell survival

  • Requires specialized expertise and facilities

Intravenous Administration

Advantages:

  • Systemic distribution

  • Simplicity of administration

  • Has been shown to increase bone formation

Limitations:

  • Non-specific tissue targeting

  • Higher risk of off-target effects

  • May require higher doses to achieve therapeutic concentrations in target tissues

Vehicle Considerations

The choice of delivery vehicle significantly impacts PDGF-BB efficacy. Studies have used 5% PEG (polyethylene glycol) in phosphate-buffered solution as a vehicle for PDGF-BB delivery . The vehicle can affect:

  • Protein stability and bioactivity

  • Tissue penetration and cellular uptake

  • Release kinetics and local retention time

What biological markers indicate successful PDGF-BB activity in mouse models?

Several biological markers can be used to assess PDGF-BB activity in mouse models:

Bone Formation Markers

  • Cellular proliferation: Increased Ki-67+ cells in bone marrow space indicates enhanced mitogenic activity of mesenchymal bone progenitor cells in response to PDGF-BB .

  • Osteoblast activity: Approximately fivefold increase in alkaline phosphatase (ALP)-stained bone surface demonstrates enhanced osteoblast function in PDGF-BB-treated animals .

  • Bone morphogenetic protein expression: Elevated Bmp2 and Bmp4 production serves as a downstream marker of PDGF-BB activity .

  • Smad signaling: Increased levels of phosphorylated Smad1/5 (pSmad1/5) indicate active BMP signaling following PDGF-BB treatment .

  • Bone strength measurements: A 45% increase in maximal load to failure directly demonstrates functional improvement from PDGF-BB treatment .

  • Microstructural changes: 20-fold increase in trabecular connectivity and substantial reduction in cortical porosity provide structural evidence of PDGF-BB effects .

Bone Resorption Markers

  • Gene expression changes: Increased expression of Csf1 and Tnfsf11 (RANKL) indicates PDGF-BB-induced osteoclastogenesis .

  • Osteoclast numbers: Elevated TRAP+ (tartrate-resistant acid phosphatase) osteoclasts at the bone surface demonstrate enhanced bone resorption activity .

  • Serum CTX-1 levels: Elevated C-terminal telopeptide of type I collagen provides a systemic marker of increased bone resorption .

In Vitro Validation Markers

  • Cell proliferation: Fibroblast proliferation assays with and without PDGF receptor blocking antibodies confirm PDGF-BB bioactivity .

  • Dose-response relationship: The characteristic trimodal dose-response curve with peak proliferation at specific concentrations (7.5 ng/ml, 50 ng/ml, and 7.5 μg/ml) serves as a fingerprint of proper PDGF-BB activity .

How can researchers reconcile in vitro and in vivo discrepancies in PDGF-BB efficacy?

Reconciling discrepancies between in vitro bioactivity and in vivo efficacy of PDGF-BB requires systematic investigation of several factors:

  • Model-specific healing mechanisms: The primary disconnect may stem from fundamental differences in healing mechanisms. While PDGF-BB shows clear bioactivity in vitro, stimulating fibroblast proliferation in a dose-dependent manner, it failed to accelerate wound healing in vivo in db/db mice using the splinted wound model . This suggests that the predominant healing mechanism (re-epithelialization versus contraction) significantly influences PDGF-BB efficacy.

  • Cell type-specific responses: Research demonstrates that PDGF-BB stimulates fibroblast proliferation but shows no significant effect on keratinocyte (HaCaT) proliferation . Since re-epithelialization depends primarily on keratinocyte function, this differential response may explain limited efficacy in splinted wounds healing primarily through re-epithelialization.

  • Microenvironmental considerations: The in vivo environment introduces complexities absent in vitro:

    • Proteolytic degradation of applied PDGF-BB

    • Competition with endogenous growth factors

    • Interaction with extracellular matrix components

    • Influence of inflammatory mediators

  • Pharmacokinetic factors: Single daily applications of PDGF-BB (as used in some studies) may provide insufficient sustained levels compared to the continuous exposure in vitro or the sustained expression achieved through gene therapy approaches .

  • Multifactorial pathology in disease models: The diabetic environment presents multiple impairments beyond what a single growth factor can address, including:

    • Impaired angiogenesis

    • Chronic inflammation

    • Oxidative stress

    • Advanced glycation end products

  • Physiological complexity: The trimodal dose-response curve observed in vitro suggests complex signaling dynamics that may be further complicated in vivo, where multiple cell types and signaling pathways interact.

Product Science Overview

Introduction

Platelet-Derived Growth Factor BB (PDGF-BB) is a member of the PDGF family, which plays a crucial role in various biological processes, including embryonic development, cell proliferation, cell migration, and angiogenesis . PDGF-BB is one of the isoforms of the PDGF family, which also includes PDGF-AA, PDGF-CC, PDGF-DD, and the heterodimer PDGF-AB .

Structure and Function

PDGF-BB is a homodimer composed of two B chains, each with a molecular weight of approximately 12.5 kDa, resulting in a total molecular weight of 25.0 kDa . The protein is produced in Escherichia coli (E. coli) and purified via sequential chromatography . PDGF-BB signals through receptor tyrosine kinases, specifically PDGFRα and PDGFRβ .

Biological Activity

PDGF-BB is a potent mitogen for cells of mesenchymal origin, such as fibroblasts, glial cells, and vascular smooth muscle cells . It has been shown to induce mesenchymal stem cell migration and angiogenesis, which are essential for proper osteogenesis during bone modeling and remodeling . Additionally, PDGF-BB is involved in the pathogenesis of various diseases, including atherosclerosis, glomerulonephritis, and cancer .

Applications

Recombinant mouse PDGF-BB is widely used in research to study its effects on cell proliferation, migration, and angiogenesis. It is also utilized in tissue engineering and regenerative medicine to promote tissue repair and regeneration .

Storage and Handling

The lyophilized protein should be stored at 2–8°C or −20°C for long-term storage, preferably desiccated . Upon reconstitution, it should be apportioned into working aliquots and stored at ≤ −20°C to avoid repeated freeze-thaw cycles .

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.