Recombinant Human Platelet-derived Growth Factor-BB (PDGFB), partial (Active) (GMP)

Shipped with Ice Packs
In Stock

Description

Definition and Functional Role

PDGF-BB is a homodimer of two B chains, with a molecular mass of ~24–28 kDa depending on glycosylation . It binds PDGFR-α and PDGFR-β tyrosine kinase receptors, activating pathways for:

  • Mitogenesis: Proliferation of fibroblasts, smooth muscle cells, and glial cells .

  • Chemotaxis: Recruitment of neutrophils, macrophages, and MSCs to wound sites .

  • Tissue Repair: Enhances extracellular matrix synthesis and osteogenic differentiation .

GMP-grade PDGF-BB is manufactured under stringent quality controls for use in ex vivo cell expansion and clinical-grade therapeutics .

Production Systems

  • Expression Hosts: Yeast (Saccharomyces cerevisiae) or E. coli .

  • Purification: Tag-free, animal component-free processes .

Stability

  • Storage: -20°C long-term; stable at 4°C for 180 days .

  • Reconstitution: 0.1–1.0 mg/mL in sterile water or 4 mM HCl .

Receptor Binding

  • PDGF-BB binds PDGFR-β with high affinity, activating MAPK and PI3K pathways .

  • Cross-reactivity with VEGF and EGF pathways observed in tissue regeneration .

Approved Uses

  • Periodontal Regeneration: Combined with β-TCP alloplasts for intrabony defects (FDA-approved) .

  • Chronic Wounds: Topical application accelerates pressure ulcer healing (ED50: 1.5–6 ng/mL) .

Emerging Applications

  • Ex Vivo MSC Expansion: Enhances proliferation without differentiation .

  • Guided Bone Regeneration (GBR): Synergistic effects with allografts/xenografts .

Efficacy in Wound Healing

Study TypeDose (µg/mL)Outcome (vs. Placebo)Reference
Phase I/II RCT10085.9% reduction in ulcer depth
Systematic Review0.1–1.092% success in gingival recession

Regulatory and Compliance Standards

  • Guidelines: USP <1043>, ISO 20399:2022, Ph. Eur. 5.2.12 .

  • GMP Compliance: Full traceability of raw materials, bioburden testing (USP <61>) .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered solution of 20mM NaAc-HAc, pH 4.5.
Form
Lyophilized powder
Lead Time
Typically, we can ship the products within 5-10 business days after receiving your order. Delivery time may vary depending on the method of purchase or location. Please consult your local distributor for specific delivery details.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend briefly centrifuging the vial prior to opening to ensure the contents settle at the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We suggest adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final concentration of glycerol is 50%, which you can use as a reference.
Shelf Life
The shelf life is influenced by several factors, including storage conditions, buffer components, storage temperature, and the inherent stability of the protein itself. Generally, the shelf life of liquid form is 6 months at -20°C/-80°C. For the lyophilized form, the shelf life is 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is necessary for multiple use. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
Becaplermin; c sis; FLJ12858; Oncogene SIS; PDGF 2; PDGF B chain; PDGF Beta; PDGF subunit B; PDGF-2; PDGF2; Pdgfb; PDGFB_HUMAN; Platelet derived growth factor 2; Platelet derived growth factor B chain; Platelet derived growth factor beta; Platelet derived growth factor beta polypeptide (oncogene SIS); Platelet derived growth factor beta polypeptide (simian sarcoma viral (v sis) oncogene homolog); Platelet derived growth factor beta polypeptide; Platelet-derived growth factor B chain; Platelet-derived growth factor beta polypeptide; Platelet-derived growth factor subunit B; Proto-oncogene c-Sis; Simian sarcoma viral (v sis) oncogene homolog; SIS; SSV; v sis platelet derived growth factor beta polypeptide
Datasheet & Coa
Please contact us to get it.
Expression Region
82-190aa
Mol. Weight
12.4kDa
Protein Length
Partial
Purity
> 98 % by SDS-PAGE and HPLC analyses.
Research Area
Cancer
Source
E.Coli
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function
Platelet-derived growth factor-BB (PDGF-BB) is a critical growth factor that plays a significant role in regulating embryonic development, cell proliferation, cell migration, survival, and chemotaxis. It acts as a potent mitogen for cells of mesenchymal origin. PDGFB is essential for normal proliferation and recruitment of pericytes and vascular smooth muscle cells in various tissues, including the central nervous system, skin, lung, heart, and placenta. It is also required for normal blood vessel development and for the proper development of kidney glomeruli. Furthermore, PDGFB plays a vital role in wound healing. Its signaling is modulated through the formation of heterodimers with PDGFA.
Gene References Into Functions
  1. Two potential susceptibility loci for prostate cancer risk were identified, located in PDGFB at 22q13.1 PMID: 29168174
  2. Treating HepG2 cells with hepatotoxicants led to a significant increase in mRNA expression of platelet-derived growth factor BB (PDGF-BB) and transforming growth factor beta (TGFbeta). PMID: 29558627
  3. The morphology and immunophenotype of all 6 cases were similar to those with the canonical COL1A1-PDGFB fusion. None of the cases showed fibrosarcomatous transformation. This study highlights that the COL6A3-PDGFD fusion product is rare in dermatofibrosarcoma protuberans and is associated with a potential predilection for breast cancer. PMID: 30014607
  4. PDGF-B expression was detected in ovarian tumor microvessels in 72% of cases. High expression of PDGF in pericapillary cells was strongly associated with high expression of this marker in cancer cells. Significant correlations between PDGF-B and nestin expression in malignant tumor microvessels were also observed. PMID: 28397199
  5. SOX7 transcription factor mediates PDGF-BB-induced IL-33 expression. PMID: 27150562
  6. High expression of PDGF-BB might be involved in the pathogenesis of Graves' disease. CCR2-positive macrophages may induce the expression of PDGF-BB through HIF-1alpha signaling. PMID: 29319128
  7. rhPDGF-BB promoted the proliferation of hADSCs via the miR-363/PI3K/Akt pathway, suggesting that rhPDGF-BB combined with ADSCs could treat Achilles tendinitis through the miR-363/PI3K/Akt pathway. PMID: 28766166
  8. This study identifies mbPDGF-BB in cell-derived extracellular vesicles as a relevant mediator of diabetes-associated vascular smooth muscle cell resistance to apoptosis. PMID: 29386225
  9. This review highlights PDGFB as one of the common genes involved in brain calcification. PMID: 28162874
  10. Dermatofibrosarcoma protuberans (DFSP), a rare skin tumor, is characterized by the translocation of the PDGFB gene to the collagen 1A1 gene. PMID: 28940884
  11. Transglutaminase type 2 affects cell migration through post-translational modification of PDGF-BB. PMID: 27633721
  12. PDGF-B rs1800818 polymorphism might play a role in mediating susceptibility to severe fever with thrombocytopenia syndrome in Chinese individuals. PMID: 27147565
  13. PDAP-1 acts as an effector of PDGF signaling in glioma cells PMID: 27448842
  14. High PDGFB expression is associated with gastric cancer. PMID: 28423550
  15. Regenerating and necrotic muscle fibers in muscle biopsy samples from Duchenne muscular dystrophy patients expressed PDGF-BB. PMID: 28618254
  16. Elevated PDGFB expression was observed in 29% of patients with papillary renal cell carcinoma. PMID: 27989785
  17. High-throughput affinity plasma proteomic profiling is a valuable research approach to identify potential candidate biomarkers for thrombosis-related disorders. Our study suggests a novel association of PDGFB plasma levels with venous thromboembolism. PMID: 27742707
  18. High PDGFB expression is linked to glioma. PMID: 26951930
  19. A cell-autonomous positive-signaling circuit is associated with the PDGF-NO-ID4-regulatory axis in glioblastoma cells. PMID: 28327358
  20. SphK1 is regulated by PDGF-BB in pulmonary artery smooth muscle cells via the transcription factor Egr-1, promoting cell proliferation. PMID: 27099350
  21. PDGF-BB regulates the proliferation and differentiation of human melanocytes in a differentiation-stage-specific manner. PMID: 27289338
  22. This study indicates that the expression of PDGFB is significantly downregulated in keloid fibroblasts compared to normal human fibroblasts PMID: 27465069
  23. This research demonstrated that knockdown of SCARA5 inhibits PDGFBB-induced HASMC proliferation and migration through suppression of the PDGF signaling pathway. PMID: 27035566
  24. The simultaneous action of PDGF-B/PDGFRbeta and VEGF165b on the same type of receptor may explain the resistance to antiangiogenic therapy, which depends on the degree of modulation of PDGFRbeta phosphorylation. PMID: 27127135
  25. The expression of angiogenesis markers VEGF-A, VEGFR, PDGFbetabeta, PDGFR, CCND1, and CA9 was assessed by immunohistochemistry and correlated with overall survival and progression-free survival in patients with renal cell carcinoma undergoing therapy with Sunitinib. However, no correlation was found between expression of angiogenesis markers and clinical outcome. PMID: 28011500
  26. More than 65% of cases exhibited PDGF-BB mRNA amplification, confirming immunohistochemical results. This validates PDGF-BB as a potential therapeutic and prognostic tool for assessing the aggressiveness of ovarian cancer. PMID: 27807074
  27. Case Report: congenital atrophic dermatofibrosarcoma protuberans with COL1A1-PDGFB rearrangement. PMID: 26932148
  28. Placental endothelial cell-derived PDGF-BB recruits human placental multipotent mesenchymal stromal cells involved in vascular development via PDGFR-beta/STAT3 activation PMID: 26353894
  29. These findings suggest that PDGF-B signaling might play a role in endothelial and cardiomyocyte recovery from ischemia-reperfusion injury after heart transplantation. PMID: 26371596
  30. Lessons from SLC20A2, PDGFB, and PDGFRB mutation carriers. Three causative genes have been identified: SLC20A2, PDGFRB, and, more recently, PDGFB. The associated phenotype of PDGFB mutations has not yet been extensively studied. PMID: 26129893
  31. Loss-of-function mutations in PDGFB or PDGFRB cause Primary Familial Brain Calcification. PMID: 26599395
  32. A three-factor model revealed significant gene-gene interaction for PDGFB +286A>G, PDGFB +1135A>C, and HER2 Ile165Val SNPs with gallbladder cancer (GBC). Protein-protein interaction showed a significant association of PDGFB and HER2 with the EGFR receptor signaling pathway. PMID: 26320430
  33. Regulation of Hyaluronan (HA) Metabolism Mediated by HYBID (Hyaluronan-binding Protein Involved in HA Depolymerization, KIAA1199) and HA Synthases in Growth Factor-stimulated Fibroblasts. PMID: 26518873
  34. Electron microscopy structure of PDGFRB [a full-length human platelet-derived growth factor receptor], in complex with its ligand PDGF-B. PMID: 26463591
  35. TM expression in corneal epithelium was modulated during the corneal wound healing process and may be regulated by PDGF-BB. Additionally, rTMD23 has therapeutic potential in corneal injury. PMID: 25816372
  36. Our results demonstrated that PDGF-B promotes tumor growth and progression in clear cell renal cell carcinoma. PMID: 25766258
  37. PDGFB and IL18R1 are plausible candidates for studying the pathophysiology of these disorders in the context of TLR4 activation. PMID: 25327457
  38. TAZ promotes neuroblastoma cell proliferation and tumorigenicity through upregulation of the expression of PDGF-beta genes. PMID: 25940705
  39. Phloretin inhibits PDGF-BB-induced thoracic aorta smooth muscle cell proliferation and migration. PMID: 25945863
  40. COL1A1-PDGFbeta translocation is specific to dermatofibrosarcoma protuberans. Platelet-derived growth factor may have acted in an autocrine manner to cause cell division, which might have led to the development of dermatofibrosarcoma protuberans. PMID: 25924890
  41. The current study demonstrated, for the first time, that ligustrazine downregulated PDGF-BB-induced VSMC proliferation and migration partly, at least, through inhibiting the activation of the ERK and P38 MAPK signaling. PMID: 25738255
  42. Cytoplasmic expression of VEGF, VEGFR2, PDGF-B, and PDGFR-beta in RCC tumor cells varies across different pathologic stages and cell types. Notably, VEGF and PDGF-B expression are higher in papillary than in clear cell renal cell carcinoma. PMID: 25550804
  43. COL1A1-PDGFB is a useful and accurate tool for diagnosing DFSP [Dermatofibrosarcoma protuberans] in Koreans. PMID: 25683993
  44. Significant diurnal variations in platelet counts and TGF-b1 and PDGF-BB levels were not observed in platelet-rich plasma. PMID: 24878758
  45. This report found lowered PDGF-BB levels in acute pancreatitis and increased PDGF-BB levels in chronic pancreatitis. PMID: 25278706
  46. rhPDGF-BB delivery on a collagen scaffold enhanced cellular proliferation and angiogenesis during the early phase of healing after rotator cuff repair. PMID: 25349036
  47. Mutations in PDGFB cause primary familial brain calcifications. PMID: 25212438
  48. Gain of PDGFB is associated with response to therapy in metastatic renal cell carcinoma. PMID: 24524969
  49. These results suggest that PDGF-BB promotes pulmonary artery smooth muscle cells proliferation and survival, likely mediated via the JNK pathway. PMID: 24804810
  50. PDGFB hypomethylation is a favorable prognostic biomarker in primary myelofibrosis. PMID: 25498506

Show More

Hide All

Database Links

HGNC: 8800

OMIM: 190040

KEGG: hsa:5155

STRING: 9606.ENSP00000330382

UniGene: Hs.1976

Involvement In Disease
Basal ganglia calcification, idiopathic, 5 (IBGC5)
Protein Families
PDGF/VEGF growth factor family
Subcellular Location
Secreted. Note=Released by platelets upon wounding.
Tissue Specificity
Expressed at high levels in the heart, brain (sustantia nigra), placenta and fetal kidney. Expressed at moderate levels in the brain (hippocampus), skeletal muscle, kidney and lung.

Q&A

What is the molecular structure of recombinant human PDGF-BB?

Recombinant human PDGF-BB is a homodimeric protein composed of two B chains encoded by the PDGFB gene. Under reducing conditions, it appears as a single band at approximately 13 kDa, while under non-reducing conditions, it appears as a band at 28 kDa, representing its dimeric structure. This structural configuration is critical for its biological activity, as the dimerization enables proper receptor binding and subsequent signal transduction. The protein exhibits specific folding patterns that are essential for maintaining its biological activity when used in experimental settings .

How does PDGF-BB differ from other PDGF isoforms?

PDGF exists in three dimeric isoforms: PDGF-AA, PDGF-AB, and PDGF-BB, each formed by the combination of A and B chains. These isoforms are differentially expressed across various cell types and exhibit distinct binding affinities to the two PDGF receptors (alpha and beta). PDGF-BB is unique in that it can bind to both alpha and beta receptors with high affinity, whereas PDGF-AA binds only to alpha receptors. This broader receptor interaction profile gives PDGF-BB a wider range of biological activities and target cells compared to other isoforms. Additionally, PDGF-BB demonstrates more potent mitogenic activity in certain contexts, making it particularly valuable for research applications requiring robust cellular responses .

What are the optimal storage and handling conditions for maintaining PDGF-BB activity?

For optimal preservation of PDGF-BB activity, the recombinant protein should be stored at -20°C to -80°C, preferably in small aliquots to avoid repeated freeze-thaw cycles. When reconstituting lyophilized PDGF-BB, it's recommended to use sterile buffer solutions such as PBS containing a carrier protein (0.1-1% BSA or HSA) to prevent adhesion to tubes and maintain stability. For working solutions, PDGF-BB should be kept at 2-8°C for immediate use and discarded after 24 hours. Researchers should avoid vortexing as this can cause protein denaturation; instead, gentle mixing by pipetting or flicking is preferred. Activity testing before critical experiments is advisable, particularly if the protein has been stored for extended periods, as activity can diminish over time even under optimal storage conditions .

What is the typical biological activity of recombinant human PDGF-BB in cellular assays?

Recombinant human PDGF-BB demonstrates potent mitogenic activity in various cell types, particularly fibroblasts. In standardized proliferation assays using the NR6R-3T3 mouse fibroblast cell line, PDGF-BB stimulates cell proliferation in a dose-dependent manner with an ED50 (effective dose for 50% response) typically ranging from 1.5-6 ng/mL. This dose-response relationship forms a sigmoidal curve, with detection of activity possible at concentrations as low as 0.5 ng/mL and reaching plateau effects at approximately 10-20 ng/mL. The protein also exhibits chemotactic activity for fibroblasts, smooth muscle cells, neutrophils, and mononuclear cells, with chemotactic responses observable at concentrations 5-10 fold lower than those required for mitogenic effects .

How does mechanical stress regulate PDGF-BB expression in endothelial cells?

Mechanical forces, particularly cyclic strain, significantly influence PDGF-BB expression in endothelial cells through transcriptional regulation. When bovine aortic endothelial cells are subjected to 10% average strain (but not 6%), a 2.6-fold increase in PDGF-B steady-state mRNA and immunoreactive protein is observed. Nuclear runoff transcription assays confirm this is due to increased transcription rather than mRNA stability. The mechanotransduction pathway involves strain-responsive elements in the PDGF-B promoter region, particularly between positions -313 and -153, which account for approximately 55% of the strain-induced activity. While a shear stress response element (SSRE) at position -125 binds nuclear proteins from strain-exposed cells, mutation analysis reveals it is not essential for strain-induced expression, suggesting multiple regulatory elements contribute to the mechanosensitive response .

What role does PDGF-BB play in tumor microenvironment remodeling?

PDGF-BB serves as a critical mediator in tumor microenvironment remodeling, particularly in extracellular matrix (ECM) formation and cancer-associated fibroblast recruitment. Studies using platelet-specific PDGFB knockout mice demonstrated that platelet-derived PDGF-BB significantly contributes to ECM composition in tumors. Ablation of platelet-specific PDGFB results in reduced collagen 1 and fibronectin deposition in the tumor microenvironment, with a significant decrease in collagen 1 transcription. Interestingly, while primary tumor growth remains unaffected by the absence of platelet-derived PDGF-BB, the number of circulating tumor cells and liver metastases increases due to compromised vascular barrier integrity. This suggests a dual role where PDGF-BB contributes to ECM stability that may restrict tumor cell dissemination while simultaneously promoting a microenvironment that supports tumor growth through fibroblast recruitment and activation .

How can researchers optimize PDGF-BB dosage in experimental settings?

Optimizing PDGF-BB dosage requires a systematic approach considering both concentration and expression systems. First, establish a dose-response curve using a range of concentrations (typically 0.1-100 ng/mL) with your specific cell type, measuring relevant endpoints (proliferation, migration, gene expression). In gene therapy approaches, regulation of PDGF-BB expression can be achieved by selecting promoters of appropriate strength. Research has demonstrated that using relatively weak physiologic promoters (like the phosphoglycerate kinase or PGK promoter) can yield significant biological effects without causing systemic elevation of PDGF-BB levels. For example, in bone formation studies, the PGK promoter driving PDGFB expression produced optimal bone formation without adverse effects such as osteomalacia or secondary hyperparathyroidism. When comparing promoters, evaluate their relative strength using reporter genes like GFP before proceeding with PDGFB expression constructs .

What methods are recommended for measuring PDGF-BB-induced cellular responses?

For comprehensive assessment of PDGF-BB-induced cellular responses, a multi-parameter approach is recommended. Proliferation can be measured using BrdU incorporation, Ki-67 immunostaining, or metabolic assays such as MTT or Alamar Blue. Cell migration should be assessed through wound healing assays, Boyden chamber assays, or time-lapse microscopy to capture dynamic responses. For signal transduction analysis, Western blotting for phosphorylated downstream effectors (Akt, ERK, STAT) at multiple time points (5 min to 24 hours) provides kinetic information. RNA-seq or targeted qPCR arrays can identify transcriptional changes at 4-24 hours post-stimulation. In tissue-specific contexts, such as bone formation, advanced imaging techniques including micro-computed tomography (μCT) enable quantification of structural parameters like trabecular bone volume, thickness, and connectivity density. Functional outcomes should also be measured; for example, in bone studies, three-point bending mechanical testing can determine if PDGF-BB treatment increases load-to-failure force and stiffness .

What are the key considerations when designing gene therapy experiments using PDGFB?

When designing gene therapy experiments using PDGFB, several critical factors must be considered. First, promoter selection significantly impacts therapeutic outcomes—strong promoters like SFFV can lead to excessive PDGF-BB production causing adverse effects, while weaker promoters like PGK or truncated PGK (P200) can provide therapeutic benefits without complications. Second, delivery method optimization is essential; studies have successfully used lentiviral vectors to transduce Sca1+ stem cells, achieving therapeutic effects with MOIs as low as 0.1. Third, establish appropriate controls, including empty vector and irrelevant protein expression (e.g., GFP) to distinguish specific PDGFB effects from vector-related responses. Fourth, comprehensive safety assessment must include monitoring for ectopic tissue formation, hyperplasia, and effects on major organs (particularly spleen enlargement due to extramedullary hematopoiesis when bone marrow space is compromised). Finally, consider the preconditioning regimen; while irradiation facilitates engraftment in research settings, alternative non-irradiation protocols would be necessary for clinical translation in non-life-threatening conditions .

How does PDGF-BB-based stem cell gene therapy affect bone strength and architecture?

PDGF-BB-based stem cell gene therapy significantly enhances bone strength and architecture through multiple mechanisms. Studies using lentiviral vectors expressing PDGFB under the control of the PGK promoter demonstrated a 45% increase in maximum load-to-failure in the midshaft of femurs and a 46% increase in stiffness compared to controls. These mechanical improvements correlate directly with both cortical bone mass and trabecular bone mass at the midshaft. The therapy produces a remarkable 20-fold increase in trabecular connectivity and substantial reduction in cortical porosity (40% decrease), both critical parameters for bone mechanical performance. Microcomputed tomography analysis reveals that treated bones develop thick, well-connected trabeculae throughout the marrow cavity at both metaphysis and midshaft regions, with the new bone formation occurring specifically on the endosteal surface rather than the periosteal surface. These architectural changes result in mechanically functional, high-quality bone tissue that maintains elevated alkaline phosphatase levels over extended periods (3+ months), suggesting sustained anabolic effects without the resistance to therapy often seen in conventional osteoporosis treatments .

What are the potential limitations and adverse effects of PDGF-BB therapy in research models?

While PDGF-BB therapy shows promising results, researchers must carefully consider several limitations and potential adverse effects. At high systemic concentrations, PDGF-BB can cause osteomalacia and secondary hyperparathyroidism, highlighting the importance of dose optimization. When using gene therapy approaches, spleen enlargement has been observed in mice receiving PGK-PDGFB treatment, likely due to increased extramedullary hematopoiesis when substantial marrow cavity space becomes occupied by newly formed bone. The preconditioning requirement of irradiation for stem cell engraftment presents another limitation, though alternative non-irradiation protocols could potentially be developed. Long-term studies are needed to assess potential resistance to therapy after prolonged treatment, similar to what occurs with other anabolic therapies. Additionally, the effectiveness of PDGF-BB therapy has primarily been demonstrated in normal bone mass models rather than osteoporotic models, leaving uncertainty about translational efficacy in disease states. Finally, the current rigid promoter system for dosage control lacks the flexibility needed for personalized medicine applications, suggesting a need for transcriptional regulation systems that allow more precise PDGFB dose adjustment .

How does the absence of platelet-derived PDGF-BB affect tumor metastasis and extracellular matrix composition?

The absence of platelet-derived PDGF-BB creates a complex dual effect on tumor progression and metastasis through alterations in extracellular matrix composition. Studies using platelet-specific PDGFB knockout mice revealed that while primary tumor growth remains unaffected, the number of circulating tumor cells and liver metastases significantly increases. This paradoxical effect stems from compromised vascular barrier integrity in the absence of platelet-derived PDGF-BB. Immunohistochemical analysis demonstrated reduced collagen 1 and fibronectin deposition in tumors from knockout mice, with collagen fibers appearing distorted and less elongated. Quantitative PCR confirmed a significant reduction in collagen 1 transcription, though fibronectin gene expression remained unchanged, suggesting post-transcriptional regulation for the latter. These findings indicate that platelet-derived PDGF-BB contributes substantially to extracellular matrix formation in the tumor microenvironment, creating a structure that may physically restrict tumor cell dissemination despite potentially supporting tumor growth through other mechanisms. This nuanced relationship between PDGF-BB, extracellular matrix composition, and metastatic potential highlights the importance of context-specific consideration when targeting PDGF-BB therapeutically in cancer .

What analytical methods should be used to verify recombinant PDGF-BB quality and activity?

Comprehensive quality assessment of recombinant PDGF-BB requires multiple analytical methods targeting different characteristics. Protein purity should be verified through SDS-PAGE under both reducing and non-reducing conditions, with silver staining revealing single bands at approximately 13 kDa and 28 kDa, respectively. Mass spectrometry provides precise molecular weight confirmation and can detect post-translational modifications. Biological activity must be assessed using standardized cell-based assays, such as proliferation assays with NR6R-3T3 mouse fibroblast cells, establishing a dose-response curve to determine the ED50 (typically 1.5-6 ng/mL for high-quality preparations). Western blot analysis using specific antibodies confirms immunoreactivity and can detect degradation products. Endotoxin testing using the Limulus Amebocyte Lysate (LAL) assay is essential, particularly for preparations intended for in vivo research, with levels below 1 EU/μg protein considered acceptable. For GMP-grade preparations, additional testing including sterility, mycoplasma, and virus testing may be necessary. Finally, stability studies under various storage conditions help establish optimal handling protocols to maintain activity throughout the experimental timeline .

How can researchers distinguish between endogenous and exogenous PDGF-BB in experimental systems?

Distinguishing between endogenous and exogenous PDGF-BB in experimental systems requires strategic experimental design and specialized analytical techniques. One effective approach is to use epitope-tagged recombinant PDGF-BB (e.g., His-tag, FLAG-tag, or HA-tag) that can be selectively detected using tag-specific antibodies via Western blot, ELISA, or immunocytochemistry. Species-specific antibodies can differentiate between human recombinant PDGF-BB and endogenous animal PDGF-BB in xenogeneic systems (e.g., human PDGF-BB in mouse cells). For gene therapy approaches, incorporating unique untranslated regions or codon-optimized sequences allows selective detection of transcripts using specific PCR primers. Temporal analysis immediately following exogenous administration can capture the acute response before substantial endogenous production occurs. In systems requiring long-term distinction, CRISPR/Cas9 gene editing to create cells with PDGFB knockout followed by complementation with detectable exogenous PDGF-BB provides a clean background. Finally, absolute quantification using mass spectrometry with isotope-labeled standards can provide precise measurements of both endogenous and exogenous protein concentrations simultaneously .

What are the considerations for scaling up PDGF-BB production for advanced preclinical studies?

Scaling up PDGF-BB production for advanced preclinical studies requires careful consideration of expression systems, purification strategies, and quality control processes. Mammalian expression systems (CHO or HEK293 cells) are preferred over bacterial systems as they produce properly folded and post-translationally modified PDGF-BB with higher biological activity. For large-scale production, stable cell lines with inducible promoters offer better consistency than transient transfection methods. Bioreactor cultivation using perfusion or fed-batch processes optimizes yields while maintaining protein quality. Purification should employ a multi-step approach, typically beginning with affinity chromatography (heparin-Sepharose exploits PDGF-BB's heparin-binding properties), followed by ion-exchange and size-exclusion chromatography to achieve >95% purity. Critical quality attributes to monitor throughout scaling include specific activity (bioassays), aggregation state (analytical ultracentrifugation or dynamic light scattering), and glycosylation patterns (lectin binding assays or mass spectrometry). Batch-to-batch consistency must be verified with comprehensive comparability studies, and stability-indicating assays should be developed to establish appropriate storage conditions and shelf-life. Finally, the entire production process should be documented following good manufacturing practice (GMP) guidelines if the material is intended for advanced preclinical studies leading toward clinical applications .

How does PDGF-BB compare to other growth factors in bone regeneration research?

In bone regeneration research, PDGF-BB demonstrates distinct advantages and mechanisms compared to other commonly studied growth factors. Unlike Bone Morphogenetic Proteins (BMPs) which primarily induce direct osteoblast differentiation, PDGF-BB functions predominantly as a chemotactic and mitogenic factor for mesenchymal stem cells, osteoprogenitors, and fibroblasts. This recruitment and expansion of progenitor populations creates a cellular reservoir for subsequent differentiation. Compared to Fibroblast Growth Factor 2 (FGF2), PDGF-BB circulates at concentrations approximately 100-fold higher, potentially offering a better safety profile. While Vascular Endothelial Growth Factor (VEGF) primarily targets endothelial cells for angiogenesis, PDGF-BB affects both the vascular and osteoblastic lineages, promoting coordinated vascularization and bone formation. In comparative studies, PDGF-BB uniquely produces substantial increases in trabecular connectivity (20-fold) and reductions in cortical porosity, architectural features critical for mechanical strength that are not consistently achieved with other growth factors. Additionally, PDGF-BB-based therapies have demonstrated sustained anabolic effects without the resistance phenomenon observed after prolonged BMP treatment, suggesting different regulatory mechanisms governing long-term response .

What explanations exist for contradictory findings in PDGF-BB research across different experimental models?

Contradictory findings in PDGF-BB research can be attributed to several methodological and biological factors. Dosage variations significantly impact outcomes—high systemic concentrations can cause osteomalacia and secondary hyperparathyroidism, while optimal lower doses produce beneficial bone formation without adverse effects. The delivery method (bolus injection versus sustained release or gene therapy) dramatically alters the temporal profile of PDGF-BB exposure and subsequent cellular responses. Species differences in PDGF receptor distribution and downstream signaling pathways may explain contradictory results between rodent and larger animal models. The disease state of the experimental model is crucial; normal bone mass models may respond differently than osteoporotic or inflammatory models due to altered receptor expression and cellular composition. Age-related differences in stem cell populations and receptor responsiveness further complicate cross-study comparisons. The microenvironmental context, particularly mechanical loading conditions, interacts with PDGF-BB signaling, potentially explaining site-specific variations in outcomes. Finally, the presence of cofactors or inhibitors in different experimental systems may modulate PDGF-BB activity—for instance, thrombospondin expression is modified by PDGF-BB but may in turn regulate its activity, creating complex feedback systems that vary across experimental contexts .

What emerging technologies are advancing PDGF-BB research and applications?

Emerging technologies are revolutionizing PDGF-BB research across multiple fronts. Advanced biomaterial delivery systems, including injectable hydrogels with tunable degradation profiles and 3D-printed scaffolds with precise spatial control, enable sustained and localized PDGF-BB delivery with minimal systemic exposure. CRISPR/Cas9 gene editing allows precise modification of PDGF-BB sequence, its receptors, or downstream signaling components to elucidate specific molecular mechanisms. Single-cell RNA sequencing combined with spatial transcriptomics provides unprecedented insights into cell-specific responses to PDGF-BB within heterogeneous tissues, revealing previously unrecognized target cell populations and response patterns. Optogenetic and chemogenetic approaches enable temporal control of PDGF-BB signaling, allowing researchers to dissect acute versus sustained effects. Computational modeling using machine learning algorithms can predict PDGF-BB distribution, receptor binding dynamics, and downstream effects across diverse tissues and disease states. Finally, organoid and microphysiological systems ("organs-on-chips") bridge the gap between traditional cell culture and animal models, providing controlled yet physiologically relevant environments to study PDGF-BB effects on tissue development, homeostasis, and pathology .

What are the key unresolved questions regarding PDGF-BB's mechanism of action in tissue regeneration?

Despite extensive research, several key questions regarding PDGF-BB's mechanism of action in tissue regeneration remain unresolved. First, the temporal dynamics of PDGF-BB signaling in coordinating the sequential phases of regeneration (inflammation, proliferation, and remodeling) are poorly understood, particularly how initial PDGF-BB exposure programs long-term cellular behavior. Second, the precise identity and fate of PDGF-BB-responsive stem/progenitor cells in various tissues require clarification—whether PDGF-BB primarily expands existing progenitor pools or reprograms committed cells toward progenitor-like states remains debated. Third, the mechanisms underlying PDGF-BB's apparent resistance to therapeutic desensitization (unlike other growth factors where efficacy diminishes over time) need investigation. Fourth, the interaction between PDGF-BB signaling and mechanical forces in driving tissue-specific architectural organization requires further study, particularly how PDGF-BB might modulate mechanotransduction pathways. Fifth, the differential effects of matrix-bound versus soluble PDGF-BB on cellular behavior and tissue organization remain to be fully elucidated. Finally, the potential role of PDGF-BB in reprogramming the immune microenvironment during regeneration represents an emerging area requiring systematic investigation, particularly how it might shift macrophage polarization from pro-inflammatory to pro-regenerative phenotypes .

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.