HGF (32-285) Human is a recombinant protein fragment corresponding to amino acids 32-285 of the full-length human Hepatocyte Growth Factor (also known as Hepapoietin A or Scatter Factor) . This particular fragment encompasses the N-terminal domain and portions of the first kringle domain of the complete HGF protein, representing approximately one-third of the full-length molecule . The protein is primarily used in research settings to study specific aspects of HGF biology and signaling without the complexity of the complete protein structure.
HGF was originally discovered in 1984 as a potent mitogen for hepatocytes in primary culture and was later identified as a cell motility factor (scatter factor) for epithelial cells . It has since been recognized as a pleiotropic growth factor involved in embryogenesis, tissue regeneration, wound healing, and cancer progression . The specific fragment HGF (32-285) represents an important functional region that contributes to these biological activities.
The HGF (32-285) Human fragment has the following physical characteristics:
HGF (32-285) Human is typically produced through recombinant expression systems, most commonly in Escherichia coli (E. coli) . This bacterial expression system allows for cost-effective production of the protein fragment for research purposes. The recombinant production typically involves:
Cloning of the cDNA encoding amino acids 32-285 of human HGF
Expression in E. coli under controlled conditions
Purification using affinity chromatography, often facilitated by fusion tags
Quality control testing including SDS-PAGE for purity assessment
The HGF (32-285) fragment contains regions important for binding to the MET receptor, which is the only known receptor for HGF . The binding of HGF to MET occurs through multiple interfaces:
The N-terminal domain contributes to high-affinity binding to MET
The first kringle domain (K1) interacts with the Sema domain of MET
While full-length HGF demonstrates a wide range of biological activities, the HGF (32-285) fragment shares some but not all of these functions. Based on studies of similar fragments like NK1, HGF (32-285) likely exhibits:
Partial agonistic activity on the MET receptor, though typically weaker than full-length HGF
Ability to induce some cell migration and morphogenesis, albeit at reduced potency
Potential competitive effects against full-length HGF when both are present
Research on NK1 variants (structurally similar to HGF 32-285) has shown that dimerization plays a crucial role in receptor activation . The ability of HGF (32-285) to dimerize would significantly impact its biological activity.
HGF (32-285) Human serves as a valuable tool in fundamental research into HGF-MET signaling:
Structure-function studies to understand the role of specific domains in HGF activity
Comparison studies with full-length HGF and other fragments to delineate domain-specific contributions to signaling
Development of novel MET agonists and antagonists for therapeutic applications
Based on the regenerative properties of full-length HGF, the (32-285) fragment has potential applications in:
Given the involvement of HGF-MET signaling in cancer progression, HGF (32-285) serves as a tool in oncology research:
Development of potential MET antagonists to inhibit cancer metastasis
Investigation of epithelial-mesenchymal transition (EMT) in cancer cells
Full-length HGF exhibits the most potent and diverse biological activities, including:
In comparison, HGF (32-285) and similar fragments typically show:
Reduced potency in MET activation compared to full-length HGF
Retained ability to bind MET but with altered signaling dynamics
Potential antagonistic effects against full-length HGF when both are present
The study of HGF (32-285) Human continues to evolve with several promising research directions:
Engineering more stable variants with enhanced agonist properties for regenerative medicine applications
Developing multimeric forms of the fragment to enhance receptor activation, similar to the K1B/S complex approach described in research
Utilizing the fragment as a starting point for designing MET-targeted therapeutics
Further exploring the structure-function relationships within this region to better understand HGF-MET signaling mechanisms
HGF (32-285) Human is a recombinant protein fragment containing amino acids 32-285 of the full-length human Hepatocyte Growth Factor. It is typically produced in Escherichia coli as a single, non-glycosylated polypeptide chain with a molecular mass of approximately 29.8 kDa . This protein represents a functional domain of HGF that maintains many of the biological activities of the complete protein while offering advantages in terms of production and experimental manipulation. The protein is also known by several synonyms including Scatter Factor (SF), Hepatopoietin (HPTA), HGFB, F-TCF, and DFNB39 .
HGF (32-285) functions as a potent mitogen for mature parenchymal hepatocyte cells and acts as a growth factor for a broad spectrum of tissues and cell types . In research contexts, it serves as an important tool for studying cellular processes including proliferation, motility, and differentiation. HGF (32-285) has been demonstrated to exert significant effects on human mesenchymal stem cells (hMSCs), where it promotes osteogenic differentiation through the transcription of key markers such as osteocalcin, osterix, and osteoprotegerin . Additionally, it synergizes with Interleukin-3 and GM-CSF to stimulate colony formation of hematopoietic progenitor cells in vitro, making it valuable for hematopoiesis research .
The HGF (32-285) fragment contains 255 amino acids with the sequence: MQRKRRNTIH EFKKSAKTTL IKIDPALKIK TKKVNTADQC ANRCTRNKGL PFTCKAFVFD KARKQCLWFP FNSMSSGVKK EFGHEFDLYE NKDYIRNCII GKGRSYKGTV SITKSGIKCQ PWSSMIPHEH SYRGKDLQEN YCRNPRGEEG GPWCFTSNPE VRYEVCDIPQ CSEVECMTCN GESYRGLMDH TESGKICQRW DHQTPHRHKF LPERYPDKGF DDNYCRNPDG QPRPWCYTLD PHTRWEYCAI KTCET . This segment represents the N-terminal portion of the complete HGF protein (P14210), which contains important domains for receptor binding and biological activity.
For optimal stability and activity maintenance, HGF (32-285) should be stored at 4°C if the entire vial will be used within 2-4 weeks . For longer periods, the protein should be stored frozen at -20°C. To minimize activity loss during long-term storage, it is recommended to add a carrier protein such as 0.1% Human Serum Albumin (HSA) or Bovine Serum Albumin (BSA) . Multiple freeze-thaw cycles should be strictly avoided as they can lead to significant degradation of protein structure and function. For research requiring consistent protein activity over extended periods, it is advisable to prepare aliquots upon initial thawing to minimize freeze-thaw events.
HGF (32-285) is typically formulated as a protein solution (1mg/ml) containing 20mM Tris-HCl buffer (pH 8.0), 10% glycerol, and 0.4M Urea . For cell culture applications, the protein should be diluted to the appropriate working concentration in sterile cell culture medium immediately before use. Based on experimental protocols from the literature, an optimal working concentration of 40 ng/mL has been established for many cell-based assays, particularly when working with human mesenchymal stem cells . When planning experiments, consider performing a dose-response assessment (e.g., 10, 20, 40, and 100 ng/mL) to determine the optimal concentration for your specific cell type and experimental endpoint.
Several approaches can be employed to confirm the biological activity of HGF (32-285):
Phosphorylation assays: HGF activates multiple signaling pathways including rapid phosphorylation of p38, ERK, and AKT/PI3K. Western blot analysis using phospho-specific antibodies can detect these activation events within minutes of treatment .
Proliferation assays: HGF (32-285) promotes cell proliferation in responsive cell types. A simple hemocytometer-based cell counting method after 5 days of treatment can confirm activity .
Differentiation markers: In mesenchymal stem cells, HGF (32-285) increases expression of osteogenic markers. RT-qPCR analysis of genes such as osteocalcin, osterix, and osteoprotegerin can validate functional activity .
Functional blocking: Using HGF-neutralizing antibodies (e.g., MAB294 at 100-300 ng/mL) or c-Met inhibitors (e.g., PHA665752) alongside HGF treatment provides important controls to confirm specificity of observed effects .
When encountering contradictory results across different cell types, several systematic approaches can help resolve discrepancies:
Receptor expression profiling: Quantify c-Met receptor expression levels across cell types using flow cytometry or Western blotting, as differential receptor densities can significantly impact response magnitude.
Pathway analysis: Implement comprehensive phosphoproteomic analysis to map activated signaling networks in different cell types, as downstream pathway availability varies between cellular contexts.
Temporal dynamics assessment: Conduct time-course experiments (0-72 hours) measuring both immediate (signaling) and delayed (transcriptional) responses, as timing differences often explain apparent contradictions.
Co-factor identification: Analyze culture medium composition for growth factors that might synergize with or antagonize HGF signaling, particularly when using serum-containing media which contains variable growth factor concentrations.
Genetic background characterization: Sequence key components of the HGF signaling pathway to identify polymorphisms that might affect response characteristics between donor-derived primary cells.
Distinguishing direct from indirect effects in complex models requires sophisticated experimental designs:
Cell-specific receptor knockout: Implement conditional c-Met receptor deletion in specific cell populations using CRISPR/Cas9 or siRNA approaches.
Conditioned media experiments: Compare direct HGF treatment with conditioned media from HGF-treated cells to identify secondary paracrine mediators.
Single-cell transcriptomics: Apply scRNA-seq to identify cell type-specific responses within heterogeneous populations, revealing which cells respond directly versus those affected through paracrine mechanisms.
Pharmacological pathway dissection: Use selective inhibitors of potential mediator pathways in combination with HGF treatment to block indirect effects while preserving direct signaling.
Temporal segregation: Implement pulse-chase experiments with labeled HGF followed by washout periods to separate immediate direct effects from delayed indirect responses.
A robust experimental design with HGF (32-285) should include the following controls:
Control Type | Implementation | Purpose |
---|---|---|
Vehicle control | Buffer components without HGF | Accounts for effects of carrier solution |
Receptor blocking | c-Met inhibitor (PHA665752) + HGF | Confirms specificity to c-Met receptor |
Neutralizing antibody | Anti-HGF antibody (e.g., MAB294, 100-300 ng/mL) + HGF | Validates HGF-specific effects |
Positive control | Known HGF-responsive cell line | Confirms activity of the protein lot |
Dose-response | Multiple concentrations (10-100 ng/mL) | Establishes optimal working concentration |
Time course | Measurements at multiple timepoints | Determines temporal dynamics of response |
Pathway inhibitors | Specific inhibitors for p38, ERK, or PI3K | Identifies pathway dependencies |
siRNA knockdown | Targeting c-Met or downstream mediators | Confirms molecular mechanisms |
Concentration optimization should be approached systematically based on the experimental endpoint:
For immediate signaling events (0-60 minutes), begin with a range of 10-100 ng/mL HGF and measure phosphorylation of key pathway components (p38, ERK, AKT) using Western blotting or ELISA-based methods. The optimal concentration may differ between pathways, and this information should guide subsequent experiments targeting specific mechanisms.
For transcriptional responses (6-48 hours), initial testing should include concentrations from 10-40 ng/mL, with gene expression analysis performed using RT-qPCR for known HGF-responsive genes. Published data indicates that 40 ng/mL is often optimal for transcriptional effects in mesenchymal stem cells .
For long-term phenotypic changes such as differentiation or mineralization, which may require repeated treatments over days to weeks, lower concentrations (20-40 ng/mL) are typically more appropriate to avoid receptor desensitization. Medium changes containing fresh HGF should be performed every 3-4 days to maintain consistent signaling .
Primary cell cultures present inherent variability that can be minimized through several methodological approaches:
Donor pooling: Using pooled cells from multiple donors can reduce donor-specific variability while maintaining primary cell characteristics.
Passage standardization: Limit experiments to specific passage numbers (typically passages 3-7) to avoid senescence-related changes in receptor expression.
Receptor quantification: Implement flow cytometry-based quantification of c-Met receptor expression as a pre-experiment quality control measure.
Serum standardization: Either use serum-free conditions or implement batch-testing of serum lots to select those with minimal impact on baseline HGF signaling.
Synchronized cell cycle: Implement serum starvation (6-12 hours) prior to HGF treatment to synchronize cells in G0/G1, reducing cell cycle-dependent variability in responses.
Internal normalization: Include housekeeping genes or proteins that remain stable during HGF treatment as internal controls for quantitative analyses.
The functional comparison between HGF variants and other growth factors reveals distinct biological profiles:
Growth Factor | Molecular Weight | Receptor | Primary Signaling Pathways | Key Effects on MSCs | Comparative Advantage |
---|---|---|---|---|---|
HGF (32-285) | 29.8 kDa | c-Met | p38 MAPK, ERK, PI3K/AKT | Promotes osteogenic differentiation, enhances migration | Higher stability, better production efficiency |
Full-length HGF | 80-95 kDa | c-Met | p38 MAPK, ERK, PI3K/AKT, STAT3 | Broader spectrum of activity, stronger pro-migration effects | Complete biological activity profile |
FGF-2 | 18 kDa | FGFR1-4 | ERK, PI3K/AKT | Maintains stemness, promotes proliferation | Superior for maintaining undifferentiated state |
BMP-2 | 26 kDa | BMPR-IA/IB | SMAD 1/5/8 | Strong osteogenic induction | More potent for bone formation |
PDGF-BB | 24.3 kDa | PDGFR-α/β | ERK, PI3K, PLCγ | Enhanced proliferation and migration | Superior chemotactic properties |
HGF (32-285) maintains many of the biological activities of full-length HGF while offering advantages in terms of production efficiency and stability. While it effectively activates the p38 MAPK pathway critical for osteogenic differentiation , its effects on STAT3 signaling may be attenuated compared to the full-length protein. This makes HGF (32-285) particularly suited for research focused on osteogenic differentiation and migration, while full-length HGF may be preferred for studies requiring the complete spectrum of biological activities.
Several key biological effects of HGF (32-285) have been consistently observed across diverse experimental systems:
Promotion of cell migration: HGF (32-285) enhances cellular motility in various cell types, including mesenchymal stem cells, epithelial cells, and certain cancer cell lines.
Activation of p38 MAPK signaling: Rapid phosphorylation of p38 MAPK, particularly the α and β isoforms, has been documented in multiple cell types following HGF (32-285) treatment .
Osteogenic differentiation: HGF (32-285) consistently promotes osteogenic differentiation of mesenchymal stem cells, as evidenced by increased expression of osteocalcin, osterix, and osteoprotegerin .
Synergy with hematopoietic factors: HGF (32-285) synergizes with Interleukin-3 and GM-CSF to enhance colony formation of hematopoietic progenitor cells .
c-Met receptor dependent signaling: The biological effects of HGF (32-285) are consistently abolished by c-Met inhibitors or receptor knockdown, confirming the specificity of its mechanism across systems .
These consistent observations provide a solid foundation for researchers to build upon when designing experiments in novel cell types or experimental conditions.
Long-term differentiation studies with HGF (32-285) present several challenges that require specific methodological solutions:
Protein stability: HGF (32-285) may degrade during extended culture periods. Solution: Implement a regular supplementation schedule with fresh protein every 3-4 days rather than relying on a single initial treatment .
Receptor desensitization: Continuous exposure to high concentrations can downregulate c-Met receptor expression. Solution: Use moderate concentrations (20-40 ng/mL) and consider implementing pulsed treatment protocols with recovery periods.
Serum interactions: Components in serum may interact with HGF signaling over time. Solution: Consider defined serum-free media formulations for critical experiments, particularly when comparing conditions.
Paracrine signaling changes: Long-term HGF treatment alters the secretory profile of treated cells. Solution: Implement regular medium changes to prevent accumulation of secondary mediators that might confound results.
Heterogeneous responses: Extended differentiation protocols can lead to population heterogeneity. Solution: Implement single-cell analysis techniques or cell sorting to identify and characterize responder subpopulations.
When encountering unexpected results with HGF (32-285), implementing a systematic troubleshooting approach can identify underlying issues:
Verify protein activity: Conduct a simple phospho-p38 Western blot in a known responsive cell line to confirm the activity of your HGF (32-285) preparation.
Assess receptor expression: Quantify c-Met receptor levels in your experimental cells using flow cytometry or Western blotting, as expression levels vary widely between cell types and culture conditions.
Examine buffer compatibility: The Tris-HCl and urea components in HGF formulation may interact with certain media components. Test dilution in different buffers before adding to cells.
Evaluate batch-to-batch variability: If using a new lot of HGF (32-285), run side-by-side comparisons with previous lots on a standardized assay to identify potential activity differences.
Check for inhibitory factors: Some cell types secrete HGF antagonists or express truncated c-Met receptors that can act as decoys. Analyze culture supernatants for potential interfering molecules.
Consider timing dynamics: HGF responses follow complex temporal patterns. Implement a detailed time-course analysis (minutes to days) to ensure you're not missing the relevant window for your endpoint.
Examine experimental conditions: Cell density, attachment substrate, and oxygen tension can all significantly impact HGF responsiveness. Systematically vary these parameters to identify optimal conditions for your system.
Advanced analytical approaches provide deeper insights into HGF (32-285) mechanisms:
Phosphoproteomics: Mass spectrometry-based phosphoproteomic analysis can comprehensively map signaling networks activated by HGF beyond the canonical pathways, revealing unexpected connections and feedback mechanisms.
ChIP-seq: Chromatin immunoprecipitation followed by sequencing identifies the genome-wide binding patterns of transcription factors activated downstream of HGF signaling, revealing the complete transcriptional program.
Proximity labeling: BioID or APEX2-based proximity labeling coupled with mass spectrometry can identify proteins that dynamically interact with c-Met receptor following HGF binding.
Live-cell biosensors: FRET-based biosensors for key kinases (p38, ERK) allow real-time visualization of signaling dynamics in living cells with subcellular resolution.
Ribosome profiling: Provides genome-wide information on actively translated mRNAs following HGF treatment, distinguishing between transcriptional and translational regulation.
Spatial transcriptomics: In complex tissue models, spatial transcriptomics can map HGF-induced gene expression changes with preserved spatial information, revealing intercellular communication networks.
Hepatoma-Derived Growth Factor (HDGF) is a protein that was initially identified in research aimed at discovering novel growth factors for hepatoma cells. HDGF is a member of a new gene family that includes four related proteins. This protein plays a significant role in cell growth, angiogenesis, and apoptosis, making it a crucial factor in both normal physiological processes and the progression of various malignant diseases .
HDGF was first purified from the conditioned medium of a hepatoma cell line. The protein is composed of 285 amino acids, with the recombinant form typically including the amino acids from position 32 to 285. This truncated form retains the biological activity of the full-length protein and is often used in research and therapeutic applications .
HDGF functions as a growth factor, angiogenic factor, and anti-apoptotic factor. It is ubiquitously expressed in non-cancerous tissues and is involved in organ development and tissue repair. In cancerous tissues, HDGF expression is often elevated, and it contributes to tumor growth and progression by promoting cell proliferation, angiogenesis, and resistance to apoptosis .
The high expression of HDGF has been closely associated with unfavorable clinical outcomes in several malignant diseases, including hepatocellular carcinoma (HCC). Studies have shown that HDGF promotes the proliferation of HCC cells and that reducing its expression can inhibit tumor growth. This makes HDGF a potential target for therapeutic interventions in cancer treatment .