HGF B Human

Hepatocyte Growth Factor B Chain Human Recombinant
Shipped with Ice Packs
In Stock

Description

Functional Roles

HGF β mediates biological activity by binding to the c-Met receptor, initiating downstream signaling pathways :

  • Cell Motility and Morphogenesis: Facilitates epithelial-mesenchymal interactions critical for embryogenesis .

  • Tissue Regeneration: Promotes organ repair in liver, kidney, and lung injuries .

  • Cancer Progression: Enhances tumor invasiveness, angiogenesis, and chemoresistance in gliomas and other cancers .

3.1. Mechanistic Insights

  • HGF β alone binds c-Met with a dissociation constant (K<sub>d</sub>) of 90 nM, inducing receptor dimerization and activation .

  • Mutations in the Met-binding interface (e.g., Arg695) disrupt signaling, confirming its role in complex stability .

3.2. Therapeutic Applications

  • Antibody Targeting: Monoclonal antibodies against HGF β (e.g., #52445) inhibit tumor growth by blocking c-Met activation .

  • siRNA Strategies: Silencing HGF β reduces glioma cell proliferation and migration in vitro .

4.1. Disease Biomarkers

  • Elevated serum HGF β levels correlate with hepatic fibrosis in hepatitis B and poor prognosis in heart failure .

  • In gliomas, high tumor-derived HGF β expression predicts shorter survival (HR = 2.8, P = 0.008) .

4.2. Therapeutic Challenges

  • Rapid hepatic clearance (t<sub>1/2</sub> = 4 minutes) limits exogenous HGF β efficacy .

  • Small-molecule modulators (e.g., Dihexa) enhance HGF β activity for neurodegenerative disease treatment .

Detection and Research Tools

MethodApplicationExample
Western BlottingDetects endogenous HGF β (35–85 kDa bands) HGF β (D6S7D) XP® Rabbit mAb
Recombinant ProductionGenerates bioactive HGF heterodimer (78.0 kDa) Prospec Bio HGF Protein (CYT-244)
siRNA TransfectionReduces HGF β mRNA/protein in glioma models HGF siRNA (Ribo Biotechnology)

Future Directions

  • Targeted Drug Delivery: Nanoparticle formulations to prolong HGF β half-life in regenerative therapies .

  • Combination Therapies: Co-administering HGF β inhibitors with chemotherapy to overcome resistance .

Product Specs

Introduction
Hepatocyte Growth Factor (HGF) is a versatile growth factor that influences both cell growth and movement. It exhibits a potent mitogenic effect on hepatocytes and primary epithelial cells. HGF acts synergistically with Interleukin-3 and GM-CSF to promote colony formation of hematopoietic progenitor cells in vitro, suggesting a potential role in modulating hematopoiesis. HGF is initially secreted as a single, inactive polypeptide chain. It undergoes cleavage by serine proteases into a 69kDa Alpha chain and a 34kDa Beta chain. The active heterodimeric molecule is formed through a disulfide bond linking the alpha and beta chains.
Description
Recombinant Human HGF-B, produced in E. coli, is a single, non-glycosylated polypeptide chain. It consists of 234 amino acids (fragment 495-728) and has a molecular weight of 34kDa. The protein includes a 4.5kDa amino-terminal hexahistidine tag. HGF-B is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
HGF-B protein is supplied in a buffer containing 25mM Sodium Acetate (pH 4.8), 1mM EDTA, and 50% glycerol.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For extended storage, freeze at -20°C. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 95.0% using SDS-PAGE analysis.
Synonyms
Scatter Factor, SF, Hepatopoietin, HPTA, HGF, HGFB, F-TCF, DFNB39, Hepatocyte growth factor, Hepatocyte growth factor beta chain.
Source
Escherichia Coli.
Amino Acid Sequence
VVNGIPTRTNIGWMVSLRYRNKHICGGSLIKESWVLTAR
QCFPSRDLKDYEAWLGIHDVHGRGDEKCKQVLNVSQLV
YGPEGSDLVLMKLARPAVLDDFVSTIDLPNYGCTIPEKTS
CSVYGWGYTGLINYDGLLRVAHLYIMGNEKCSQHHRGK
VTLNESEICAGAEKIGSGPCEGDYGGPLVCEQHKMRMV
LGVIVPGRGCAIPNRPGIFVRVAYYAKWIHKIILTYKVPQS.

Q&A

What is Hepatocyte Growth Factor (HGF) and what are its primary biological functions?

Hepatocyte Growth Factor (HGF) is a multifunctional cytokine that plays critical roles in various biological processes including angiogenesis, cell proliferation, anti-fibrosis, and antiapoptosis . Initially identified as a potent mitogen for hepatocytes, HGF is now recognized as a pleiotropic growth factor with effects extending beyond the liver. HGF is primarily produced by mesenchymal cells and acts on epithelial and endothelial cells expressing the c-Met receptor tyrosine kinase .

The primary biological functions of HGF include:

  • Liver regeneration and hepatoprotection

  • Tissue repair and wound healing

  • Embryonic development

  • Angiogenesis (formation of new blood vessels)

  • Modulation of immune responses

  • Anti-fibrotic activity in multiple organs

  • Neurotropic effects in the central nervous system

In research settings, understanding these diverse functions requires multiple experimental approaches, including in vitro cell culture systems, animal models of tissue injury, and clinical evaluations of HGF levels in various disease states.

How are HGF levels measured in clinical and research settings?

The most common methodology for measuring HGF levels in clinical and research settings is the Enzyme-Linked Immunosorbent Assay (ELISA). As described in the referenced study, HGF measurement typically follows this protocol:

  • Sample preparation: Biological samples (serum, cerebrospinal fluid, etc.) are centrifuged at 1000g for 15 minutes prior to analysis .

  • ELISA procedure: Commercial ELISA kits (such as those from Biosource International Inc.) are commonly employed, following the supplier's recommendations .

  • Calibration: Recombinant human HGF is used as a calibrator with standard concentrations (typically ranging from approximately 150 to 10,000 pg/ml) .

  • Sample analysis: Samples are measured in duplicates to ensure reliability .

  • Quantification: Results are expressed in pg/ml .

For research purposes requiring higher sensitivity or specificity, alternative methods include:

  • Quantitative PCR for HGF mRNA expression

  • Western blotting for protein analysis

  • Immunohistochemistry for tissue localization

  • Cell-based bioassays to measure functional activity

When interpreting results, researchers should consider sample handling procedures, assay detection limits, and potential cross-reactivity with other growth factors.

What is the relationship between HGF and its receptor c-Met in normal physiology?

The biological effects of HGF are mediated through binding to its specific receptor, c-Met, which is a transmembrane receptor tyrosine kinase. This interaction initiates a complex signaling cascade that regulates various cellular responses. The HGF/c-Met signaling axis demonstrates several important characteristics:

  • Cell type specificity: While HGF is primarily produced by mesenchymal cells, c-Met is expressed on epithelial cells, endothelial cells, and some immune cells like dendritic cells . This creates a paracrine signaling system.

  • Tissue distribution: Research has confirmed that dendritic cells express the receptor for HGF (c-Met), which is not expressed in T cells . This differential expression pattern is critical for understanding the cell-specific effects of HGF.

  • Signaling mechanism: Upon HGF binding, c-Met undergoes dimerization and autophosphorylation, activating multiple downstream pathways including PI3K/Akt, Ras/MAPK, and STAT3.

  • Physiological regulation: In healthy tissues, HGF/c-Met signaling is tightly regulated through feedback mechanisms, receptor internalization, and degradation.

  • Developmental importance: HGF and c-Met are both detected in developing and adult mammalian brains, suggesting an important role as a neurotrophic factor .

Understanding this relationship is fundamental to investigating both normal physiological processes and pathological conditions where HGF/c-Met signaling may be dysregulated.

How do HGF levels correlate with liver damage in patients with chronic hepatitis B?

Research demonstrates a significant correlation between serum HGF levels and markers of liver damage in patients with chronic hepatitis B. The relationship appears to be multifaceted and provides insights into disease progression and severity.

Studies have established the following correlations:

  • Relationship with viral replication: Serum HGF levels in patients with chronic hepatitis B show a statistically significant correlation with HBV-DNA levels (r: 0.951, p<0.05) . This strong correlation suggests that serum HGF may serve as a secondary marker of viral replication.

  • Association with liver enzymes: HGF levels correlate significantly with serum alanine aminotransferase (ALT) levels (r: 0.816, p<0.05) , as illustrated in Figure 1 of the referenced study. This indicates a relationship between HGF production and ongoing hepatocyte damage.

  • Correlation with histopathological findings: HGF levels show significant correlation with:

    • Fibrosis score (r: 0.750, p<0.05)

    • Hepatic activity index (HAI) (r: 0.459, p<0.05)

These correlations suggest that serum HGF levels in chronic hepatitis B may reflect:

  • The extent of viral replication

  • The degree of necro-inflammatory activity in the liver

  • The structural progression of liver disease

The correlation between HGF and fibrosis is particularly notable since HGF is known to have anti-fibrotic properties in experimental models. This paradoxical relationship may represent a compensatory response to liver injury, where increased HGF production occurs as an attempt to counteract fibrosis progression.

What is the role of HGF in immune regulation, particularly regarding dendritic cell function?

HGF has emerged as a significant immunoregulatory molecule with particular effects on dendritic cell (DC) function. Research has revealed several mechanisms through which HGF modulates immune responses:

  • Effects on dendritic cells:

    • DCs express the c-Met receptor, making them direct targets of HGF signaling

    • HGF treatment both in vitro and in vivo potently suppresses DC functions, particularly their antigen-presenting capacity

    • This suppression leads to downregulation of antigen-induced Th1- and Th2-type immune responses

  • Impact on allergic airway inflammation:

    • Exogenous administration of HGF expression plasmid into antigen-primed mice markedly suppresses:

      • Development of airway eosinophilia

      • Airway hyperresponsiveness

      • Antigen-presenting capacity of DCs in the lung

  • Mechanism of immunosuppression:

    • HGF exhibits these immunosuppressive effects without upregulation of traditional immunoregulatory cytokines like IL-10 or TGF-β

    • This suggests a unique mechanism of action distinct from other immunomodulatory factors

  • Endogenous HGF regulation:

    • Expression of endogenous HGF in the lung significantly increases following antigen sensitization and inhalation challenges

    • Neutralization of endogenous HGF in vivo significantly increases airway eosinophilia and hyperresponsiveness

These findings highlight a novel role for HGF as a potential therapeutic agent in immune-mediated disorders such as asthma. The ability of HGF to suppress DC function without inducing known immunosuppressive cytokines suggests a unique mechanism that could be exploited for targeted immunomodulation with potentially fewer side effects than current immunosuppressive therapies.

How does HGF contribute to the pathophysiology of meningitis, particularly tuberculous meningitis?

HGF plays a significant role in the pathophysiology of meningitis, with particularly notable findings in tuberculous meningitis. The research reveals several important aspects of HGF's involvement:

  • Elevated HGF levels in meningitis:

    • CSF HGF levels are significantly higher in patients with both acute bacterial meningitis and tuberculous meningitis compared to control groups (p<0.05)

    • Notably, CSF HGF levels in tuberculous meningitis are statistically significantly higher than those in acute bacterial meningitis (p<0.05)

  • Localization and synthesis of HGF in the CNS:

    • mRNA for HGF and its activator HGFA is expressed in white matter astrocytes in human brain tissue

    • Most HGF in CSF is produced intrathecally, with only a small amount being of extrathecal origin

    • HGF is released differently at different sites of the brain, which may explain the particularly high levels in tuberculous meningitis

  • Potential mechanisms in tuberculous meningitis:

    • There appears to be a similarity between brain sites affected in tuberculous meningitis and sites where HGF is more commonly identified

    • This suggests that the particularly high CSF HGF levels in tuberculous meningitis may be related to the specific pattern of brain involvement

  • Diagnostic implications:

    • The differential elevation of HGF in tuberculous versus bacterial meningitis suggests that "evaluation of HGF levels in CSF may provide additional information in the differential diagnosis of meningitis"

This research represents the first documentation of CSF HGF levels in tuberculous meningitis, highlighting a potentially important biomarker for differential diagnosis. The findings suggest that HGF may be involved in the brain's response to infection, particularly in tuberculous meningitis, though the precise mechanisms require further investigation.

What are the optimal experimental approaches to study HGF signaling in different cell types?

Studying HGF signaling across different cell types requires a multi-faceted experimental approach that accounts for the complex biology of the HGF/c-Met axis. Researchers should consider the following methodological strategies:

  • Receptor expression analysis:

    • Flow cytometry to quantify c-Met expression on cell surfaces

    • RT-PCR and western blotting to assess c-Met at mRNA and protein levels

    • Single-cell RNA sequencing to identify cell populations expressing c-Met

    • Immunohistochemistry to visualize receptor distribution in tissues

  • Signaling pathway investigation:

    • Phospho-specific antibodies to detect activated c-Met and downstream mediators

    • Inhibitor studies using selective c-Met inhibitors to confirm signaling specificity

    • Time-course experiments to capture transient signaling events

    • Pathway-specific reporter assays to quantify transcriptional responses

  • Cell-specific functional assays:

    • For hepatocytes: Proliferation assays, metabolic function tests

    • For immune cells: Antigen presentation assays, cytokine production measurement

    • For epithelial cells: Migration, invasion, and wound healing assays

    • For endothelial cells: Tube formation and angiogenesis models

  • Genetic manipulation approaches:

    • CRISPR/Cas9 editing of c-Met or downstream components

    • Conditional knockout models to study cell-specific effects

    • Expression of constitutively active c-Met to mimic sustained signaling

    • siRNA knockdown for transient reduction of signaling components

  • In vivo models with cell type-specific readouts:

    • Transgenic mice expressing reporters under c-Met responsive elements

    • Cell-specific conditional deletion of c-Met in target tissues

    • Bone marrow chimeras to distinguish hematopoietic vs. non-hematopoietic responses

    • Lineage tracing to identify cellular targets of HGF in complex tissues

These approaches should be tailored to the specific research question and cell types of interest. For instance, when studying dendritic cells, researchers demonstrated that these cells express c-Met (unlike T cells), allowing HGF to directly modulate their antigen-presenting capacity . Similar cell-specific analyses can reveal the diverse functions of HGF across different tissues and disease states.

How should researchers design studies to evaluate HGF as a potential therapeutic agent in immune-mediated disorders?

Designing rigorous studies to evaluate HGF as a therapeutic agent in immune-mediated disorders requires careful consideration of multiple experimental parameters. Based on existing research, particularly regarding HGF's immunomodulatory effects, the following framework is recommended:

  • Preclinical model selection and validation:

    • Choose models that recapitulate key aspects of human immune pathology

    • For allergic airway inflammation: Consider ovalbumin or house dust mite sensitization models

    • For autoimmune conditions: Select models with established pathogenic mechanisms

    • Validate models by confirming typical disease parameters before HGF intervention

  • HGF delivery strategy considerations:

    • Recombinant protein administration: Determine optimal dose, frequency, and route

    • Gene therapy approach: Select appropriate vectors (plasmid, viral) and promoters

    • Cell-based delivery: Consider engineered cells that overexpress HGF

    • Pharmacological inducers: Identify compounds that upregulate endogenous HGF

  • Timing of intervention:

    • Preventive protocol: Administer HGF before disease induction

    • Therapeutic protocol: Introduce HGF after disease establishment

    • Comparative analysis: Directly compare preventive vs. therapeutic efficacy

  • Comprehensive outcome assessment:

    • Clinical parameters: Disease-specific symptoms and signs

    • Cellular analysis:

      • Immune cell profiling by flow cytometry

      • Assessment of dendritic cell function (antigen presentation capacity)

      • T cell subset analysis (Th1/Th2/Th17/Treg balance)

    • Molecular markers:

      • Cytokine/chemokine profiles

      • Tissue-specific inflammatory mediators

      • Signaling pathway activation states

  • Mechanistic investigation:

    • Blocking studies: Use neutralizing antibodies against HGF or c-Met inhibitors

    • Cell-specific effects: Isolate and test immune cell subsets ex vivo after in vivo treatment

    • Combine with other immunomodulators to identify synergistic effects

    • Include controls to rule out effects through IL-10 or TGF-β upregulation

  • Translation-focused elements:

    • Dose-response studies to establish minimum effective dose

    • Toxicity and off-target effect assessment

    • Pharmacokinetic/pharmacodynamic analysis

    • Biomarker identification for potential patient stratification

When implementing this framework, researchers should be aware that HGF exhibits immunosuppressive effects without upregulating traditional immunoregulatory cytokines like IL-10 or TGF-β , suggesting a unique mechanism that warrants careful characterization in each immune disorder context.

What are the potential clinical applications of measuring HGF levels in patients with hepatitis B?

The measurement of HGF levels in hepatitis B patients offers several potential clinical applications that could enhance disease management and prognostication:

  • Disease activity assessment:

    • The significant correlation between serum HGF levels and ALT (r: 0.816, p<0.05) suggests HGF could serve as a biomarker of ongoing hepatocyte damage

    • HGF levels might provide complementary information to traditional liver enzymes, particularly in cases with discordant enzyme patterns

  • Viral replication monitoring:

    • The strong correlation between serum HGF and HBV-DNA levels (r: 0.951, p<0.05) indicates HGF could serve as an alternate marker of viral replication

    • This could be particularly valuable in resource-limited settings where nucleic acid testing is less accessible

  • Liver fibrosis evaluation:

    • HGF levels correlate significantly with fibrosis score (r: 0.750, p<0.05)

    • This correlation suggests HGF could serve as a non-invasive biomarker for fibrosis, potentially reducing the need for liver biopsies

    • Integrated into multi-parameter models, HGF might improve the accuracy of non-invasive fibrosis assessment

  • Prognostic stratification:

    • Different HGF patterns in acute versus chronic hepatitis B suggest potential for distinguishing disease phases

    • Serial HGF measurements might help identify patients at higher risk of disease progression

  • Treatment response prediction:

    • Baseline HGF levels might predict response to antiviral therapy

    • Changes in HGF levels during treatment could serve as early indicators of response

    • Post-treatment HGF patterns might help identify patients at risk for relapse

  • HGF-targeted interventions:

    • Understanding HGF's role in liver regeneration could inform the development of therapeutic strategies

    • Modulation of the HGF pathway might represent a novel approach to enhance liver repair in chronic hepatitis B

While these applications show promise, several research priorities should be addressed to validate HGF's clinical utility, including larger prospective studies, standardization of measurement methods, and direct comparisons with established biomarkers. The potential integration of HGF measurement into clinical algorithms for hepatitis B management represents an important area for translational research.

How might the immunoregulatory properties of HGF be harnessed for therapeutic purposes?

The immunoregulatory properties of HGF offer significant potential for therapeutic applications in various immune-mediated disorders. Based on current research findings, several strategic approaches could be developed:

  • Allergic and inflammatory airway diseases:

    • HGF administration has demonstrated suppression of airway eosinophilia and airway hyperresponsiveness in experimental models

    • Potential delivery methods include:

      • Aerosolized recombinant HGF for direct lung delivery

      • Gene therapy using HGF expression plasmids

      • Cell-based therapies using HGF-secreting mesenchymal stem cells

  • Autoimmune disorders:

    • HGF's ability to suppress dendritic cell function and subsequent T cell responses could be beneficial in conditions like:

      • Rheumatoid arthritis

      • Multiple sclerosis

      • Inflammatory bowel disease

      • Type 1 diabetes

  • Transplantation:

    • HGF's immunomodulatory effects could help prevent graft rejection

    • Combined with standard immunosuppressants, HGF might allow dose reduction of conventional drugs, potentially reducing side effects

  • Chronic inflammatory liver diseases:

    • Beyond hepatitis B, HGF's dual role in liver regeneration and immune modulation makes it an attractive candidate for:

      • Non-alcoholic steatohepatitis (NASH)

      • Autoimmune hepatitis

      • Primary biliary cholangitis

  • Therapeutic development considerations:

    • Advantages of HGF-based approaches:

      • Novel mechanism distinct from IL-10 or TGF-β mediated immunosuppression

      • Potential for tissue-specific effects via targeted delivery

      • Combined regenerative and immunomodulatory properties

    • Challenges to address:

      • Optimal dosing and administration routes

      • Potential for unwanted effects (e.g., tumor promotion)

      • Development of stable recombinant proteins or effective gene delivery systems

  • Combination therapy strategies:

    • HGF combined with conventional immunosuppressants

    • Sequential therapy using HGF at specific disease stages

    • Synergistic combinations with other biologics

The therapeutic potential of HGF's immunoregulatory properties is particularly promising because it offers a mechanistically distinct approach compared to current immunomodulatory drugs. The ability to suppress dendritic cell function while potentially promoting tissue repair represents a unique therapeutic profile that warrants further translational investigation.

What are the key methodological challenges in studying the diverse biological roles of HGF?

Investigating the multifaceted roles of HGF presents several methodological challenges that researchers must address to advance our understanding of this complex growth factor:

To overcome these challenges, researchers should consider:

  • Developing standardized protocols for HGF detection and functional assessment

  • Creating reporter systems to visualize HGF signaling in real-time

  • Establishing tissue-specific and inducible genetic models

  • Employing systems biology approaches to capture complex signaling networks

  • Enhancing collaboration between basic scientists and clinicians to facilitate translational research

Addressing these methodological challenges will be crucial for unlocking the full potential of HGF in both basic research and therapeutic applications.

What are the promising future research directions for understanding HGF's role in human health and disease?

The current understanding of HGF biology reveals several promising research directions that could significantly advance both basic science knowledge and therapeutic applications:

  • Systems-level analysis of HGF signaling networks:

    • Application of proteomics and phosphoproteomics to map complete HGF signaling networks

    • Single-cell analysis to decipher cell-specific responses to HGF

    • Network modeling to predict outcomes of HGF pathway modulation

    • Integrative multi-omics approaches to contextualize HGF signaling within broader cellular processes

  • Mechanistic investigation of HGF's immunomodulatory properties:

    • Detailed characterization of HGF effects on dendritic cell subsets beyond antigen presentation

    • Examination of potential direct or indirect effects on other immune cell populations

    • Investigation of the unique immunosuppressive mechanism that operates independently of IL-10 and TGF-β

    • Exploration of potential epigenetic modulation by HGF in immune cells

  • Tissue-specific roles in pathological conditions:

    • Further investigation of HGF's role in tuberculous meningitis, extending the novel findings

    • Exploration of the apparent paradox between HGF's anti-fibrotic properties and its correlation with fibrosis in chronic hepatitis B

    • Investigation of tissue microenvironment factors that modify HGF activity in different organs

    • Examination of the HGF axis in emerging infectious diseases

  • Therapeutic development and precision medicine:

    • Development of targeted delivery systems for HGF or HGF mimetics

    • Creation of tissue-specific c-Met modulators with improved safety profiles

    • Identification of biomarkers to predict response to HGF-based therapies

    • Design of combination approaches that leverage HGF's pleiotropic effects

  • Translational research priorities:

    • Validation of HGF as a biomarker in large, prospective cohorts of hepatitis B patients

    • Clinical trials of HGF-based therapies in immune-mediated disorders

    • Development of point-of-care testing for HGF to facilitate clinical applications

    • Investigation of HGF genetics and pharmacogenomics to personalize therapeutic approaches

  • Novel technological approaches:

    • Development of optogenetic tools to spatiotemporally control HGF signaling

    • Creation of engineered cells with synthetic HGF regulatory circuits

    • Application of CRISPR screening to identify new regulators and effectors of HGF signaling

    • Use of organ-on-chip technologies to model complex HGF-mediated interactions

These research directions represent opportunities to advance our understanding of HGF biology while developing novel diagnostic and therapeutic approaches. The multifunctional nature of HGF – spanning regeneration, immunomodulation, and tissue protection – makes it a particularly attractive target for translational research with potential impact across multiple medical specialties.

Product Science Overview

Structure and Function

HGF is initially secreted as a single inactive polypeptide. This precursor is cleaved by serine proteases into two chains: a 69-kDa alpha-chain and a 34-kDa beta-chain. These chains are linked by a disulfide bond to form the active, heterodimeric molecule . The beta-chain, in particular, is essential for the biological activity of HGF, as it contains the receptor-binding site necessary for activating the c-Met receptor .

Recombinant Human HGF

Recombinant human HGF (rh-HGF) is produced using DNA technology, where the gene encoding HGF is inserted into a host cell line, such as CHO (Chinese Hamster Ovary) cells, to produce the protein . The recombinant protein is then purified to achieve high levels of purity and activity. The recombinant form of HGF is used in various research and clinical applications due to its ability to stimulate hepatocyte proliferation and act as an anti-apoptotic factor .

Clinical Applications

HGF has shown potential as a therapeutic agent for treating fatal liver diseases, such as fulminant hepatitis (FH) and late-onset hepatic failure (LOHF). Clinical trials have been conducted to evaluate the safety, pharmacokinetics, and clinical efficacy of rh-HGF in patients with these conditions . Although some adverse effects, such as a decrease in blood pressure and renal toxicity, were observed in preclinical studies, these effects were manageable and did not require cessation of treatment .

Research and Development

The development of rh-HGF has been a significant milestone in the field of regenerative medicine. Researchers have successfully cloned the cDNA of human HGF, elucidated its primary structure, and identified it as a novel growth factor with unique structural characteristics . This has paved the way for further studies on the therapeutic potential of HGF in various diseases and conditions.

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.