Recombinant Human Hepatocyte growth factor receptor (MET), partial (Active)

Shipped with Ice Packs
In Stock

Description

Mechanism of Activation

MET activation involves asymmetric dimerization induced by HGF or its splice variant NK1 :

  • HGF Binding: One HGF molecule bridges two MET receptors via distinct interfaces (N-SEMA and SPH-SEMA), inducing conformational changes that trigger kinase activation .

  • NK1 Binding: NK1 forms a symmetric dimer, recruiting two MET receptors in a parallel orientation .

  • Heparin Stabilization: Enhances HGF-MET binding by acting as a molecular "glue" between MET's IPT1 domain and HGF's N domain .

Critical Residues for Activation:

  • HGF: K34, R35, R36 (N domain); mutations here reduce MET activation .

  • MET: R592, K595, K599 (IPT1 domain); disruption diminishes heparin-mediated stabilization .

Functional Roles in Biological Processes

MET signaling is essential for:

Liver Regeneration

  • Post-Hepatectomy: MET activation triggers ERK1/2 and PI3K/AKT pathways, driving hepatocyte proliferation .

  • Anti-Apoptosis: Protects hepatocytes from Fas-induced apoptosis via STAT3 and NF-κB pathways .

Cancer and Metastasis

  • Tumor Growth: Aberrant MET activation promotes invasiveness and resistance to targeted therapies .

  • Stem Cell Support: MET signaling sustains hepatic stem cells (oval cells) during chronic injury .

Research Applications

Recombinant MET (partial) is utilized in:

Drug Development

  • PROTAC Degradation: Used to validate degradation of integral membrane proteins like MET .

  • Inhibitor Screening: Assess kinase inhibitors targeting MET’s tyrosine kinase domain .

Therapeutic Potential

  • Liver Failure: Recombinant HGF (acting via MET) showed hepatoprotective effects in phase I/II trials for fulminant hepatitis .

  • Neurodegeneration: Intrathecal HGF administration is under trial for ALS and spinal cord injury .

Challenges in Targeting MET

  • Tumor Resistance: MET amplification or mutation drives resistance to EGFR inhibitors .

  • Dual Roles: MET activation aids tissue repair but exacerbates cancer progression .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered 1xPBS, pH 7.4.
Form
Lyophilized powder
Lead Time
Typically, we can ship products within 5-10 business days after receiving your order. Delivery time may vary depending on the purchase method and location. Please consult your local distributors for specific delivery timelines.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend centrifuging the vial briefly before opening to ensure the contents settle to the bottom. Reconstitute the protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our default final glycerol concentration is 50%. Customers may use this 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. Generally, the shelf life of liquid formulations is 6 months at -20°C/-80°C. Lyophilized formulations typically have a shelf life of 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
C-terminal hFc-tagged
Synonyms
AUTS9; c met; D249; Hepatocyte growth factor receptor; HGF; HGF receptor; HGF/SF receptor; HGFR; MET; Met proto oncogene; Met proto oncogene tyrosine kinase; MET proto oncogene; receptor tyrosine kinase; Met proto-oncogene (hepatocyte growth factor receptor); Met proto-oncogene; Met protooncogene; MET_HUMAN; Oncogene MET; Par4; Proto-oncogene c-Met; RCCP2; Scatter factor receptor; SF receptor; Tyrosine-protein kinase Met
Datasheet & Coa
Please contact us to get it.
Expression Region
25-932aa
Mol. Weight
128.4 kDa
Protein Length
Extracellular Domain
Purity
Greater than 95% as determined by SDS-PAGE.
Research Area
Signal Transduction
Source
Mammalian cell
Species
Homo sapiens (Human)
Target Names
MET
Uniprot No.

Target Background

Function
Hepatocyte growth factor receptor (MET) is a receptor tyrosine kinase that transduces signals from the extracellular matrix into the cytoplasm by binding to the hepatocyte growth factor/HGF ligand. It plays a crucial role in regulating various physiological processes, including cell proliferation, scattering, morphogenesis, and survival. Upon ligand binding at the cell surface, MET undergoes autophosphorylation on its intracellular domain, creating docking sites for downstream signaling molecules. Following activation by the ligand, MET interacts with PI3-kinase subunit PIK3R1, PLCG1, SRC, GRB2, STAT3, or the adapter GAB1. The recruitment of these downstream effectors by MET activates several signaling cascades, including RAS-ERK, PI3 kinase-AKT, or PLCgamma-PKC. RAS-ERK activation is associated with morphogenetic effects, while PI3K/AKT coordinates prosurvival effects. During embryonic development, MET signaling participates in gastrulation, muscle and neuronal precursor development and migration, angiogenesis, and kidney formation. In adults, it is involved in wound healing, organ regeneration, and tissue remodeling. It also promotes hematopoietic cell differentiation and proliferation. MET may regulate cortical bone osteogenesis. It serves as a receptor for Listeria monocytogenes internalin InlB, facilitating the entry of the pathogen into cells.
Gene References Into Functions
  1. The miR-19a/c-Met pathway plays a crucial role in acquired resistance to gefitinib, suggesting that manipulation of miR-19a may offer a therapeutic strategy for overcoming acquired gefitinib resistance. PMID: 28592790
  2. Expression of C-Met and HER2 proteins in lung adenocarcinoma is highly correlated. Further investigation is warranted to determine whether their co-expression exhibits synergy in the targeted therapy of lung adenocarcinoma. PMID: 29400000
  3. MET overexpression is more frequently observed in high-grade myxofibrosarcoma and the epithelioid variant. Chromosome 7 polysomy, rather than MET gene regional amplification, may account for the overexpression of MET protein. PMID: 30126419
  4. miR-449a suppresses hepatocellular carcinoma tumorigenesis by down-regulating activity in the c-Met/ERK pathway. PMID: 30108016
  5. MET amplifications were identified in two cases of endometrial clear-cell carcinoma with mixed features. PMID: 29633423
  6. NGS enables the detection of low-abundant ctDNA in blood based on ultra-deep sequencing. A patient who benefited from crizotinib despite the low abundance of MET exon 14 skipping demonstrates that targeted therapy can be chosen even with low abundance of gene mutations. PMID: 29110851
  7. The interplay of dual MET/HER2 overexpression in the AKT and ERK pathways for esophageal cancer is described. Combination therapy could be a novel strategy for EAC with amplification of both MET and HER2. PMID: 29223420
  8. MET inactivation in the context of the BRAF-activating mutation is driven through a negative feedback loop involving inactivation of PP2A phosphatase, which in turn leads to phosphorylation on MET inhibitory Ser985. PMID: 30224486
  9. MET Exon 14 Skipping Mutations in Non-small Cell Lung Cancer PMID: 30037377
  10. MET activation, by either METex14 mutations or amplification, is a characteristic of a subset of early stage NSCLCs and may coexist with ERBB2 amplification. PMID: 29139039
  11. Serum level of miR-658 is significantly lower in the NM group than in the DM group. The levels of PAX3 and MET are also lower in the NM group. Both overexpression and silencing of miR-658 significantly up-regulate or down-regulate the levels of PAX3 and MET in gastric cell lines. PMID: 29630524
  12. MiR-206 inhibits the development of epithelial ovarian cancer cell by directly targeting c-Met and inhibiting the c-Met/AKT/mTOR signaling pathway. PMID: 29807226
  13. These results suggest that gastric cancer progression is not associated with a unique signaling pathway and that a feedback loop may exist between the HGF/c-Met and Notch1 signaling pathways, which may result in therapeutic resistance. PMID: 29781036
  14. Comparative analysis revealed a strong association between MET expression and MET amplification (85% concurrence) in primary stomach tumors and matched liver metastasis. Survival analyses revealed that both MET amplification and MET overexpression were prognostic of poor outcomes. PMID: 29790169
  15. High c-met expression is associated with oral squamous cell carcinoma. PMID: 29286169
  16. FOXO1 serves as an important linker between HER2 and MET signaling pathways through negative crosstalks and is a key regulator of the acquired lapatinib resistance in HER2-positive GC cells. PMID: 28343375
  17. Analysis of how the cMET blockade augments radiation therapy in patients with NF2 PMID: 29440379
  18. These findings highlight the relevance of cross-species protein interactions between murine feeder cells and human epithelial cells in 3T3-J2 co-culture and demonstrate that STAT6 phosphorylation occurs in response to MET activation in epithelial cells. However, STAT6 nuclear translocation does not occur in response to HGF, precluding the transcriptional activity of STAT6. PMID: 29771943
  19. c-Met-activated Mesenchymal Stem Cells (MSC) pre-exposed to hypoxia interact with PrPC at the site of ischemic injury to increase the efficiency of MSC transplantation. PMID: 29705776
  20. A novel G-quadruplex motif formed in the Human MET promoter region. PMID: 29054971
  21. A METex14 del mutation-positive NSCLC patient who responded to crizotinib but later relapsed, demonstrated a mixed response to glesatinib including reduction in size of a MET Y1230H mutation-positive liver metastasis and concurrent loss of detection of this mutation in plasma DNA. These data demonstrate that glesatinib exhibits a distinct mechanism of target inhibition and can overcome resistance to PMID: 28765324
  22. This study demonstrates that simultaneous inhibition of c-Met and Src signaling in MD-MSCs triggers apoptosis and reveals vulnerable pathways that could be exploited to develop NF2 therapies. PMID: 28775147
  23. Prolonged treatment with a single HGF/c-Met or Hh inhibitor leads to resistance to these single inhibitors, likely because the single c-Met treatment leads to enhanced expression of Shh, and vice versa. Targeting both the HGF/c-Met and Hh pathways simultaneously overcame the resistance to the single-inhibitor treatment and led to a more potent antitumor effect in combination with the chemotherapy treatment. PMID: 28864680
  24. We identified unique and tumor-specific tyrosine phosphorylation rewiring in tumors resistant to treatment with the irreversible third-generation EGFR-inhibitor, osimertinib, or the novel dual-targeting EGFR/Met antibody, JNJ-61186372. PMID: 28830985
  25. TGF-beta negatively controls the HGF/c-MET pathway by regulating stemness in glioblastoma. PMID: 29238047
  26. The preclinical efficacy and safety data provide a clear rationale for the ongoing clinical studies of Sym015 in patients with MET-amplified tumors. PMID: 28679766
  27. High MET expression is associated with malignant pleural mesothelioma. PMID: 28560410
  28. Real-time PCR and western blotting revealed that Huaier extract decreased p65 and c-Met expression and increased IkappaBalpha expression, while paclitaxel increased p65 expression and reduced IkappaBalpha and c-Met expression. The molecular mechanisms may involve the inhibition of the NF-kappaB pathway and c-Met expression PMID: 29039556
  29. Data found that the expression of c-Met was significantly increased in human oral squamous cell carcinoma (OSCC) tissues compared to normal mucosa adjacent to the tumor, but was not correlated with clinicopathological parameters. Further findings indicated the potential role of c-Met in the progression of OSCC. PMID: 29115556
  30. S49076 exerts its cytotoxic activity at low doses on MET-dependent cells through MET inhibition, whereas it inhibits growth of MET-independent cells at higher but clinically relevant doses by targeting Aurora B PMID: 28619752
  31. MET expression was shown to be significantly reduced in the superior temporal gyrus cortex of autism spectrum disorders individuals. PMID: 28322981
  32. In SCCHN, immunohistochemical overexpression of c-MET above cut-off levels III and particularly II was associated with inferior survival outcomes and advanced disease PMID: 29103754
  33. Here we present a case series of three patients who achieved were cMET amplified and showed partial response on Crizotinib PMID: 29199685
  34. c-Met/beta1 integrin complex whose ligand-independent cross-activation and robust affinity for fibronectin drives invasive oncologic processes. PMID: 28973887
  35. tivantinib did not suppress MET signaling, and selective MET inhibitors demonstrated an antiproliferative effect only in MHCC97H, the unique cell line displaying MET gene amplification. HCC tumors with high expression of cell proliferation genes defined a group of patients with poor survival. PMID: 28246274
  36. Studies show that MET mutations have been found in cancer of unknown primary origin (CUP) being clustered to the SEMA and TK domain of the receptor. The biomechanical properties of MET mutants might trigger the hyper-invasive phenotype associated with CUP. [review] PMID: 29037604
  37. Data show that Kruppel like factor 4 (KLF4) was overexpressed in met proto-oncogene protein (c-Met)-overexpressing non-small-cell lung cancer (NSCLC) cells and tissues. PMID: 29624806
  38. SOCS1 attenuates migration and invasion properties of hepatocellular carcinoma cells at least partly via modulation of MET-mediated epithelial-mesenchymal transition, and controls invasive tumor growth. PMID: 29085209
  39. The authors reconfirmed EGFR mutation as a strong predictive marker of Non-Small-Cell Lung Cancer. However, c-MET positivity was not associated with response or progression-free survival, although c-MET overexpression correlated with some clinical characteristics. PMID: 29502124
  40. Findings show oncogene E5 is primarily responsible for Met upregulation; E5-induced Met contributes to the motility of HPV-containing cells; these studies show a new role for E5 in epithelial-stromal interactions, with implications for cancer development PMID: 29609071
  41. EGFR T790M mutation and cMET amplification are main mechanisms leading to EGFR TKI resistance in lung adenocarcinoma. PMID: 29616327
  42. MET activation is associated with drug resistance in chronic myeloid leukemia. PMID: 28418880
  43. High glucose activated Met receptor in HK2 cells independently of HGF, via induction of integrin a5b1 and downstream signaling. This mode of Met activation was associated with tubular cell damage and apoptosis and it may represent a novel pathogenic mechanism and a treatment target in diabetic nephropathy. PMID: 28819999
  44. The purpose of this study was to explore gene copy number (GCN) variation of EGFR, HER2, c-MYC, and MET in patients with primary colorectal cancer. PMID: 28764718
  45. HGF/c-MET pathway mediates VEGFR inhibitor resistance and vascular remodeling in NSCLC. PMID: 28559461
  46. Because c-Met is strongly associated with pathological grade, stage, and disease-specific survival, c-Met levels may have potential to predict patient prognosis and to guide clinical diagnosis and treatment of patients with renal cell carcinoma PMID: 28427859
  47. miR-1 is downregulated in ovarian cancer tissues, and may play a tumor suppressive role by inhibiting c-Met expression and its effects on the regulation of cell proliferation, migration, and invasion PMID: 28698064
  48. Proto-oncogene proteins c-met (MET) mutations Y1248H and D1246N confer resistance in vitro and in vivo. PMID: 28396313
  49. MET overexpression is found in 23.8% of surgically resected NSCLC. MET amplification prevails in 4.6% and is associated with MET overexpression. Both have no influence on prognosis. PMID: 28838386
  50. The study highlights the role of tissue differentiation on pathological response to neoadjuvant chemotherapy in gastric cancer and shows no impact between FOXP3, HER2, and MET expression in terms of tumor regression grading PMID: 29696715

Show More

Hide All

Database Links

HGNC: 7029

OMIM: 114550

KEGG: hsa:4233

STRING: 9606.ENSP00000317272

UniGene: Hs.132966

Involvement In Disease
Hepatocellular carcinoma (HCC); Renal cell carcinoma papillary (RCCP); Deafness, autosomal recessive, 97 (DFNB97); Osteofibrous dysplasia (OSFD)
Protein Families
Protein kinase superfamily, Tyr protein kinase family
Subcellular Location
Membrane; Single-pass type I membrane protein.; [Isoform 3]: Secreted.
Tissue Specificity
Expressed in normal hepatocytes as well as in epithelial cells lining the stomach, the small and the large intestine. Found also in basal keratinocytes of esophagus and skin. High levels are found in liver, gastrointestinal tract, thyroid and kidney. Also

Q&A

What is the functional significance of c-Met receptor in liver regeneration?

The c-Met receptor plays an indispensable role in liver regeneration and repair processes. After partial hepatectomy (PHx), c-Met activation occurs rapidly, with tyrosine phosphorylation beginning within 5 minutes and peaking at approximately 60 minutes post-surgery . This activation initiates critical signaling cascades that drive hepatocyte proliferation.

Functional studies have demonstrated that mice lacking c-Met experience liver necrosis, jaundice, delayed regeneration, and significantly higher mortality rates compared to wild-type controls . The HGF/c-Met signaling pathway activates multiple downstream pathways essential for regeneration, including JAK/STAT3, PI3K/Akt/NF-κB, and Ras/Raf pathways, which collectively initiate cell proliferation programs .

Researchers investigating liver regeneration should note that c-Met signaling is particularly critical during the initial stages of regeneration, making this receptor an essential component in experimental models of liver repair and regeneration.

How does c-Met receptor activation influence intracellular signaling cascades?

Upon HGF binding, c-Met undergoes tyrosine phosphorylation, triggering multiple downstream signaling pathways that regulate cellular responses. The activation sequence begins with:

  • HGF binding and c-Met tyrosine residue phosphorylation

  • Induction of Wnt-independent nuclear translocation of β-catenin

  • Subsequent internalization and degradation of c-Met via the ubiquitin-proteasome pathway

  • Activation of multiple signaling branches

Key pathways activated by HGF/c-Met include:

Signaling PathwayFunction in RegenerationKey Mediators
JAK/STAT3Cell survival and proliferationSTAT3 transcription factors
PI3K/Akt/NF-κBAnti-apoptotic effectsAkt, NF-κB
Ras/Raf/MAPKMitotic signalingERK1/2

The Ras pathway is particularly significant, as c-Met activates ERK1/2 (two mitogen-activated protein kinases) to transmit mitotic signals. Research has demonstrated that in c-Met mutant mice, phosphorylation of ERK1/2 is absent, directly correlating with impaired liver regeneration . This indicates that ERK1/2 activation is completely dependent on c-Met signaling during the liver regeneration process.

How can researchers differentiate between active and inactive forms of recombinant c-Met?

Researchers can differentiate between active and inactive forms of recombinant c-Met through several methodological approaches:

Phosphorylation Status Assessment:
The most direct method involves detecting phosphorylated tyrosine residues using phospho-specific antibodies through Western blotting or immunoprecipitation. Active c-Met shows phosphorylation at specific tyrosine residues, particularly in the kinase domain.

Functional Assays:
Measuring downstream pathway activation through ERK1/2 phosphorylation provides an excellent functional readout. Studies have shown that during liver regeneration, ERK1/2 phosphorylation is entirely dependent on c-Met activation . Researchers can use selective inhibitors like SGX523 to confirm c-Met-specific effects, as this compound blocks c-Met phosphorylation and was shown to eliminate enhanced liver regeneration in experimental models .

Structural Analysis:
The partial active form typically refers to a truncated version containing the kinase domain, which can be distinguished from full-length receptor through molecular weight analysis or domain-specific antibodies.

When planning experiments involving recombinant c-Met, researchers should include appropriate controls to verify the activation state and consider the timing of examination, as c-Met is rapidly degraded after activation through the ubiquitin-proteasome pathway .

How do age-related changes affect c-Met expression and function in regenerative models?

Age significantly impacts c-Met expression and function, with important implications for regenerative research. Clinical studies involving 130 patients who underwent hepatectomy revealed that both HGF and c-Met expression levels were substantially lower in older patients compared to younger cohorts .

This age-dependent decline in c-Met expression correlates directly with regenerative capacity. Six months after partial hepatectomy, liver volume increase was significantly greater in younger patients than in elderly individuals . This finding suggests that age is an important variable to control in experimental designs involving c-Met and liver regeneration.

Researchers studying regenerative processes should:

  • Stratify experimental groups by age

  • Consider age as a covariate in statistical analyses

  • Potentially adjust dosing of recombinant HGF in older subjects to account for reduced receptor availability

  • Examine age-dependent changes in downstream signaling pathways

These considerations are essential for accurate interpretation of data from regenerative models and may help explain variability in experimental outcomes between different age groups.

What methodological approaches can optimize detection of c-Met activation in complex tissue samples?

Detecting c-Met activation in complex tissue samples requires sophisticated methodological approaches to ensure specificity and sensitivity:

Tissue-Specific Phospho-Profiling:
For heterogeneous tissue samples, researchers should employ phospho-specific antibodies against multiple c-Met tyrosine residues, as different residues may show variable phosphorylation patterns depending on activation context. Post-partial hepatectomy, c-Met phosphorylation follows a specific temporal pattern, beginning within 5 minutes and peaking at 60 minutes .

Multiplexed Analysis:
Combining immunohistochemistry with digital imaging allows spatial mapping of activated c-Met within specific cell populations. This approach is particularly valuable in liver samples where different cell types (hepatocytes, stellate cells, etc.) show differential c-Met expression and activation patterns.

Downstream Signaling Verification:
Confirmation of c-Met activity should include assessment of multiple downstream pathways. During liver regeneration, c-Met activates JAK/STAT3, PI3K/Akt/NF-κB, and Ras/Raf pathways . Examining these multiple readouts provides more robust confirmation of functional c-Met activation.

Temporal Considerations:
Researchers must consider the rapid dynamics of c-Met activation and degradation. After activation, c-Met is promptly internalized and degraded via the ubiquitin-proteasome pathway , necessitating careful timing of sample collection in experimental designs.

When analyzing complex tissue samples, these methodological refinements help overcome common challenges such as cell-type heterogeneity, temporal signaling dynamics, and activation pathway crosstalk.

How do regulatory non-coding RNAs influence c-Met expression and signaling?

Recent research has highlighted the significant role of non-coding RNAs in regulating the HGF/c-Met axis during liver regeneration. These regulatory elements add an important layer of post-transcriptional control that researchers must consider when studying c-Met function.

The research review indicates that non-coding RNAs participate in regulating liver regeneration through the HGF/c-Met pathway, though specific mechanisms were not detailed in the search results . This represents an emerging area of investigation that connects epigenetic regulation with growth factor receptor signaling.

For researchers investigating c-Met regulation, experimental approaches should include:

  • Profiling of microRNAs predicted to target c-Met mRNA

  • Functional validation using antisense oligonucleotides or CRISPR-based approaches

  • Assessment of long non-coding RNAs that may interact with the c-Met promoter region

  • Integration of non-coding RNA expression data with c-Met protein levels and activation status

Understanding these regulatory relationships may reveal new opportunities for modulating c-Met signaling in research and therapeutic applications, particularly in contexts where direct receptor targeting has proven challenging.

What are the optimal conditions for studying recombinant human c-Met activity in in vitro models?

Establishing optimal conditions for studying recombinant human c-Met activity in vitro requires careful attention to several experimental parameters:

Activation Conditions:
For studying c-Met activation, researchers should note that HGF binding to c-Met drives phosphorylation of c-Met tyrosine residues, followed by activation of downstream signaling . In experimental designs, both single-chain HGF and two-chain (active form) HGF should be considered, as they exist in different proportions in normal tissue with single-chain being dominant in normal liver .

Temporal Dynamics:
C-Met activation occurs rapidly, with phosphorylation beginning within minutes of stimulation. Studies should include early time points (5-60 minutes) to capture the peak phosphorylation that occurs at approximately 60 minutes post-stimulation . Additionally, researchers should account for the subsequent rapid internalization and degradation of c-Met through the ubiquitin-proteasome pathway .

Environmental Factors:
Research has shown that hypoxic conditions dramatically decrease HGF expression in hepatic stellate cells and c-Met expression in hepatocytes . Therefore, oxygen tension should be carefully controlled and reported in experimental protocols.

Pathway Specificity Controls:
To confirm c-Met-specific effects, selective inhibitors like SGX523 should be employed as controls. This compound blocks c-Met phosphorylation and has been shown to eliminate enhanced effects on liver regeneration in experimental models .

These considerations will help researchers develop robust protocols for studying recombinant human c-Met activity that account for the complex dynamics of receptor activation, signaling, and degradation.

How can researchers effectively evaluate crosstalk between c-Met and other signaling pathways?

Evaluating crosstalk between c-Met and other signaling pathways requires sophisticated experimental approaches that can distinguish direct c-Met effects from secondary pathway interactions:

Sequential Inhibition Strategy:
Researchers should employ selective inhibitors in sequence to delineate pathway hierarchies. For example, studies have demonstrated that PPARγ regulates liver regeneration by influencing the HGF/c-Met/ERK1/2 pathway. The PPARγ antagonist GW9662 was shown to accelerate liver regeneration, but this effect was eliminated when c-Met phosphorylation was blocked by SGX523 . This sequential inhibition approach helps establish causality in signaling networks.

Pathway Convergence Analysis:
Several pathways interact with c-Met signaling. For instance, the bone morphogenetic protein (BMP)9 and HGF/c-Met signaling axes establish a signal crossover through ALK1 by modulating SMAD1 (pro-survival) and p38MAPK (pro-apoptotic) pathways . Researchers should design experiments that can track multiple pathway components simultaneously through techniques like multiplexed phospho-flow cytometry or mass cytometry.

Transcription Factor Activity Mapping:
Downstream effects of pathway crosstalk can be evaluated through transcription factor activity assays. Research has shown that the cellular transcription factor late SV40 factor (LSF) regulates osteopontin, which then activates c-Met through interaction with CD44 . Monitoring transcription factor activity provides insight into the functional outcomes of pathway interactions.

These methodological approaches enable researchers to construct more complete models of how c-Met integrates with broader cellular signaling networks, particularly in complex biological processes like liver regeneration and response to injury.

What considerations are important when translating c-Met research from rodent models to human applications?

Translating c-Met research from rodent models to human applications requires careful attention to several key considerations:

Species-Specific Differences:
While the HGF/c-Met signaling pathway is conserved across mammals, there are important species-specific differences in expression patterns, regulatory mechanisms, and downstream effectors. In the PHx model, liver regeneration in rodents shows complete recovery in volume and mass within one week , but this timeline differs in humans.

Age-Dependent Variation:
Clinical studies have demonstrated significant age-related differences in HGF and c-Met expression. In a study of 130 patients undergoing hepatectomy, older patients showed substantially lower expression of both HGF and c-Met compared to younger patients, correlating with reduced regenerative capacity . Age stratification is therefore critical when designing translational studies.

Therapeutic Formulation Considerations:
Engineered forms of HGF have shown promise in translational research. For example, 1K1, a synthetic small molecule derived from the naturally occurring HGF fragment NK1, demonstrates anti-fibrotic properties and promotes liver regeneration in rodents with better stability and easier production characteristics than native HGF . Such engineered variants may bridge the gap between basic research and clinical application.

Activation and Delivery Methods:
The method of delivering recombinant proteins affects outcomes. Studies showed that administering recombinant human HGF-activator via the portal vein significantly increased liver regeneration rates compared to control groups . Administration route and activation state of recombinant proteins should be carefully considered in translational design.

By addressing these translational considerations, researchers can develop more effective strategies for applying c-Met research findings from rodent models to human therapeutic applications in liver diseases and regenerative medicine.

How should researchers interpret contradictory data regarding c-Met activation in different experimental models?

Interpreting contradictory data regarding c-Met activation across experimental models requires systematic analysis of several key variables:

Model-Specific Activation Context:
Different experimental models may represent distinct physiological or pathological states that influence c-Met activation. For example, c-Met signaling in liver regeneration after partial hepatectomy may differ from that observed in models of acute liver injury or chronic fibrosis. The research shows that HGF/c-Met plays critical roles in liver fibrosis, post-inflammatory hepatocyte regeneration, and post-transplantation liver regeneration , suggesting context-dependent functions.

Pathway Component Assessment:
Comprehensive analysis of multiple pathway components can help resolve contradictions. For instance, in cases where direct c-Met activation measurements conflict, examining downstream effectors like ERK1/2 phosphorylation, which is completely dependent on c-Met during liver regeneration , can provide clarification.

Biological Variable Stratification:
Age-related differences can explain contradictory findings. Clinical studies demonstrate that HGF and c-Met expression levels are significantly lower in older patients compared to younger patients . Stratifying data by age or other biological variables may resolve apparent contradictions.

When confronted with contradictory data, researchers should systematically evaluate these factors and consider integrating findings through computational modeling to generate testable hypotheses that may reconcile disparate observations.

What statistical approaches are most appropriate for analyzing c-Met phosphorylation dynamics in time-course experiments?

Time-course experiments investigating c-Met phosphorylation dynamics require specialized statistical approaches to properly capture the complex temporal patterns:

Mixed-Effects Modeling:
Given the rapid changes in c-Met phosphorylation status (beginning within 5 minutes and peaking at 60 minutes post-stimulation ), mixed-effects models that account for both fixed (treatment, time) and random (subject-specific) effects are optimal for analyzing longitudinal data with potential missing timepoints.

Functional Data Analysis:
The continuous nature of phosphorylation dynamics is better captured by functional data analysis approaches that model the entire phosphorylation curve rather than discrete timepoints. This is particularly important for c-Met, which undergoes rapid activation followed by ubiquitin-proteasome-mediated degradation .

Pathway-Informed Bayesian Methods:
Incorporating prior knowledge about the HGF/c-Met signaling pathway into Bayesian models can improve estimation of phosphorylation parameters. For instance, knowing that HGF binding triggers c-Met phosphorylation followed by β-catenin translocation provides temporal constraints that can inform statistical models.

Sample Size Considerations:
For detecting meaningful differences in c-Met phosphorylation, researchers should conduct power analyses that account for:

  • The expected effect size (often large for phosphorylation events)

  • Biological variability between samples

  • Technical variability in phospho-protein detection

  • Multiple testing correction for pathway analysis

These statistical approaches help researchers extract meaningful patterns from time-course phosphorylation data while accounting for the complex dynamics of c-Met activation, signaling, and degradation observed in biological systems.

How can researchers distinguish between direct and indirect effects on c-Met signaling in complex biological systems?

Distinguishing between direct and indirect effects on c-Met signaling in complex biological systems requires sophisticated experimental designs and analytical approaches:

Selective Inhibition Strategy:
Using highly selective c-Met inhibitors like SGX523 provides a direct way to isolate c-Met-specific effects. Research has demonstrated that SGX523 eliminates enhanced liver regeneration effects of PPARγ antagonist GW9662, confirming that PPARγ inhibits liver growth and hepatocyte proliferation specifically through the HGF/c-Met/ERK1/2 pathway .

Temporal Sequence Mapping:
Direct c-Met effects typically occur rapidly, with phosphorylation beginning within minutes of stimulation . By establishing detailed temporal sequences of signaling events, researchers can distinguish primary (direct) from secondary (indirect) effects based on their kinetics.

Genetic Rescue Experiments:
Complementing pharmacological approaches with genetic rescue experiments can provide definitive evidence of direct c-Met involvement. In c-Met mutant mice, liver regeneration is impaired and ERK1/2 phosphorylation is absent , demonstrating direct dependence of this pathway on c-Met.

Pathway Crossover Analysis:
Research has identified specific crossover points between signaling pathways. For example, BMP9 and HGF/c-Met signaling establish a signal crossover through ALK1 by modulating SMAD1 and p38MAPK . Targeted analysis of these nodes helps delineate where pathways directly interact versus where they operate independently.

These methodological approaches provide researchers with a framework for distinguishing direct c-Met-mediated effects from indirect influences or pathway crosstalk, enabling more precise understanding of c-Met's role in complex biological processes such as liver regeneration.

What are the most promising approaches for targeting c-Met in regenerative medicine applications?

Several promising approaches for targeting c-Met in regenerative medicine applications have emerged from recent research:

Recombinant HGF Administration:
Direct administration of recombinant human HGF (rh-HGF) has shown significant therapeutic potential. Studies demonstrate that rh-HGF can inhibit hepatocyte death and stabilize structural and vascular integrity in mice with acute liver failure . The method of administration matters significantly - injection of HGF into the portal vein has been shown to cause hepatocyte proliferation and hepatomegaly in normal rats and mice .

HGF Activator Therapy:
Recombinant human HGF-activator (rh-HGF-activator) administered via the portal vein significantly increases liver regeneration rates compared to control groups . This approach leverages the body's existing HGF while enhancing its activation state.

Engineered HGF Fragments:
Synthetic small molecules based on natural HGF fragments show promise. The engineered molecule 1K1, derived from the naturally occurring HGF fragment NK1, demonstrates anti-fibrotic properties and promotes liver regeneration in rodents with improved stability and production characteristics . Such engineered variants may overcome the pharmacokinetic and production challenges of full-length HGF.

Natural Compound Modulators:
Several natural compounds have shown efficacy in modulating HGF/c-Met signaling. Schisandra Chinensis and Resina Draconis significantly increase HGF expression levels, promoting liver function recovery and ameliorating acute liver injury by enhancing cell proliferation . These natural compounds may offer complementary approaches with potentially favorable safety profiles.

These diverse approaches highlight the therapeutic potential of targeting the HGF/c-Met axis in regenerative medicine, particularly for liver diseases where restoration of tissue function is critical.

How can researchers monitor potential off-target effects when modulating c-Met signaling in experimental models?

Monitoring potential off-target effects when modulating c-Met signaling requires comprehensive assessment strategies:

Broad Pathway Profiling:
Since c-Met activates multiple downstream pathways including JAK/STAT3, PI3K/Akt/NF-κB, and Ras/Raf , researchers should employ phospho-proteomic approaches to evaluate changes across these and other pathways simultaneously. This broad profiling can identify unexpected pathway activation or inhibition that may indicate off-target effects.

Tissue-Specific Analysis:
The effects of c-Met modulation may vary across tissues. Research has shown that HGF/c-Met signaling is essential for liver regeneration , but it also plays roles in other tissues. Multi-tissue analysis can identify off-target effects that may be restricted to specific organ systems.

Cell-Type Specificity Assessment:
Within tissues, different cell types may respond differently to c-Met modulation. For example, under hypoxic conditions, HGF expression in hepatic stellate cells and c-Met expression in hepatocytes both decrease dramatically . Single-cell analysis techniques can resolve cell-type-specific responses that might be missed in bulk tissue analysis.

Developmental Timing Considerations:
The biological functions of HGF/c-Met are extensive and diverse, particularly during embryonic development. Targeted destruction of HGF or c-Met leads to embryonic lethality in mice with specific damage to liver and placental development . Age-appropriate models should be used to avoid misinterpreting developmental effects as off-target effects.

By implementing these monitoring strategies, researchers can better differentiate between intended on-target effects and potential off-target consequences of c-Met modulation, enhancing both the safety and specificity of experimental interventions targeting this pathway.

What biomarkers most accurately reflect c-Met activation status in clinical samples?

Accurate assessment of c-Met activation status in clinical samples requires a multi-biomarker approach that reflects the complex dynamics of receptor activation:

Phosphorylated c-Met Residues:
The primary direct biomarkers are phosphorylated tyrosine residues on the c-Met receptor itself. After partial hepatectomy, c-Met tyrosine phosphorylation begins within 5 minutes and peaks at 60 minutes . Phospho-specific antibodies against these residues provide the most direct measurement of activation status.

ERK1/2 Phosphorylation:
Research has established that ERK1/2 activation is completely dependent on c-Met during liver regeneration . Therefore, phospho-ERK1/2 serves as a reliable downstream biomarker that reflects functional c-Met signaling, particularly in liver tissue samples.

Nuclear β-catenin Localization:
HGF binding to c-Met induces Wnt-independent nuclear translocation of β-catenin . Immunohistochemical assessment of nuclear β-catenin localization provides an additional readout of functional c-Met activation.

Age-Adjusted Reference Ranges:
Clinical studies have demonstrated that HGF and c-Met expression levels are significantly lower in older patients compared to younger patients . Biomarker interpretation therefore requires age-appropriate reference ranges to accurately assess activation status relative to biological norm.

These biomarkers, when assessed in combination, provide a comprehensive picture of c-Met activation status in clinical samples. This multi-parameter approach is particularly important given the rapid dynamics of c-Met activation and degradation, which might lead to false negatives if relying on a single biomarker at a single timepoint.

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 2025 TheBiotek. All Rights Reserved.