MET Human

Met Proto-Oncogene Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to MET Human

The MET human receptor, encoded by the MET gene (GeneID: 4233), is a receptor tyrosine kinase (RTK) critical for embryonic development, tissue repair, and signal transduction. It is activated by hepatocyte growth factor (HGF), triggering pathways involved in cell proliferation, survival, and migration. Aberrant activation of MET is implicated in oncogenesis, making it a therapeutic target in cancers such as kidney, liver, and lung tumors .

Domain Architecture

DomainFunctionKey Residues/Features
Sema domainBinds HGF; critical for receptor dimerizationBlades 2–3 interact with HGF β-chain .
PSI domainLinks Sema and IPT repeats; stabilizes extracellular interactionsCysteine-rich sequence .
IPT repeatsMediate receptor clustering and ligand recognitionImmunoglobulin-like folds .
Tyrosine kinase domain (TKD)Phosphorylates downstream targets (e.g., MAPK, PI3K/AKT)Y1234/Y1235: Autophosphorylation sites .

Physiological Roles

MET signaling is essential for:

  • Embryogenesis: Placental development, muscle formation, and neural tube morphogenesis .

  • Tissue repair: Wound healing, hepatocyte regeneration, and angiogenesis .

Oncogenic Activation

MET deregulation in cancers occurs via:

  1. Overexpression: Observed in gastric, lung, and breast cancers .

  2. Gene amplification: Seen in non-small cell lung cancer (NSCLC) and gastric adenocarcinomas .

  3. Mutations: Germline/somatic mutations in kinase or extracellular domains .

Table 1: MET Mutations and Associated Cancers

Mutation TypeLocationCancer TypeFunctional Impact
Exon 14 skippingSplice variantNSCLC, gastric, renalConstitutive activation; crizotinib resistance .
Y1235DKinase domainPapillary renal carcinomaHyperactivation; oncogenic transformation .
G1181RKinase domainNSCLC, gastricTKI resistance (e.g., capmatinib) .

Therapeutic Resistance

Acquired resistance to MET inhibitors (e.g., crizotinib) often involves secondary mutations:

  • D1246N/H: Reduces drug binding affinity .

  • Y1248H/S: Disrupts ATP-binding pocket .

Natural Compound Inhibitors

CompoundSourceMechanismEfficacy in Cancers
CurcuminTurmericInhibits MET phosphorylationGastric, breast .
ResveratrolGrapes, berriesSuppresses MET/STAT3 signalingLung, liver .
QuercetinCitrus, onionsDownregulates MET expressionProstate, colon .

Tissue Microarray Data

MET overexpression is observed in:

  • Lung cancers: Bronchioalveolar junctions co-express MET and stem cell factor (SCF) .

  • Gastric cancers: MET amplification correlates with sensitivity to PHA665752 .

Product Specs

Introduction
The mesenchymal-epithelial transition factor (c-MET) is a receptor tyrosine kinase with implications in oncogenesis. Its primary ligand, hepatocyte growth factor (HGF), is a heterodimer typically produced by mesenchymal cells. c-MET expression is generally restricted to stem, progenitor, and wound-healing cells in adults. However, it plays a crucial role in embryonic development, particularly in epithelial cell invasive growth and epithelial-mesenchymal transition (EMT). Dysregulation of the HGF/MET pathway is associated with various cancers, and c-MET mutations are linked to poor prognoses due to their potential to stimulate tumor growth, angiogenesis, and metastasis.
Description
This product consists of the recombinant human Met Proto-Oncogene (amino acids 1039-1345), produced in insect cells and with a molecular weight of 34.6 kDa. The protein is purified using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The MET protein is supplied in a solution of 50mM Tris, 300mM NaCl, and 10% Glycerol, at pH 7.5, with a concentration of 1mg/ml.
Stability
For optimal storage, keep the vial at 4°C if it will be used within 2-4 weeks. For longer-term storage, freeze the solution at -20°C. Repeated freezing and thawing of the product should be avoided.
Purity
The purity of this product is greater than 90.0%, as determined by SDS-PAGE analysis.
Synonyms
Hepatocyte growth factor receptor, HGF receptor, HGF/SF receptor, Proto-oncogene c-Met, Scatter factor receptor, SF receptor, Tyrosine-protein kinase Met, MET,HGFR, AUTS9, RCCP2.
Source
Insect cells.
Amino Acid Sequence
DSDISSPLLQNTVHIDLSALNPELVQAVQHVVIGPSSLIVHFNEVIGRGHFGCVYHGTL
LDNDGKKIHCAVKSLNRITDIGEVSQFLTEGIIMKDFSHPNVLSLLGICLRSEGSPLVVL
PYMKHGDLRNFIRNETHNPTVKDLIGFGLQVAKGMKYLASKKFVHRDLAARNCMLDE
KFTVKVADFGLARDMYDKEYYSVHNKTGAKLPVKWMALESLQTQKFTTKSDVWSFG
VLLWELMTRGAPPYPDVNTFDITVYLLQGRRLLQPEYCPDPLYEVMLKCWHPKAEM
RPSFSELVSRISAIFSTFI.

Q&A

What is the human MET oncogene and what is its significance in cancer research?

The human MET oncogene was originally isolated from a chemically transformed human cell line, MNNG-HOS. It's related to protein kinase oncogenes and growth factor receptors, with nucleotide sequencing showing it's more closely homologous to tyrosine kinases than to serine/threonine kinases . Within the tyrosine kinase family, MET domains are most closely related to the human insulin receptor and the viral abl gene . The significance of MET in cancer research stems from its chromosomal location at human chromosome 7 band 7q21-q31, a region associated with nonrandom chromosomal deletions observed in some patients with acute nonlymphocytic leukemia . Understanding MET is crucial for developing targeted therapies for cancers where this oncogene plays a role in tumor progression and metastasis. The gene's relationship to both cancer pathways and growth factor signaling makes it a valuable target for therapeutic intervention research.

How does the MET signaling pathway function in normal human cells versus cancer cells?

In normal human cells, the MET signaling pathway is tightly regulated and primarily functions in embryonic development, tissue regeneration, and wound healing. The pathway is activated when the hepatocyte growth factor (HGF) binds to the MET receptor, triggering autophosphorylation of tyrosine residues in the kinase domain and subsequent activation of downstream signaling cascades that regulate cell proliferation, survival, and motility . In cancer cells, aberrant MET signaling can occur through various mechanisms including gene amplification, overexpression, mutations, or inappropriate ligand production. These alterations lead to constitutive activation of the pathway, resulting in dysregulated cell growth, enhanced cell survival, increased invasiveness, and promotion of metastasis. The difference between normal and cancer cell signaling through MET offers opportunities for therapeutic targeting that exploits cancer-specific alterations while minimizing effects on normal cells.

What are the primary methods for detecting MET expression in human tissue samples?

Researchers typically employ multiple complementary techniques to detect MET expression in human tissue samples. Immunohistochemistry (IHC) remains the most widely used method for protein-level detection in clinical samples, allowing visualization of MET expression patterns within tissue architecture. Fluorescence in situ hybridization (FISH) is employed to detect MET gene amplification at the DNA level . Quantitative real-time PCR provides precise measurement of MET mRNA expression levels. For more in-depth analysis, Western blotting can quantify total and phosphorylated MET protein levels. Next-generation sequencing approaches enable comprehensive characterization of MET genetic alterations including mutations, amplifications, and rearrangements. Mass spectrometry-based proteomics offers detailed protein-level quantification and can identify post-translational modifications. When selecting detection methods, researchers should consider the specific research question, available sample types, and required sensitivity/specificity parameters.

How can researchers design experiments to study MET-dependent metabolite changes in human cancer models?

When designing experiments to study MET-dependent metabolite changes in human cancer models, researchers should employ a multi-faceted approach that combines genetic manipulation of MET expression with comprehensive metabolite profiling. Begin by establishing appropriate model systems including cell lines with genetic knockdown/knockout of MET using CRISPR-Cas9 or shRNA technologies alongside MET-overexpressing lines . Paired patient-derived xenograft models with varying MET expression levels can provide more translational relevance. For metabolite profiling, employ both targeted and untargeted metabolomic approaches using liquid chromatography-mass spectrometry (LC-MS) or gas chromatography-mass spectrometry (GC-MS) . Researchers should pay particular attention to four types of metabolites: active metabolites that exert pharmacological effects similar to the parent compound; reactive metabolites with electrophilic reactivity that might cause toxicity; disproportionate human metabolites with exposures substantially higher in humans compared to animal models; and unique human metabolites found only in human samples . Incorporating stable isotope tracing can help elucidate specific metabolic pathways affected by MET signaling. Statistical methods including principal component analysis and partial least squares discriminant analysis should be employed to identify significant metabolite changes correlated with MET activity.

What are the current challenges in addressing contradictions in MET Human research data?

Addressing contradictions in MET Human research data presents several significant challenges. Firstly, biological heterogeneity across cancer types and patient populations can lead to seemingly contradictory findings about MET's role . Different experimental models (cell lines, PDX models, transgenic animals) may yield inconsistent results due to varying MET dependency contexts. Technical variability in detection methods introduces another layer of complexity, as different antibodies for immunohistochemistry or varying thresholds for defining "MET positivity" contribute to discrepancies . Contradictions also arise from context-dependent MET signaling, where the oncogene may function differently depending on the presence of other genetic alterations or signaling pathway activities. To address these contradictions systematically, researchers should implement structured approaches that explicitly account for experimental variables and contexts . This includes standardizing detection methods across studies, carefully documenting experimental conditions, and utilizing meta-analysis techniques to integrate findings across multiple datasets. Computational modeling approaches can help reconcile seemingly contradictory data by identifying conditional dependencies and context-specific effects. Finally, robust statistical methods for handling inconsistent data, such as those developed for dialogue contradiction detection, can be adapted for research data analysis .

How can metabolite profiling and identification studies enhance understanding of MET-targeted therapeutics?

Metabolite profiling and identification studies significantly enhance understanding of MET-targeted therapeutics through comprehensive characterization of drug metabolism and identification of potential biomarkers for treatment response. These studies are critical for elucidating the metabolic fate of MET inhibitors and understanding how metabolites contribute to efficacy or toxicity profiles . In vitro MetID studies provide early insights into metabolic soft spots of compounds, potential reactive metabolites, and cross-species comparisons to guide toxicology species selection . This information helps optimize drug design by identifying structural modifications that might improve pharmacokinetic properties. In vivo MetID studies establish in vitro-in vivo correlation and compare metabolism across species to ensure reliable prediction of human metabolism . Particularly important is the evaluation of metabolite exposure in humans versus preclinical models to determine the appropriateness of selected toxicological species from the Metabolite in Safety Testing (MIST) perspective . Advanced metabolomic approaches can also identify downstream metabolic signatures associated with MET inhibition, potentially revealing resistance mechanisms or synergistic therapeutic targets. When integrated with pharmacodynamic and clinical response data, metabolite profiles may yield predictive biomarkers for patient stratification and treatment monitoring.

What methodologies are recommended for conducting human factors studies in MET research?

For conducting human factors studies in MET research, a comprehensive methodology incorporating both formative and summative evaluations is recommended. Formative evaluations should be implemented early in the research process using product prototypes to gather specific user feedback for design development . As the research progresses toward final design stages, late-stage formative evaluations structured more like summative testing should focus on end-to-end testing . For early-stage formative evaluations, methodologies should include contextual inquiry/ethnographic research to understand user behavior in natural settings . When transitioning to summative evaluation, researchers should test the final production equivalent design with participants given specific use scenarios and tasks, with all sessions video recorded for detailed analysis . This approach ensures that all design controls and user interface requirements effectively eliminate or reduce risk. Human factors studies should follow established MHRA and FDA Guidelines to maintain compliance standards . For complex systems involving software components, such as those found in intensive care monitors, ventilators, and administration equipment, usability studies should combine risk analysis with iterative design in a comprehensive usability engineering process . The goal throughout should be to demonstrate device performance in environments that accurately simulate real-world usage conditions while identifying any safety or application problems before clinical trials.

How do researchers effectively analyze contradictions in human-MET interaction data?

To effectively analyze contradictions in human-MET interaction data, researchers should implement a structured approach that explicitly accounts for the natural structure of human interactions. Rather than using unstructured approaches where all interaction data is simply concatenated for analysis, a structured utterance-based approach that pairs interactions separately before analysis has proven more robust and transferable to out-of-distribution scenarios . This methodology is particularly important when analyzing potential contradictions in how humans interact with medical technologies or experimental protocols. When implementing this approach, researchers should create a framework similar to the DialoguE COntradiction DEtection task (DECODE) methodology, which involves carefully collecting and analyzing instances where contradictions occur . Analysis should distinguish between different types of contradictions, such as those related to procedural understanding, risk interpretation, or technical comprehension. Transformer-based models trained on contradiction detection can be helpful, but only when explicitly structured to account for the sequential and contextual nature of human-technology interactions . For rigorous evaluation, researchers should develop both in-distribution test sets based on controlled study conditions and out-of-distribution test sets that reflect real-world variability, particularly focusing on scenarios where contradictions might lead to safety concerns or data integrity issues .

What are the best practices for designing human factors studies for MET-based diagnostic devices?

Best practices for designing human factors studies for MET-based diagnostic devices should follow a systematic approach that evaluates usability throughout the development lifecycle. Begin with a heuristic review conducted by human factors experts to identify potential usability issues early in the design process . This should be followed by formative evaluations using representative user groups to gather feedback on prototype designs. When selecting participants, ensure diversity across relevant demographics, experience levels, and physical capabilities to represent the full spectrum of intended users . Study environments should simulate the actual context of use, whether clinical settings, laboratories, or point-of-care locations. Define clear, measurable success criteria for critical tasks based on intended use and risk analysis. For data collection, employ a mixed-methods approach combining quantitative metrics (time on task, error rates, success rates) with qualitative assessments (think-aloud protocols, post-task interviews, satisfaction ratings) . Video recording of sessions is essential for comprehensive analysis of user interactions . Final summative evaluation should be conducted on production-equivalent devices, focusing on end-to-end testing with realistic use scenarios . Throughout the process, maintain detailed documentation of methodologies, participant characteristics, results, and design iterations to satisfy regulatory requirements. The entire study should adhere to ethical standards for human subjects research while following MHRA and FDA guidelines for medical device human factors studies .

What quantitative research methodologies are most effective for studying MET oncogene expression patterns?

The most effective quantitative research methodologies for studying MET oncogene expression patterns employ a multi-modal approach combining genomic, transcriptomic, and proteomic techniques with robust statistical analysis. Digital droplet PCR (ddPCR) offers superior precision for absolute quantification of MET gene copy numbers, while RNA sequencing provides comprehensive transcriptome-wide expression analysis with the ability to detect splice variants . For protein-level quantification, reverse phase protein arrays (RPPA) and quantitative immunofluorescence enable high-throughput analysis across large sample cohorts with precise signal quantification . Single-cell RNA sequencing is particularly valuable for characterizing cellular heterogeneity in MET expression within tumor microenvironments. When analyzing the resulting data, researchers should employ appropriate statistical methods including non-parametric tests for non-normally distributed expression data and multiple comparison corrections when assessing expression across various tissue types or treatment conditions . Multivariate analysis techniques such as principal component analysis and hierarchical clustering help identify patterns in expression data correlated with clinical outcomes. Computational approaches including machine learning algorithms can identify complex relationships between MET expression patterns and other molecular features or clinical parameters . Longitudinal studies tracking MET expression changes over time, particularly in response to treatments, provide dynamic information not captured in single timepoint analyses.

How should researchers design mixed-method studies to investigate both molecular and clinical aspects of MET Human research?

When designing mixed-method studies to investigate both molecular and clinical aspects of MET Human research, researchers should implement a sequential explanatory design that integrates quantitative molecular data with qualitative clinical observations . Begin by clearly defining research questions that address both molecular mechanisms and clinical implications, then develop a structured research methodology that incorporates appropriate methods for each domain . For the molecular component, employ high-throughput genomic, transcriptomic, and proteomic techniques to quantitatively characterize MET alterations, expression patterns, and downstream signaling effects. This should include targeted sequencing of MET and related pathway genes, RNA sequencing for expression analysis, and proteomic approaches to evaluate activation status . For the clinical component, collect comprehensive data on patient characteristics, treatment responses, progression patterns, and survival outcomes. Patient-reported outcomes should be systematically gathered using validated instruments. The integration phase is critical - employ statistical methods that can correlate molecular findings with clinical outcomes, such as multivariate regression models, survival analyses, and machine learning approaches . Triangulation between molecular and clinical data should be facilitated through regular multidisciplinary team discussions including molecular biologists, clinical oncologists, pathologists, and biostatisticians. This collaborative approach ensures that molecular findings are interpreted in clinically relevant contexts and that unusual clinical observations prompt targeted molecular investigations .

What sampling approaches are most appropriate for MET expression studies in diverse human populations?

For MET expression studies in diverse human populations, researchers should implement a strategic multi-level sampling approach that ensures both statistical power and comprehensive representation. Begin with a stratified random sampling design that accounts for key demographic and clinical variables including age, sex, ethnicity, geographical region, and disease subtype . Sample size calculations should consider the expected effect size of MET expression differences between subgroups, with power analysis conducted to determine minimum cohort sizes needed for detecting clinically meaningful variations . For rare populations or underrepresented groups, purposive sampling may be necessary to ensure adequate representation. When studying genetic determinants of MET expression, ancestry-informed sampling is essential, utilizing ancestry informative markers or genome-wide data to characterize genetic background beyond self-reported ethnicity . Temporal sampling considerations are also important - longitudinal sampling enables assessment of MET expression changes over time, particularly relevant for treatment response studies. For tissue sampling, standardized protocols must address pre-analytical variables including ischemia time, fixation methods, and storage conditions that can affect MET detection . Digital pathology approaches with automated quantification can reduce observer bias in expression analysis. Researchers should also consider sampling from multiple tumor regions to account for intratumoral heterogeneity in MET expression. Finally, all sampling approaches should adhere to ethical guidelines for human subjects research, with appropriate informed consent procedures and institutional review board approvals .

What statistical approaches are recommended for analyzing metabolite data in MET-focused research?

For analyzing metabolite data in MET-focused research, researchers should employ a comprehensive statistical framework that addresses the complex, multivariate nature of metabolomic datasets. Initial data preprocessing is critical and should include normalization procedures to account for technical variations, such as intensity normalization, internal standard normalization, or probabilistic quotient normalization . Missing value imputation strategies should be carefully selected based on the missing data mechanism. For exploratory analysis, unsupervised methods including principal component analysis (PCA) and hierarchical clustering help visualize natural groupings and identify outliers without imposing a priori assumptions . When testing specific hypotheses about MET-related metabolite changes, supervised methods such as partial least squares discriminant analysis (PLS-DA) or orthogonal projections to latent structures (OPLS) can identify discriminatory metabolites while accounting for confounding variables . For hypothesis testing of individual metabolites, appropriate multiple testing corrections (Benjamini-Hochberg or similar methods) must be applied to control false discovery rates. Pathway enrichment analysis tools help contextualize findings within biological systems, identifying metabolic pathways significantly affected by MET alterations . For longitudinal metabolite data, mixed-effects models can account for repeated measures while assessing time-dependent changes. Network analysis approaches, including correlation networks and Gaussian graphical models, reveal complex relationships between metabolites and their association with MET expression or activity . All statistical analyses should include appropriate validation strategies, such as cross-validation or independent test sets, to ensure robustness of findings.

How can machine learning approaches enhance the interpretation of complex MET Human research data?

Machine learning approaches significantly enhance interpretation of complex MET Human research data by uncovering patterns and relationships that traditional statistical methods might miss. For MET research applications, supervised learning algorithms including random forests, support vector machines, and gradient boosting can classify samples based on MET expression patterns and predict clinical outcomes such as treatment response or prognosis . Deep learning approaches, particularly convolutional neural networks applied to histopathology images, can quantify MET expression and localization patterns from tissue samples with greater consistency than manual scoring . Unsupervised learning methods including autoencoders and self-organizing maps help identify novel molecular subtypes characterized by distinct MET-related features without relying on predefined classifications. For integrating multi-omics data (genomic, transcriptomic, proteomic, and metabolomic), techniques such as similarity network fusion and multi-view clustering create unified representations that capture complementary aspects of MET biology . Natural language processing algorithms can extract MET-related information from unstructured clinical notes and research literature, enabling knowledge synthesis across thousands of documents . Interpretability is crucial - techniques such as SHAP (SHapley Additive exPlanations) values and LIME (Local Interpretable Model-agnostic Explanations) help identify the features driving model predictions, translating complex models into biologically meaningful insights . Researchers should implement rigorous validation protocols including external validation on independent datasets and assessment of model calibration to ensure reliable performance in new research contexts.

What methodologies help researchers identify and resolve contradictions in MET signaling pathway data?

To identify and resolve contradictions in MET signaling pathway data, researchers should implement a comprehensive methodology that combines structured data comparison with mechanistic validation. Begin by constructing a formal ontology of MET signaling components and interactions, providing a standardized framework for comparing findings across studies . For each apparent contradiction, implement a structured approach similar to the DialoguE COntradiction DEtection task (DECODE) methodology, explicitly pairing contradictory findings and analyzing contextual factors that might explain differences . Create a contradiction resolution matrix that categorizes discrepancies based on potential sources: experimental conditions (cell types, time points, stimulation protocols), detection methods (antibodies, activity assays, readout systems), or biological contexts (genetic background, microenvironment factors). For methodological contradictions, conduct head-to-head comparisons using standardized systems to determine which finding is more reproducible. For biological contradictions, design experiments specifically testing context-dependency hypotheses, such as genetic dependency screens in diverse cell lines or conditional knockout models . Network modeling approaches, including causal Bayesian networks and logic-based models, can integrate seemingly contradictory data by identifying conditional dependencies that explain context-specific behaviors. Time-resolved studies of MET signaling may reveal temporal dynamics that reconcile apparently contradictory steady-state observations. Finally, single-cell approaches can determine whether population-level contradictions reflect differences in cellular heterogeneity rather than true biological disagreement .

How can researchers validate contradictory findings about MET oncogene function across different experimental models?

Validating contradictory findings about MET oncogene function across different experimental models requires a systematic cross-validation approach that bridges in vitro, in vivo, and clinical contexts. First, create a standardized experimental framework where identical genetic manipulations of MET (CRISPR knockouts, point mutations, or overexpression systems) are implemented across multiple model systems including cell lines, organoids, and animal models . Ensure genetic backgrounds are well-characterized, as modifier genes may explain context-dependent MET functions. For each contradictory finding, design parallel experiments that maintain identical conditions except for the model system, allowing direct comparison of MET effects . Employ multi-modal readouts measuring the same endpoints (proliferation, migration, signaling) using different methodologies to confirm findings are not artifacts of particular assay systems. Dose-response studies are particularly important, as contradictions may reflect threshold effects rather than true biological differences. For in vivo studies, utilize both xenograft and genetically engineered mouse models with conditional MET alterations to evaluate tissue-specific effects . Patient-derived models including PDX and organoids directly link experimental findings to clinical samples. Single-cell approaches can determine whether apparent contradictions reflect cellular heterogeneity, with different cell populations responding differently to MET alterations . Finally, translate findings to human clinical samples using digital spatial profiling, multiplex immunofluorescence, or similar technologies that can correlate MET activity with downstream effects in preserved tissue architecture. This comprehensive cross-validation approach can determine whether contradictions reflect true biological complexity or methodological artifacts.

What approaches can identify reliable biomarkers for MET-targeted therapies despite contradictory research data?

To identify reliable biomarkers for MET-targeted therapies despite contradictory research data, researchers should implement a convergent validation approach that integrates multiple data types while accounting for contextual factors. Begin with a systematic review of contradictory biomarker findings, categorizing discrepancies based on study characteristics, patient populations, and analytical methods . Implement meta-analysis techniques specifically designed to handle heterogeneity, such as random-effects models and subgroup analyses, to identify consistent biomarker signals across diverse studies . For prospective biomarker validation, design studies with pre-specified analysis plans that include multiple candidate biomarkers evaluated simultaneously, enabling direct comparison of predictive performance. Multi-parameter biomarker approaches are particularly valuable when single markers show contradictory results - combining genomic, transcriptomic, and protein-based markers into integrated signatures often yields more robust prediction than any single marker . Longitudinal sampling before, during, and after MET-targeted therapy can identify dynamic biomarkers that better reflect treatment response than baseline measurements alone. Machine learning algorithms trained on multi-omics data can discover complex biomarker patterns not evident in single-marker analyses, with ensemble methods particularly useful for integrating contradictory signals . To address biological context, implement biomarker assessment in defined molecular subtypes, as MET dependency may vary across tumor contexts. Functional validation is essential - techniques such as CRISPR screening or pharmacologic inhibition can directly test whether biomarker-defined populations show differential dependence on MET signaling . Finally, establish multi-institutional consortia for biomarker validation using standardized assays, shared sample repositories, and harmonized clinical data to generate the large, consistent datasets needed to resolve contradictions.

What emerging technologies are revolutionizing the study of MET Human interactions in precision medicine?

Emerging technologies are fundamentally transforming MET Human research in precision medicine, creating unprecedented opportunities for therapeutic targeting and patient stratification. Single-cell multi-omics approaches now allow simultaneous profiling of MET genomic alterations, expression levels, and downstream pathway activation within individual cells, revealing previously hidden heterogeneity in MET signaling dynamics . Spatial transcriptomics and proteomics technologies, including GeoMx Digital Spatial Profiling and Visium Spatial Gene Expression, are providing critical insights into MET activity within preserved tissue architecture, showing how MET-expressing cells interact with the tumor microenvironment . CRISPR-based functional genomics screens have evolved to enable high-throughput assessment of synthetic lethal interactions with MET alterations, identifying novel combination therapy opportunities and resistance mechanisms . In the clinical domain, liquid biopsy approaches incorporating circulating tumor DNA, extracellular vesicles, and circulating tumor cells allow non-invasive monitoring of MET alterations during treatment, enabling dynamic treatment adjustments . Artificial intelligence platforms integrating radiomics with molecular data are identifying imaging signatures that correlate with MET pathway activation, potentially offering non-invasive biomarkers. For therapeutic development, proteolysis-targeting chimeras (PROTACs) directed against MET represent a paradigm shift, inducing degradation rather than just inhibition of the receptor . Patient-derived organoid platforms enable rapid ex vivo testing of MET-targeted therapies against individual patient samples, facilitating truly personalized treatment selection. These technological advances collectively enable a systems biology approach to MET research, viewing the oncogene within its full biological context rather than in isolation.

What research methodology trends are shaping the future of integrated MET Human studies?

The future of integrated MET Human studies is being shaped by several transformative research methodology trends that emphasize holistic understanding across scales and contexts. Team science approaches featuring multidisciplinary collaborations between molecular biologists, clinical oncologists, computational scientists, and patient advocates are becoming essential for comprehensive MET research programs . These collaborations are increasingly formalized through consortia with standardized protocols and shared resources. Methodologically, the integration of real-world evidence with traditional clinical trial data is bridging the gap between controlled research environments and clinical practice, providing insights into MET-targeted therapies' effectiveness in diverse patient populations . Digital health technologies including wearable devices and mobile applications are enabling continuous remote monitoring of patient outcomes in MET-targeted therapy trials, capturing richer data on treatment responses and adverse events. Multi-scale integration methodologies are emerging that connect molecular MET signaling to cellular behaviors, tissue organization, and ultimately patient outcomes through hierarchical modeling approaches . Adaptive trial designs with biomarker-driven treatment assignment are accelerating the clinical translation of MET research findings, while master protocol frameworks allow simultaneous evaluation of multiple MET-targeted agents or combinations. Computational approaches including digital twins of individual patients' tumors may enable in silico prediction of responses to MET-targeted therapies before actual treatment . Patient-partnered research methodologies are increasingly incorporating patient priorities and experiences into study design and outcome selection, ensuring MET research addresses endpoints meaningful to those most affected. These methodology trends collectively represent a shift toward more integrated, adaptive, and patient-centered research paradigms that better capture the complexity of MET biology in human disease.

Product Science Overview

Introduction

The Met proto-oncogene, also known as the hepatocyte growth factor receptor, is a member of the receptor tyrosine kinase family. It plays a crucial role in various cellular processes, including growth, survival, and migration. The human recombinant form of this protein is of significant interest in both research and therapeutic contexts due to its involvement in cancer and other diseases.

Structure and Function

The Met proto-oncogene encodes a receptor tyrosine kinase that binds to its ligand, hepatocyte growth factor (HGF). This interaction induces dimerization and activation of the receptor, which subsequently triggers a cascade of downstream signaling pathways. These pathways are involved in cellular processes such as proliferation, differentiation, and motility .

The Met receptor is a heterodimer composed of an alpha and a beta subunit linked by disulfide bonds. The alpha subunit is extracellular, while the beta subunit spans the membrane and contains the intracellular tyrosine kinase domain .

Role in Cancer

Alterations in the Met proto-oncogene, such as gene amplification, mutations, and overexpression, have been implicated in various cancers, including renal cell carcinoma, hepatocellular carcinoma, and head and neck cancers . These alterations can lead to constitutive activation of the Met receptor, driving tumor growth, invasion, and metastasis .

Preparation Methods

The human recombinant Met proto-oncogene is typically produced using recombinant DNA technology. This involves cloning the Met gene into an expression vector, which is then introduced into a suitable host cell line, such as Escherichia coli or mammalian cells. The host cells are cultured under specific conditions to express the recombinant protein, which is subsequently purified using techniques such as affinity chromatography .

Synthetic Routes and Reaction Conditions

The production of recombinant Met proto-oncogene involves several steps, including gene cloning, expression, and purification. The gene is first cloned into an expression vector, which is then introduced into host cells. The cells are cultured under optimal conditions to ensure high-level expression of the recombinant protein. The protein is then purified using various chromatographic techniques to achieve the desired purity and activity .

Industrial Production Methods

Industrial production of recombinant Met proto-oncogene typically involves large-scale fermentation processes. Host cells expressing the recombinant protein are cultured in bioreactors under controlled conditions to maximize yield. The protein is then harvested and purified using scalable purification methods, such as affinity and ion-exchange chromatography .

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.