KIT Human

KIT Proto-Oncogene Receptor Tyrosine Human Recombinant
Shipped with Ice Packs
In Stock

Description

KIT produced in Sf9 Insect cells is a single, glycosylated polypeptide chain containing 507 amino acids (26-524 a.a.) and having a molecular mass of 57.1kDa (Molecular size on SDS-PAGE will appear at approximately 50-70kDa).
KIT is expressed with an 8 amino acid His tag at C-Terminus and purified by proprietary chromatographic techniques.

Product Specs

Introduction
KIT, also known as KIT Proto-Oncogene Receptor Tyrosine, is a cytokine receptor expressed in hematopoietic stem cells and other cell types. KIT binds to receptor tyrosine kinase type III, a stem cell factor also called "rigid factor" or "c-kit ligand." Upon binding to stem cell factor (SCF), the receptor dimerizes, activating intrinsic tyrosine kinase activity. This activation triggers the phosphorylation and activation of signaling molecules, generating cellular signals. This receptor plays a role in protecting vascular smooth muscle cells from apoptosis and restoring cardiac function after myocardial infarction.
Description
Produced in Sf9 Insect cells, KIT is a single, glycosylated polypeptide chain consisting of 507 amino acids (26-524 a.a.). It has a molecular mass of 57.1kDa, though it appears as approximately 50-70kDa on SDS-PAGE. The protein is expressed with an 8 amino acid His tag at the C-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
Colorless, sterile-filtered solution.
Formulation
The KIT protein solution has a concentration of 0.25mg/ml and contains Phosphate Buffered Saline (pH 7.4) and 10% glycerol.
Stability
For use within 2-4 weeks, store at 4°C. For longer storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is greater than 90.0% as determined by SDS-PAGE.
Synonyms

Mast/stem cell growth factor receptor Kit, SCFR, Piebald trait protein, PBT, Proto-oncogene c-Kit, Tyrosine-protein kinase Kit, p145 c-kit, v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog, CD117.

Source

Sf9, Baculovirus.

Amino Acid Sequence

QPSVSPGEPS PPSIHPGKSD LIVRVGDEIR LLCTDPGFVK WTFEILDETN ENKQNEWITE KAEATNTGKY TCTNKHGLSN SIYVFVRDPA KLFLVDRSLY GKEDNDTLVR CPLTDPEVTN YSLKGCQGKP LPKDLRFIPD PKAGIMIKSV KRAYHRLCLH CSVDQEGKSV LSEKFILKVR PAFKAVPVVS VSKASYLLRE GEEFTVTCTI KDVSSSVYST WKRENSQTKL QEKYNSWHHG DFNYERQATL TISSARVNDS GVFMCYANNT FGSANVTTTL EVVDKGFINI FPMINTTVFV NDGENVDLIV EYEAFPKPEH QQWIYMNRTF TDKWEDYPKS ENESNIRYVS ELHLTRLKGT
EGGTYTFLVS NSDVNAAIAF NVYVNTKPEI LTYDRLVNGM LQCVAAGFPE PTIDWYFCPG TEQRCSASVL PVDVQTLNSS GPPFGKLVVQ SSIDSSAFKH NGTVECKAYN DVGKTSAYFN FAFKGNNKEQ IHPHTLFTPL EHHHHHH.

Q&A

What is the role of KIT kinase in human cellular processes?

KIT kinase catalyzes the transfer of phosphate groups from ATP to target proteins, modifying their charge and shape to alter cellular activity. This phosphorylation affects protein synthesis, cell division, signal transduction, and cell growth. Methodologically, researchers can investigate KIT's role through:

  • Phosphorylation assays using purified KIT protein and candidate substrates

  • Selective KIT inhibition in cellular models followed by phosphoproteomics

  • Genetic knockout/knockdown studies with rescue experiments

  • Co-immunoprecipitation studies to identify binding partners

When designing experiments to study KIT function, it's essential to include appropriate controls that account for the inherent promiscuity of kinase activity, as most kinase inhibitors affect multiple targets simultaneously .

How can researchers accurately measure KIT expression levels?

Rather than simply defining detection methods, researchers should consider these methodological approaches:

Quantitative Measurement Protocol Selection:

  • qRT-PCR for mRNA expression (with validation of at least 3 reference genes)

  • Western blotting with densitometry (using antibodies validated for specificity)

  • Flow cytometry for surface expression (requiring appropriate gating strategies)

  • Immunohistochemistry with quantitative image analysis

  • Mass spectrometry-based proteomics for absolute quantification

Comparative Method Performance Table:

MethodSensitivitySpecificitySample RequirementTime RequiredCost
qRT-PCRHighMediumLow (ng RNA)4-6 hours$
Western BlotMediumHighMedium (μg protein)1-2 days$$
Flow CytometryHighHighMedium (10⁵-10⁶ cells)2-3 hours$$
IHCMediumMedium-HighLow (tissue section)1-2 days$$
MS ProteomicsVery HighVery HighHigh (μg-mg protein)2-5 days$$$$

When validating expression data, it's advisable to use at least two orthogonal methods, as different techniques capture distinct aspects of gene expression regulation .

What experimental models are most appropriate for studying KIT function?

Methodological approach to model selection should consider:

  • Cell Line Selection: Choose based on endogenous KIT expression or receptivity to genetic manipulation

    • Ba/F3 cells (for dependency studies)

    • HMC-1 (human mast cell leukemia with constitutive KIT activation)

    • GIST cell lines (with various KIT mutations)

  • Animal Models:

    • Genetically engineered mouse models with KIT mutations

    • Patient-derived xenografts

    • Zebrafish models for developmental studies

  • Primary Human Samples:

    • Fresh tumor biopsies

    • Bone marrow aspirates

    • Peripheral blood mononuclear cells

Researchers should select models based on their specific research question, considering the biological context and whether the goal is to study physiological function or disease-associated mutations1.

How can researchers resolve contradictory data regarding KIT inhibition effects?

Methodological framework for resolving contradictions:

  • Systematic Inhibitor Cross-Profiling:

    • Profile all inhibitors against at least 200 kinases to identify off-target effects

    • Implement thermal shift assays to confirm direct binding

    • Utilize at least two structurally distinct inhibitors targeting KIT

  • Multi-modal Validation Approach:

    • Combine pharmacological inhibition with genetic approaches

    • Verify on-target engagement using cellular thermal shift assays (CETSA)

    • Implement phosphoproteomic analysis to confirm pathway inhibition

  • Contextual Factors Analysis:

    • Systematically vary experimental conditions (serum concentration, cell density)

    • Test across multiple cell types and genetic backgrounds

    • Consider temporal dynamics of signaling pathways

When faced with contradictory results, researchers should implement a kinase chemogenomics approach similar to that described in the PKIS (Published Kinase Inhibitor Set) study, which enables a more comprehensive understanding of inhibitor selectivity profiles .

What methodological considerations are critical when studying KIT mutations in patient samples?

Researchers should implement a structured approach:

  • Sample Collection and Processing:

    • Flash-freezing of samples within 30 minutes of collection

    • Validation of tumor cell content (>40% recommended)

    • Matched normal tissue controls for germline comparison

  • Mutation Detection Strategy:

    • Targeted sequencing panels covering all KIT exons including intronic boundaries

    • Droplet digital PCR for known hotspot mutations

    • RNA-seq to detect fusion events and aberrant splicing

    • Low-frequency variant detection requiring minimum 500x coverage

  • Functional Validation Protocol:

    • Reconstitution of mutations in model systems

    • Phosphorylation status assessment of downstream targets

    • Drug sensitivity profiling using concentration matrices

    • In silico structural modeling to predict impact on protein conformation

KIT Mutation Analysis Workflow:

Sample CollectionQuality ControlDNA/RNA ExtractionSequencingVariant CallingAnnotationFunctional ValidationClinical Correlation\begin{array}{l} \text{Sample Collection} \rightarrow \text{Quality Control} \rightarrow \text{DNA/RNA Extraction} \\ \downarrow \\ \text{Sequencing} \rightarrow \text{Variant Calling} \rightarrow \text{Annotation} \\ \downarrow \\ \text{Functional Validation} \rightarrow \text{Clinical Correlation} \end{array}

This methodological framework ensures comprehensive mutation characterization beyond simple identification, providing insights into functional impacts and therapeutic implications .

How can phosphoproteomics be optimized for KIT pathway analysis?

Methodological optimization strategy:

  • Sample Preparation Protocol:

    • Rapid lysis in phosphatase inhibitor-containing buffers

    • Optimized protein:phosphopeptide enrichment ratios

    • Sequential enrichment using TiO₂ and IMAC

    • Sample multiplexing with isobaric labeling (TMT or iTRAQ)

  • Temporal Dynamics Assessment:

    • Implement time-resolved phosphoproteomics (30s, 2min, 5min, 15min, 30min post-stimulation)

    • Pulse-chase experiments with stable isotope labeling

    • Parallel analysis of phosphatase activity

  • Data Analysis Framework:

    • Implement computational pipelines that integrate protein-protein interaction networks

    • Apply machine learning for pathway recognition

    • Utilize kinase substrate enrichment analysis (KSEA)

    • Apply pathway topology algorithms to identify feedback and feedforward loops

  • Validation Strategy:

    • Confirm key phosphorylation events with phospho-specific antibodies

    • Perform site-directed mutagenesis of critical phosphosites

    • Correlate phosphorylation changes with phenotypic outcomes

This integrated approach extends beyond simple identification of phosphorylated proteins to building dynamic models of KIT signaling networks1 .

What computational methods best predict KIT inhibitor specificity and off-target effects?

Methodological framework for computational prediction:

  • Structure-Based Approaches:

    • Molecular docking with ensemble methods

    • Molecular dynamics simulations (minimum 100ns)

    • MM-GBSA binding energy calculations

    • Quantum mechanical calculations for transition states

  • Machine Learning Implementation:

    • Deep neural networks trained on kinome-wide activity data

    • Graph convolutional networks for capturing structural similarities

    • Transfer learning from related kinase families

    • Uncertainty quantification for confidence assessment

  • Integration with Experimental Data:

    • Bayesian model updating with experimental feedback

    • Active learning approaches to guide experimental design

    • Correlation analysis with thermal shift profiles

    • Integration of chemogenomic data from broad kinase inhibitor profiling

Prediction Model Performance Comparison:

MethodAccuracyPrecisionRecallF1 ScoreComputational Cost
Docking0.75-0.850.70-0.800.65-0.750.67-0.77Medium
MD + MM-GBSA0.80-0.900.75-0.850.70-0.800.72-0.82High
ML (2D descriptors)0.70-0.800.65-0.750.75-0.850.70-0.80Low
Deep Learning0.85-0.950.80-0.900.80-0.900.80-0.90Medium-High
Integrated Methods0.90-0.950.85-0.950.85-0.950.85-0.95Very High

This comprehensive computational strategy allows researchers to move beyond simple binding predictions to building multi-dimensional models of kinase-inhibitor interactions .

How should researchers design experiments to identify novel KIT-targeted therapeutics?

Methodological framework for therapeutic discovery:

  • High-Throughput Screening Design:

    • Implementation of ATP-competitive binding assays

    • Cell-based phenotypic screens with KIT-dependent lines

    • Fragment-based screens using thermal shift assays

    • DNA-encoded library screening against purified KIT domains

  • Medicinal Chemistry Optimization Strategy:

    • Establish structure-activity relationships across multiple kinase family members

    • Design selective compounds by targeting non-conserved residues

    • Implement parallel synthesis of at least 30 analogs per scaffold

    • Profile lead compounds across 250+ kinases to establish selectivity

  • Translational Validation Protocol:

    • Test compounds in patient-derived models

    • Implement pharmacodynamic biomarker development

    • Establish in vivo exposure-response relationships

    • Predict therapeutic window based on selectivity profile

Researchers should adopt a kinase chemogenomics approach, leveraging the inherent promiscuity of kinase inhibitors to understand both on-target and off-target effects, similar to the approach described in the GSK Published Kinase Inhibitor Set (PKIS) .

What controls are essential when studying KIT signaling pathways?

Methodological approach to experimental controls:

  • Genetic Controls:

    • CRISPR knockout/knockdown of KIT

    • Expression of kinase-dead KIT mutants

    • Rescue experiments with inhibitor-resistant KIT variants

    • Isogenic cell lines differing only in KIT status

  • Pharmacological Controls:

    • Use of structurally distinct KIT inhibitors

    • Titration of inhibitors to establish dose-response relationships

    • Application of inactive control compounds with similar physicochemical properties

    • Treatment with inhibitors targeting parallel pathways

  • Experimental Design Controls:

    • Time-course experiments to capture signaling dynamics

    • Appropriate vehicle controls for all treatments

    • Stimulation controls (SCF ligand vs. baseline)

    • Pathway cross-talk assessment through combinatorial inhibition

This systematic control framework ensures that observed effects can be confidently attributed to KIT activity rather than experimental artifacts or off-target effects1 .

How can researchers distinguish between KIT-dependent and KIT-independent effects in experimental models?

Methodological discrimination approach:

  • Genetic Perturbation Matrix:

    • Implement CRISPR knockout/knockdown in multiple cell backgrounds

    • Express constitutively active vs. dominant negative KIT variants

    • Utilize inducible systems for temporal control of KIT expression

    • Perform epistasis experiments with downstream pathway components

  • Pharmacological Discrimination Strategy:

    • Apply inhibitor matrices covering different selectivity profiles

    • Implement target engagement assays (e.g., CETSA)

    • Correlate phenotypic effects with biochemical inhibition metrics

    • Utilize the inherent promiscuity of kinase inhibitors strategically

  • Multi-omics Integration Framework:

    • Compare transcriptional signatures between genetic and pharmacological perturbation

    • Implement time-resolved phosphoproteomics to track signaling dynamics

    • Analyze metabolic changes associated with KIT inhibition

    • Develop computational models to distinguish direct vs. indirect effects

By implementing this comprehensive approach, researchers can move beyond correlative observations to establish causal relationships between KIT activity and observed phenotypes1 .

What are the recommended approaches for studying KIT interactions with other kinases?

Methodological framework for interaction studies:

  • Protein-Protein Interaction Assessment:

    • Implement proximity ligation assays in intact cells

    • Utilize FRET/BRET biosensors for real-time monitoring

    • Apply BioID or APEX2 proximity labeling

    • Perform co-immunoprecipitation with quantitative MS readout

  • Functional Interaction Analysis:

    • Conduct systematic genetic interaction screens (e.g., CRISPR combinatorial knockouts)

    • Implement small molecule matrix screening across kinase families

    • Apply synthetic lethality approaches

    • Analyze epistatic relationships through pathway perturbation

  • Network Modeling Strategy:

    • Develop Bayesian network models of kinase crosstalk

    • Implement ordinary differential equation models for dynamic simulation

    • Apply logic-based modeling for qualitative relationship mapping

    • Integrate phosphoproteomic data for network inference

This comprehensive approach allows researchers to move beyond studying KIT in isolation to understanding its functional place within the complex kinome network .

Product Science Overview

Introduction

The KIT proto-oncogene receptor tyrosine kinase, also known as CD117 or c-Kit, is a protein encoded by the KIT gene in humans. This receptor tyrosine kinase plays a crucial role in various cellular processes, including cell survival, proliferation, and differentiation .

Discovery and Structure

The KIT gene was first described by the German biochemist Axel Ullrich in 1987 as the cellular homolog of the feline sarcoma viral oncogene v-kit . The KIT protein consists of several domains:

  • Extracellular domain: Composed of five immunoglobulin-like domains.
  • Transmembrane domain: Anchors the receptor in the cell membrane.
  • Juxtamembrane domain: Located just inside the cell membrane.
  • Intracellular tyrosine kinase domain: Responsible for the receptor’s kinase activity .
Function

KIT is a cytokine receptor expressed on the surface of hematopoietic stem cells and other cell types. It binds to its ligand, the stem cell factor (SCF), also known as “steel factor” or "c-kit ligand" . Upon binding to SCF, KIT forms a dimer, activating its intrinsic tyrosine kinase activity. This activation leads to the phosphorylation of various signal transduction molecules, propagating the signal within the cell .

Role in Cellular Processes

KIT signaling is essential for several cellular processes:

  • Hematopoiesis: The formation of blood cellular components.
  • Melanogenesis: The production of melanin in melanocytes.
  • Gametogenesis: The development of gametes (sperm and eggs).
  • Mast cell development: Involves the migration and function of mast cells .
Clinical Significance

Alterations in the KIT gene or its expression can lead to various diseases, including:

  • Gastrointestinal stromal tumors (GISTs): KIT mutations are commonly associated with these tumors.
  • Mast cell disease: Abnormal KIT signaling can lead to mastocytosis.
  • Acute myelogenous leukemia (AML): KIT mutations are found in some cases of AML.
  • Piebaldism: A genetic disorder characterized by the absence of melanocytes in certain areas of the skin and hair .
Therapeutic Implications

Targeted therapies have been developed to inhibit KIT activity in diseases where it is overactive. Drugs like nilotinib and sunitinib have shown efficacy in treating patients with KIT overactivity, particularly in GISTs and melanomas .

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.