The KSR1 Antibody, FITC conjugated consists of:
Antibody specificity: Targets KSR1, a scaffold protein regulating the Raf/MEK/ERK signaling cascade .
Conjugate: FITC fluorophore covalently linked to the antibody, enabling green fluorescence (emission: 525 nm) .
Epitope recognition:
| Parameter | Details |
|---|---|
| Host Species | Rabbit |
| Clonality | Monoclonal (e.g., clone EPR2421Y) or polyclonal |
| Reactivity | Human, Mouse, Rat |
| Applications | Flow cytometry, immunofluorescence, Western blot |
KSR1 Antibody, FITC conjugated is used to investigate:
ERK activation dynamics: KSR1 scaffolds Raf, MEK, and ERK, amplifying PDGF- or EGF-induced ERK phosphorylation .
Subcellular localization: FITC labeling reveals KSR1’s translocation to the plasma membrane upon growth factor stimulation .
Oncogenic signaling: KSR1 overexpression enhances ERK-driven proliferation and H-RasV12-mediated transformation .
Therapeutic targeting: Reduced KSR1 levels impair tumor growth in murine models .
Mast cell regulation: KSR1 modulates SCF-induced chemotaxis and FcεRI-dependent degranulation via Pak2 and ERK .
Infection response: KSR1-deficient mice exhibit heightened susceptibility to Pseudomonas aeruginosa due to dysregulated iNOS activity .
Flow cytometry: Detects KSR1 phosphorylation in HeLa cells, with lambda phosphatase treatment confirming specificity .
Western blot: Identifies KSR1 at ~110 kDa in human, mouse, and rat lysates .
Immunohistochemistry: Localizes KSR1 in human cervical carcinoma tissues .
Optimal KSR1 levels: ERK activation peaks at 14× endogenous KSR1 expression, while excess KSR1 sequesters Raf/MEK/ERK, inhibiting signaling .
Feedback regulation: KSR1 facilitates ERK-dependent phosphorylation of Raf and MEK, fine-tuning pathway activity .
Cancer: KSR1 promotes melanoma and lung adenocarcinoma progression via sustained ERK signaling .
Inflammation: KSR1 deficiency reduces mast cell-mediated allergic responses and neutrophilic inflammation .
KSR1 functions as part of a multiprotein signaling complex that promotes phosphorylation of Raf family members and activation of downstream MAP kinases . Independent of its kinase activity, KSR1 acts as an allosteric activator of BRAF; upon binding to MAP2K1/MEK1 or MAP2K2/MEK2, it dimerizes with BRAF and promotes BRAF-mediated phosphorylation of these kinases . KSR1 enhances the activation of the MAP kinase extracellular signal-regulated kinase (ERK) and promotes ERK activation in response to both EGF and cAMP signaling . Although KSR1 has a kinase-like domain, its intrinsic kinase activity remains controversial, with some protein kinase activity detected in vitro but unclear physiological relevance .
FITC-conjugated KSR1 antibodies are immunological reagents in which fluorescein isothiocyanate (FITC) has been chemically linked to antibodies targeting KSR1 protein . These antibodies are typically derived from rabbits immunized with KLH-conjugated synthetic peptides from human KSR1 or other specific immunogens corresponding to phosphorylation sites like S392 . The antibodies exist in various formats, including recombinant monoclonal variants with IgG isotypes . The FITC conjugation allows direct visualization of KSR1 in various applications, particularly flow cytometry and immunofluorescence, without requiring secondary antibody detection steps .
FITC conjugation provides a bright green fluorescence (excitation ~495 nm, emission ~519 nm) that is directly compatible with standard FITC filter sets in fluorescence microscopy and flow cytometry . Unlike Alexa Fluor 594-conjugated KSR1 antibodies, which emit in the red spectrum , FITC-conjugated antibodies allow for different multicolor experimental designs when combined with other fluorophores. While FITC offers good brightness, it can be susceptible to photobleaching and pH sensitivity compared to more stable fluorophores. The choice between FITC and other conjugates (like Alexa Fluor dyes or phycoerythrin) should be based on the experimental design, available instrumentation, and the need for multiplexing with other fluorescent markers .
FITC-conjugated KSR1 antibodies have been validated for several key applications in research settings. Western blotting (WB) can be performed at dilutions ranging from 1:300-5000, allowing researchers to detect KSR1 protein in cell and tissue lysates . Flow cytometry (FCM) is optimally performed at dilutions between 1:20-100, enabling quantitative assessment of KSR1 expression or phosphorylation states in cell populations . Immunofluorescence and immunocytochemistry (IF/ICC) applications typically utilize dilutions between 1:50-200, facilitating visualization of KSR1 subcellular localization . For phospho-specific antibodies, such as those targeting phospho-S392, flow cytometry has been specifically validated for human samples . When designing experiments, researchers should consider the specific application requirements and optimize antibody concentrations accordingly.
For optimal detection of KSR1 using FITC-conjugated antibodies, careful sample preparation is essential. For flow cytometry, cells should be fixed with 4% paraformaldehyde for 10 minutes at room temperature, followed by permeabilization with 90% methanol for 30 minutes at -20°C . When preparing samples for immunofluorescence microscopy, cells should be adhered to poly-lysine coated slides, fixed, and permeabilized before antibody staining . For studying KSR1 in the context of immune cell interactions, conjugates between effector cells (T cells or NK cells) and target cells can be formed by gentle centrifugation (30 seconds) followed by incubation at 37°C for the desired time points . Storage of FITC-conjugated antibodies should follow manufacturer recommendations, typically involving storage at -20°C in an aqueous buffered solution containing TBS, BSA, glycerol, and preservatives like Proclin300 .
Rigorous experimental design with appropriate controls is critical when using FITC-conjugated KSR1 antibodies. Isotype controls consisting of FITC-conjugated rabbit IgG with no specific target should be included to assess non-specific binding . Biological negative controls using KSR1 knockdown cells are essential to confirm antibody specificity, as demonstrated in studies using shRNA-mediated KSR1 depletion . For phospho-specific KSR1 antibodies, such as phospho-S392, samples treated with phosphatase inhibitors versus phosphatase should be compared . Positive controls should include cells known to express KSR1, such as Jurkat T cells or NK92 cells, which have been documented to express detectable levels of KSR1 . When performing multicolor flow cytometry, fluorescence minus one (FMO) controls should be included to account for spectral overlap between fluorophores.
FITC-conjugated KSR1 antibodies can be powerful tools for investigating KSR1 recruitment to the immunological synapse (IS) in lymphocytes. To study this phenomenon, researchers should conjugate T cells with antigen-presenting cells (APCs) loaded with appropriate antigens, such as superantigens (e.g., staphylococcal enterotoxin E) . After conjugation, cells should be gently centrifuged and placed on poly-lysine-coated slides for 5-10 minutes at 37°C . Following fixation and permeabilization, FITC-conjugated KSR1 antibodies can be applied to visualize KSR1 localization . Confocal microscopy with 63× objective lenses provides optimal resolution for observing KSR1 recruitment . Important controls should include conjugates formed without antigen loading and comparison with KSR1-YFP fusion proteins, which have been used to demonstrate that KSR1 is specifically recruited to the IS during T-cell activation .
To simultaneously assess KSR1 phosphorylation and protein interactions, researchers can employ a combination of techniques. For phosphorylation studies, phospho-specific antibodies such as FITC-anti-KSR1 (phospho S392) can be used in flow cytometry or microscopy approaches . To study protein interactions, immunoprecipitation followed by immunoblotting can be performed by lysing cells in ice-cold lysis buffer (containing 0.1M Tris base, 140mM NaCl, 1mM EDTA, 1% NP-40, and protease/phosphatase inhibitors) . Proteins from cell lysates should be resolved by SDS-PAGE and analyzed by immunoblotting with appropriate antibodies . For quantitative analysis of KSR1 protein levels, immunoblotting with the Odyssey system allows precise quantification . Advanced approaches may include proximity ligation assays or FRET-based methods to detect KSR1 interactions with binding partners such as Raf, MEK, and ERK in intact cells .
Investigating KSR1's role in modulating MAPK pathway sensitivity requires sophisticated experimental approaches. Flow cytometry with FITC-conjugated KSR1 antibodies allows analysis of KSR1 expression levels in relation to ERK activation in single cells . This approach has revealed that KSR1 does not change the digital (all-or-none) versus analog (graded) nature of ERK responses, but instead modulates the sensitivity of the system . To establish this relationship, researchers should stimulate cells with titrated doses of pathway activators (e.g., TCR engagement with superantigen for T cells, or CXCR4 stimulation) . Co-staining for phospho-ERK and KSR1 enables correlation between KSR1 levels and MAPK pathway activation thresholds . Complementary approaches include manipulating KSR1 expression through knockdown or overexpression systems and assessing how this alters dose-response curves for ERK activation . Time-course experiments can further reveal how KSR1 affects the kinetics of MAPK pathway activation and deactivation.
Several common pitfalls can affect experiments using FITC-conjugated KSR1 antibodies. Photobleaching is a significant concern with FITC, which can reduce signal intensity during imaging; this can be mitigated by minimizing exposure time, using anti-fade reagents, and optimizing imaging parameters . Non-specific binding may occur, particularly in fixed and permeabilized samples, and can be reduced by including appropriate blocking steps with serum or BSA and performing thorough washing steps . Autofluorescence in the FITC channel from certain cell types or fixatives can interfere with specific signal detection; this can be addressed through proper controls and background subtraction during analysis . Suboptimal fixation and permeabilization can limit antibody access to intracellular KSR1, particularly when studying subcellular localization; researchers should optimize these steps based on the cellular compartment being investigated (cytoplasm, cell membrane) . Finally, repeated freeze-thaw cycles can compromise antibody performance, so aliquoting antibodies into single-use volumes is recommended .
Data normalization and comparison across experimental conditions requires careful consideration when using FITC-conjugated KSR1 antibodies. For flow cytometry data, median fluorescence intensity (MFI) should be reported after subtracting the isotype control MFI . When comparing across experiments, normalization to internal standards or control cell lines with known KSR1 expression levels provides consistency . For imaging data, integrated fluorescence intensity within regions of interest should be measured, with background subtraction from adjacent regions . When quantifying KSR1 recruitment to specific subcellular locations (e.g., immunological synapse), the enrichment ratio should be calculated by dividing the fluorescence intensity at the region of interest by the average intensity throughout the cell . For western blot quantification, KSR1 levels should be normalized to housekeeping proteins (e.g., tubulin) and presented relative to control conditions . Statistical analysis should employ appropriate tests based on data distribution, with paired analyses used when comparing treatments on the same cell populations.
Distinguishing specific staining from background requires systematic controls and analytical approaches. Isotype controls using FITC-conjugated rabbit IgG that has no relevant specificity should be processed identically to the experimental samples to establish baseline fluorescence levels . KSR1 knockdown cells provide critical biological negative controls that define the antibody's specificity; any signal observed in knockdown cells can be considered background . For phospho-specific antibodies, treatment with phosphatase inhibitors versus phosphatase treatment can confirm signal specificity . In imaging applications, examining subcellular localization patterns can help distinguish specific staining (e.g., cytoplasmic and membrane localization for KSR1) from non-specific signals (which often appear diffuse or punctate throughout the cell) . Titration experiments with increasing antibody concentrations can help identify the optimal signal-to-noise ratio, as specific staining typically increases proportionally with concentration until saturation, while background may increase linearly .
KSR1 phosphorylation at S392 represents an important regulatory mechanism for MAPK pathway modulation. This phosphorylation site can be specifically detected using phospho-S392 antibodies conjugated to FITC . Research suggests that S392 phosphorylation affects KSR1's scaffold function by altering its interaction with other MAPK pathway components . To investigate this relationship, researchers should compare wild-type KSR1 with phosphomimetic (S392D/E) and phosphodeficient (S392A) mutants in functional assays measuring ERK activation kinetics and amplitude . Flow cytometry with dual staining for phospho-S392 KSR1 and phospho-ERK can reveal correlations between these phosphorylation events at the single-cell level . Immunoprecipitation experiments following cell stimulation can determine how S392 phosphorylation affects KSR1's ability to form complexes with Raf, MEK, and ERK . The temporal dynamics of S392 phosphorylation relative to ERK activation can provide insights into whether this modification functions as a feedback mechanism or priming event in the signaling cascade.
The relationship between KSR1 membrane recruitment and ERK activation varies across different cell types and signaling contexts. In T cells, KSR1 is recruited to the immunological synapse during activation, which correlates with ERK activation and localization to the same region . This recruitment depends on KSR1's CA3 domain, which contains conserved cysteine residues critical for membrane binding . Studies using CA3-mutated KSR1 have shown that membrane recruitment is partially dispensable for ERK activation but is required for efficient pERK accumulation at the immunological synapse . To investigate this relationship in different cell types, researchers should generate KSR1-YFP fusion constructs with intact or mutated CA3 domains and express them in KSR1-deficient cells . Live-cell imaging during stimulation can then track KSR1 membrane recruitment kinetics in relation to ERK activation measured by FRET-based sensors or phospho-ERK staining . Correlation analysis between the degree of membrane recruitment and ERK activation levels in single cells can reveal cell-type specific dependencies.
KSR1 expression levels significantly impact MAPK signaling dynamics without changing the fundamental system output characteristics. Research has demonstrated that in T cells, engagement of the T-cell receptor (TCR) with superantigen results in a digital (all-or-none) ERK response, whereas stimulation through the G-protein coupled receptor CXCR4 generates a graded (analog) ERK output . KSR1 does not rewire this system behavior but instead modulates the sensitivity threshold for activation . To investigate this relationship, researchers should establish cell systems with titratable KSR1 expression, using technologies such as doxycycline-inducible promoters or sorting cells expressing KSR1-GFP fusions into different expression level bins . Flow cytometry with phospho-ERK staining following dose titration of different stimuli can then reveal how KSR1 levels affect the EC50 (half-maximal effective concentration) for pathway activation and the steepness of the dose-response curve . Single-cell imaging approaches can further characterize how KSR1 levels affect the probability of digital responses versus the amplitude of analog responses across different signaling inputs and cell types .
Emerging technologies promise to expand the utility of FITC-conjugated KSR1 antibodies in research. Advanced super-resolution microscopy techniques, including structured illumination microscopy (SIM), stimulated emission depletion (STED), and photoactivated localization microscopy (PALM), could overcome the diffraction limit to reveal KSR1 nanoscale organization at signaling clusters and the immunological synapse with unprecedented detail . Mass cytometry (CyTOF) approaches coupled with antibody metal conjugation could allow simultaneous measurement of KSR1 expression, phosphorylation states, and dozens of other cellular parameters without fluorescence overlap constraints . Single-cell RNA-sequencing paired with protein detection (CITE-seq) could correlate KSR1 protein levels with transcriptional states across heterogeneous cell populations . Spatial transcriptomics combined with KSR1 immunofluorescence could reveal how KSR1 protein distribution relates to local gene expression patterns in tissues . These emerging technologies will provide multidimensional insights into KSR1 biology that current methods cannot achieve.
FITC-conjugated KSR1 antibodies can provide valuable insights into therapeutic resistance mechanisms in cancer treatments targeting the RAF/MEK/ERK pathway. Since KSR1 functions as an allosteric activator of BRAF and modulates MAPK pathway sensitivity, changes in KSR1 expression or phosphorylation could contribute to resistance to BRAF or MEK inhibitors . Flow cytometry with FITC-KSR1 antibodies could be used to monitor KSR1 levels in patient-derived xenografts or circulating tumor cells before and after treatment with these inhibitors . Phospho-specific antibodies targeting sites like S392 could reveal adaptive changes in KSR1 regulation during therapy . Combining KSR1 detection with phospho-ERK staining can identify populations of cells that maintain ERK signaling despite inhibitor treatment . Immunofluorescence microscopy could determine whether drug resistance correlates with altered subcellular localization of KSR1, potentially identifying new therapeutic vulnerabilities . These approaches could ultimately inform combination therapies that target both the kinase cascade and scaffold proteins like KSR1 to overcome resistance.