Detects intracellular G-CSF in PBMCs or bone marrow cells at 5–10 µL per 1×10⁶ cells .
Enables analysis of G-CSF receptor (G-CSFR) expression on CD34⁺ hematopoietic stem cells .
ELISA: Paired antibodies (e.g., BVD11-37G10 + BVD13-3A5) achieve detection limits <0.5 ng/mL .
Western Blot: Identifies G-CSF at ~18 kDa under reducing conditions .
Myeloid Expansion: G-CSF/anti-G-CSF FITC complexes enhance CD11b⁺Gr-1⁺ myeloid cell recovery by >100-fold post-chemotherapy, without impairing CD8⁺ T-cell responses .
Infection Models: FITC-labeled antibodies quantify G-CSF upregulation during Listeria monocytogenes infections, correlating with neutrophil-mediated bacterial clearance .
Autoimmunity: Detects anti-G-CSF autoantibodies in inflammatory bowel disease, aiding disease monitoring .
Transplant Monitoring: Tracks G-CSF-mediated CD34⁺ cell mobilization in autologous stem cell harvests .
Cancer Therapy: Evaluates G-CSF suppression of proinflammatory cytokines (e.g., IL-12) during immunotherapy .
Antibody-Drug Conjugates: FITC labeling aids pharmacokinetic studies of G-CSF-targeted biologics .
G-CSF (Granulocyte colony-stimulating factor) is a pleiotropic cytokine that primarily influences the differentiation, proliferation, and activation of the neutrophilic granulocyte lineage. Unlike GM-CSF and IL-3, which can stimulate cells of multiple lineages, G-CSF activity is specifically limited to neutrophilic granulocytes. The human G-CSF is an 18.8 kDa protein containing 175 amino acid residues, derived from a 207 amino acid precursor with a 29 amino acid signal peptide that undergoes proteolytic cleavage to form the mature protein . G-CSF is essential for maintaining neutrophil counts during homeostasis, with low basal levels detectable in the serum of healthy individuals. During infection, circulating levels become rapidly elevated, as G-CSF plays a critical role in activating and mobilizing mature neutrophils during the innate immune response .
G-CSF is primarily expressed by monocytes, macrophages, and bone marrow stromal cells under normal conditions. Additionally, fibroblasts can be induced to express G-CSF when stimulated by IL-17A . Under inflammatory conditions, endothelial cells can also produce this cytokine, contributing to the elevated circulating levels observed during infection . The expression pattern is tightly regulated, with significant upregulation occurring in response to pathogenic challenges, tissue damage, or other inflammatory stimuli, providing a mechanism for rapid neutrophil mobilization when needed for immune defense.
G-CSF antibodies serve unique research functions compared to other neutrophil markers by specifically recognizing granulocyte colony-stimulating factor rather than general neutrophil surface antigens. These antibodies can detect both intracellular G-CSF in producing cells and secreted G-CSF in biological samples . Unlike antibodies targeting CD11b, Ly6G, or myeloperoxidase that identify neutrophils themselves, G-CSF antibodies identify the regulatory cytokine controlling neutrophil development and function. This makes G-CSF antibodies particularly valuable for studying the mechanisms of granulopoiesis, neutrophil activation, and inflammatory signaling cascades. Furthermore, when conjugated with fluorescent markers like FITC, these antibodies enable researchers to visualize G-CSF expression patterns in different cell populations and track changes in G-CSF levels during various physiological and pathological processes .
For optimal intracellular staining of G-CSF using FITC-conjugated antibodies, researchers should follow specific methodological steps to ensure reliable and reproducible results. Begin with fixation and permeabilization of cells, as G-CSF is predominantly located intracellularly in producing cells. For flow cytometric analysis, use approximately 10μl of anti-G-CSF FITC antibody per 1,000,000 cells in a final staining volume of 100μL . The antibody concentration may require empirical determination based on your specific cell type, but typically ranges from 0.125 μg to 1 μg per test .
After staining, cells should be washed twice with permeabilization buffer to remove unbound antibody before analysis. When optimizing your protocol, consider including appropriate isotype controls and single-stained controls if performing multicolor experiments. For microscopy applications, similar fixation and permeabilization steps apply, though incubation times may need to be extended. Post-staining, mount samples in anti-fade medium without DAPI to prevent interference with the FITC signal. This methodological approach ensures specific detection of G-CSF in various cell types including monocytes, macrophages, and bone marrow stromal cells .
The excitation and emission properties of G-CSF Antibody, FITC significantly impact experimental design considerations, particularly for flow cytometry and fluorescence microscopy applications. FITC conjugates have excitation maxima in the 488-561 nm range and emission around 578 nm, requiring blue, green, or yellow-green lasers for optimal excitation . This spectral profile necessitates careful planning when designing multicolor panels to avoid fluorescence spillover into other channels, particularly PE and PerCP channels which have overlapping emission spectra with FITC.
When designing flow cytometry experiments, researchers should consider:
Compensating for spectral overlap if using PE or PerCP in the same panel
Accounting for potential autofluorescence in the FITC channel, particularly from myeloid cells
Selecting complementary fluorophores with minimal spectral overlap for other markers
Utilizing appropriate single-stained controls for accurate compensation
For microscopy applications, researchers must select filter sets that maximize FITC signal while minimizing background autofluorescence. Additionally, photobleaching can be a concern with FITC, so minimizing exposure time and using anti-fade mounting media can help preserve signal integrity during extended imaging sessions. These spectral considerations should guide experimental design to ensure optimal detection sensitivity and specificity when working with G-CSF Antibody, FITC conjugates .
FITC offers several distinct advantages for G-CSF detection compared to other fluorophores, though it also presents certain limitations that researchers should consider when designing experiments. The primary advantages include widespread compatibility with standard flow cytometers and fluorescence microscopes equipped with 488 nm lasers and appropriate filter sets, making FITC conjugates accessible for most research facilities without specialized equipment requirements . Additionally, FITC has a relatively large Stokes shift (the difference between excitation and emission wavelengths), which helps reduce self-quenching effects.
FITC has lower photostability compared to CF® dyes, Alexa Fluor® 488, or PE, potentially limiting its use in applications requiring extended imaging
FITC exhibits pH sensitivity, with reduced fluorescence in acidic environments (relevant for lysosomal or endosomal studies)
FITC has moderate brightness compared to PE or APC, potentially limiting detection of low-abundance targets
For detecting G-CSF specifically, which may be expressed at variable levels depending on cellular activation state, brighter fluorophores like PE might offer superior sensitivity for detecting low expression levels in resting cells . Conversely, FITC remains advantageous for detecting highly expressed G-CSF in activated cells or after stimulation protocols, where signal intensity is less limiting. When designing panels for simultaneous detection of G-CSF and other markers, researchers should reserve brighter fluorophores for low-abundance targets and consider FITC for more highly expressed proteins .
G-CSF Antibody, FITC serves as a powerful tool for investigating G-CSF/G-CSF receptor signaling in inflammatory diseases by enabling direct visualization and quantification of G-CSF expression patterns in various cell populations. This application is particularly valuable given recent evidence that therapeutic targeting of the G-CSF receptor can significantly reduce neutrophil trafficking and joint inflammation in antibody-mediated inflammatory arthritis . Researchers can employ this antibody in flow cytometry to identify G-CSF-producing cells within inflammatory lesions, tracking their temporal and spatial distribution during disease progression.
For studying signaling mechanisms, G-CSF Antibody, FITC can be combined with phospho-flow cytometry to simultaneously detect G-CSF expression and downstream signaling events such as STAT3 phosphorylation . This approach allows researchers to correlate G-CSF production with activation of specific signaling pathways in individual cells. Furthermore, sorting G-CSF-positive cells using FITC signal enables subsequent transcriptional analysis to identify gene expression programs associated with G-CSF production during inflammation.
In animal models of inflammatory diseases, administering G-CSF Antibody, FITC before tissue collection helps visualize sites of G-CSF production through fluorescence microscopy, illuminating the microanatomical niches where G-CSF signaling originates. This methodological approach has revealed that targeting G-CSF receptor signaling not only reduces neutrophil numbers but also alters their phenotype, with changes in surface expression of trafficking molecules like CXCR2 and CD62L that regulate neutrophil homing to inflammatory sites .
Studying G-CSF/anti-G-CSF antibody complexes for enhanced therapeutic efficacy requires sophisticated methodological approaches that integrate multiple techniques. First, researchers can use size exclusion chromatography or dynamic light scattering to verify complex formation and determine optimal ratios of G-CSF to anti-G-CSF antibody for stable complex generation. The remarkable finding that G-CSF efficacy can be enhanced over 100-fold through pre-association with an anti-G-CSF monoclonal antibody necessitates careful optimization of these ratios .
For in vitro assessment of enhanced biological activity, researchers should employ cell-based assays measuring neutrophil differentiation from hematopoietic progenitors. Comparative dose-response curves between free G-CSF and G-CSF/anti-G-CSF complexes enable precise quantification of potency enhancement. Flow cytometric analysis using FITC-labeled antibodies against CD11b and Gr-1 provides direct visualization of myeloid expansion in response to these complexes .
In vivo studies require careful experimental design:
Titration experiments comparing equivalent doses of free G-CSF versus G-CSF/anti-G-CSF complexes
Time-course analyses of neutrophil expansion in peripheral blood and tissues
Functional assessment of neutrophil bactericidal activity
Evaluation of protective immunity using bacterial challenge models like Listeria monocytogenes
Research has demonstrated that 0.015 μg of G-CSF complexed with anti-G-CSF antibody exhibits greater biological activity than 1.5 μg of G-CSF alone, indicating a ~100-fold enhancement in efficacy . This methodological framework enables comprehensive characterization of the pharmacodynamic and pharmacokinetic properties that underlie the dramatically enhanced biological activity of these complexes.
Integrating differential expression analysis of G-CSF with neutrophil trafficking studies requires a multifaceted methodological approach combining molecular techniques with in vivo imaging. Begin by establishing baseline G-CSF expression patterns in tissues of interest using quantitative RT-PCR and ELISA to measure transcript and protein levels, respectively. FITC-conjugated G-CSF antibodies can then be employed for flow cytometric analysis to identify G-CSF-producing cell populations and monitor changes in their frequency and distribution during inflammatory responses .
For trafficking studies, researchers should implement a comprehensive strategy including:
Real-time tracking of neutrophil migration using adoptive transfer of fluorescently labeled neutrophils
Correlating G-CSF expression patterns with neutrophil accumulation in tissues
Analyzing expression of trafficking molecules (CXCR2, CD62L) on neutrophils in response to G-CSF signaling
Evaluating local chemokine production (KC, MCP-1) that may be modulated by G-CSF
Studies have demonstrated that blocking G-CSF receptor signaling significantly alters neutrophil trafficking by modifying the expression of key adhesion molecules, with reduced CXCR2 and increased CD62L expression observed in neutrophils from anti-G-CSF receptor-treated mice . This altered expression profile correlates with diminished neutrophil accumulation in inflammatory sites without inducing systemic neutropenia.
To further elucidate mechanisms, RNA-sequencing of joint neutrophils following anti-G-CSF receptor therapy has revealed a transcriptional shift away from inflammatory phenotypes . This methodological integration of G-CSF expression analysis with neutrophil trafficking studies provides comprehensive insights into how G-CSF regulates not only neutrophil production but also their functional deployment during inflammatory responses.
Validating G-CSF Antibody, FITC specificity in flow cytometry requires implementing several critical controls to ensure reliable and reproducible results. First, isotype controls matched to the G-CSF antibody's host species, isotype, and FITC conjugation are essential for distinguishing specific binding from background caused by Fc receptor interactions or non-specific binding . These controls should be used at the same concentration as the G-CSF antibody.
Biological controls are equally important for validation:
Positive controls using cell types known to express G-CSF (e.g., stimulated monocytes, macrophages, or bone marrow stromal cells)
Negative controls using cell types that don't express G-CSF (e.g., lymphocytes)
Induction controls demonstrating increased G-CSF expression following appropriate stimulation (e.g., LPS treatment of monocytes)
For intracellular staining, include unstained and single-stained controls for each fluorochrome to facilitate accurate compensation in multicolor panels. Additionally, blocking experiments with unconjugated G-CSF antibody prior to staining with the FITC-conjugated version should abolish specific staining, confirming antibody specificity .
Fluorescence-minus-one (FMO) controls are particularly valuable for determining proper gating strategies, especially when G-CSF expression may be heterogeneous across cell populations. Finally, antibody titration experiments should be performed to identify the optimal concentration (typically 5 μL or 0.125 μg per test in 100 μL final volume) that maximizes specific signal while minimizing background . These methodological controls collectively ensure that the observed fluorescence genuinely represents G-CSF expression rather than technical artifacts.
Researchers can employ several quantitative methods to measure G-CSF antibody binding affinity and specificity with high precision. Surface Plasmon Resonance (SPR) represents the gold standard for determining binding kinetics, providing both association (kon) and dissociation (koff) rate constants, from which the equilibrium dissociation constant (KD) can be calculated. For this application, purified G-CSF protein is immobilized on a sensor chip, and varying concentrations of the antibody are flowed over the surface to generate binding curves .
Enzyme-Linked Immunosorbent Assay (ELISA) provides complementary quantitative data:
Direct ELISA: Coat plates with G-CSF, then apply serial dilutions of antibody
Competition ELISA: Pre-mix fixed antibody concentration with varying G-CSF concentrations
Epitope mapping: Use peptide fragments of G-CSF to identify binding regions
Flow cytometry-based approaches offer cellular context for affinity measurements through calculation of the effective KD using mean fluorescence intensity values from cells stained with serial dilutions of the FITC-conjugated antibody . To assess specificity, cross-reactivity testing against structurally related cytokines (GM-CSF, IL-3) is essential.
Western blot analysis under reducing and non-reducing conditions helps confirm recognition of the appropriate molecular weight species (18.8 kDa for human G-CSF) and determine whether the antibody recognizes conformational or linear epitopes . For functional validation, researchers should verify that the antibody can recognize both natural and recombinant G-CSF, and potentially both human and murine forms if cross-reactivity is claimed. The integration of these quantitative approaches provides comprehensive characterization of antibody binding properties essential for experimental design and interpretation.
Preserving G-CSF epitope integrity during sample preparation is crucial for accurate antibody detection and requires methodological precision. For flow cytometry and immunocytochemistry applications with FITC-conjugated antibodies, begin with gentle fixation using 2-4% paraformaldehyde for 10-15 minutes at room temperature, which adequately stabilizes cellular architecture while preserving most epitopes . Avoid methanol-based fixatives which can denature protein structure and potentially destroy conformational epitopes recognized by many G-CSF antibodies.
For intracellular staining, permeabilization requires careful optimization:
Saponin-based buffers (0.1-0.5%) provide reversible permeabilization that maintains protein structure
Triton X-100 (0.1-0.2%) offers stronger permeabilization but may disrupt some epitopes
Commercial fixation/permeabilization kits designed for cytokine detection generally yield excellent results
Temperature management is critical throughout the procedure. Maintain cells at 4°C before fixation to prevent cytokine secretion, perform antibody incubations at 4°C to reduce non-specific binding, and store samples at -20°C after reconstitution if not used immediately .
For tissue sections, antigen retrieval methods must be carefully selected. For G-CSF detection, citrate buffer (pH 6.0) heat-induced epitope retrieval typically provides optimal results without excessive background. When processing blood samples for G-CSF detection, immediate processing or addition of protease inhibitors helps prevent degradation of secreted G-CSF by neutrophil proteases.
These methodological considerations ensure optimal epitope preservation, enabling reliable and sensitive detection of G-CSF using FITC-conjugated antibodies across various experimental applications .
Addressing poor signal-to-noise ratio when using G-CSF Antibody, FITC in complex tissue samples requires systematic optimization of multiple experimental parameters. First, optimize antibody concentration through careful titration experiments to determine the minimum antibody concentration that yields maximum specific signal. Excessive antibody often increases background staining without enhancing specific signal detection .
To minimize autofluorescence, which is particularly problematic in the FITC channel:
Incorporate an autofluorescence quenching step using reagents such as Sudan Black B (0.1-0.3%)
Consider using tissue clearing techniques compatible with immunofluorescence
Implement spectral unmixing during image acquisition if using confocal microscopy
Background from non-specific binding can be reduced by extending blocking steps with species-appropriate serum (5-10%) supplemented with bovine serum albumin (1-3%) and increasing the duration to 1-2 hours . Additionally, use 0.2 μm filtered antibody preparations to remove aggregates that can contribute to background .
For flow cytometry applications, improve discrimination between positive and negative populations by:
Optimizing voltage settings for the FITC channel
Implementing stringent gating strategies based on fluorescence-minus-one controls
Using viability dyes to exclude dead cells, which often exhibit elevated autofluorescence
When analyzing tissues with high collagen content (known for autofluorescence in the FITC spectrum), consider using alternative fluorophores or implementing computational approaches for background subtraction. Multi-spectral imaging systems can help discriminate between genuine FITC signal and tissue autofluorescence by analyzing complete spectral profiles rather than single emission wavelengths .
Analyzing G-CSF expression in myeloid leukemias using FITC-conjugated antibodies requires specialized methodological considerations to ensure accurate results in these complex malignancies. Begin with sample processing optimized for leukemic cells, using density gradient centrifugation with Ficoll-Paque to isolate mononuclear cells while minimizing granulocyte contamination that could skew results. For bone marrow aspirates, red cell lysis should be performed using ammonium chloride-based buffers rather than hypotonic solutions to preserve cellular morphology .
When designing multicolor flow cytometry panels:
Include lineage markers (CD34, CD33, CD117) to identify leukemic populations
Add differentiation markers (CD11b, CD15) to determine maturation status
Incorporate viability dyes to exclude dead cells, which are often elevated in leukemic samples
Consider including markers for apoptosis (Annexin V) as G-CSF may affect cell survival
For intracellular staining, optimize fixation and permeabilization specifically for leukemic blasts, which may have different permeability characteristics than normal myeloid cells. Commercial kits designed for detecting intracellular phospho-epitopes often work well for cytokine detection in leukemic cells .
When interpreting results, implement quantitative analysis using median fluorescence intensity rather than percent positive cells, as G-CSF expression may manifest as shifts in expression level rather than distinct positive/negative populations. Additionally, compare results to age-matched normal controls processed identically, as baseline G-CSF expression varies with age and health status.
For prognostic studies, correlate G-CSF expression with clinical outcomes and genetic markers, particularly those associated with granulocytic differentiation. This methodological framework enables accurate assessment of G-CSF expression in myeloid leukemias and granulocytic sarcomas, providing valuable diagnostic and prognostic information .
Distinguishing between membrane-bound and intracellular G-CSF using immunofluorescence techniques requires a methodical differential staining approach with precise protocol modifications. Begin by implementing a sequential staining strategy where cells are first stained for membrane-bound G-CSF using G-CSF Antibody, FITC on unfixed, non-permeabilized cells at 4°C to prevent internalization during staining . After thorough washing, cells are then fixed and permeabilized for subsequent staining of intracellular G-CSF using a spectrally distinct fluorophore-conjugated G-CSF antibody (e.g., PE or APC).
For microscopy-based differentiation:
Utilize membrane markers like wheat germ agglutinin (conjugated to a far-red fluorophore) to delineate cell membranes
Perform high-resolution confocal microscopy with z-stack acquisition to precisely localize G-CSF signal relative to the membrane
Implement deconvolution algorithms to enhance spatial resolution beyond the diffraction limit
Consider super-resolution techniques (STED, PALM, STORM) for definitive localization studies
With flow cytometry, researchers can exploit the acid wash technique to distinguish surface from intracellular staining. After staining with G-CSF Antibody, FITC, briefly expose cells to acidic buffer (pH 2.0-3.0) to strip surface-bound antibodies, then compare fluorescence before and after acid treatment .
To validate findings, implement biological controls demonstrating trafficking of G-CSF:
Use Brefeldin A or Monensin treatment to block protein secretion, causing accumulation of intracellular G-CSF
Stimulate cells with LPS or other activators to induce G-CSF production and monitor changes in localization
Perform pulse-chase experiments with differentially labeled antibodies to track G-CSF movement over time
These methodological approaches enable precise discrimination between membrane-associated and intracellular G-CSF populations, providing insights into G-CSF's cellular trafficking and biological functions in neutrophil development and inflammatory responses .
G-CSF Antibody, FITC provides a powerful tool for investigating the complex relationship between G-CSF signaling and neutrophil extracellular trap (NET) formation through simultaneous visualization of both processes. Researchers can implement a multicolor flow cytometry approach combining G-CSF Antibody, FITC with DNA stains (Hoechst or DRAQ5) and citrullinated histone H3 antibodies to detect NET-forming neutrophils while simultaneously assessing G-CSF expression levels . This approach enables correlation between G-CSF signaling intensity and NET formation propensity at the single-cell level.
For microscopy-based studies, researchers should develop co-staining protocols that incorporate:
G-CSF Antibody, FITC for detecting G-CSF expression or binding
DNA stains (SYTOX dyes) for visualizing extracellular chromatin
Neutrophil elastase antibodies to confirm NET protein components
G-CSFR antibodies to assess receptor expression and distribution
This methodological framework allows for spatiotemporal analysis of G-CSF signaling events preceding NET formation. Time-lapse imaging using these markers can reveal whether G-CSF acts as a priming signal for subsequent NET induction by secondary stimuli.
To establish causality, researchers should implement G-CSF receptor blockade using neutralizing antibodies while monitoring NET formation in response to various stimuli. Studies have demonstrated that therapeutic targeting of the G-CSF receptor can significantly alter neutrophil function and trafficking in inflammatory conditions, suggesting potential impacts on NET formation . Transcriptional analysis of neutrophils following G-CSF stimulation, facilitated by G-CSF Antibody, FITC-based cell sorting, can further identify gene expression changes associated with enhanced NET formation capacity, providing mechanistic insights into this clinically relevant process.
Investigating whether G-CSF/anti-G-CSF complexes induce distinct neutrophil functional phenotypes compared to G-CSF alone requires sophisticated experimental approaches integrating functional, phenotypic, and molecular analyses. Begin by isolating neutrophils from bone marrow or peripheral blood, then expose them to equivalent bioactive concentrations of either G-CSF alone or G-CSF/anti-G-CSF complexes (accounting for the ~100-fold potency enhancement of complexes) .
For comprehensive functional phenotyping, implement a multiparameter assessment protocol:
Chemotaxis assays using transwell systems to measure directional migration toward various chemoattractants
Phagocytosis assays quantifying uptake of fluorescent bacteria or beads
Oxidative burst assessment measuring reactive oxygen species production via luminol-enhanced chemiluminescence
NET formation quantification using plate-based fluorescence assays and confirmatory microscopy
Bacterial killing assays with clinically relevant pathogens to assess microbicidal capacity
Surface phenotype characterization using flow cytometry should assess expression levels of:
Chemokine receptors (CXCR1, CXCR2) governing tissue homing
Adhesion molecules (CD62L, CD11b) mediating endothelial interactions
Fc receptors (CD16, CD32) involved in immune complex recognition
Activation markers (CD66b, CD69) indicating neutrophil priming status
Research has demonstrated that G-CSF/anti-G-CSF complexes induce more robust expansion of CD11b+Gr-1+ myeloid cells than equivalent doses of G-CSF alone, suggesting potential qualitative differences in neutrophil phenotypes . Transcriptomic profiling using RNA-sequencing of neutrophils exposed to either treatment can reveal differential gene expression patterns governing functional outcomes. Importantly, the effect of G-CSF/anti-G-CSF complexes on neutrophil responses during bacterial infection warrants investigation, as these complexes have been shown to provide enhanced protection against Listeria monocytogenes infection compared to G-CSF alone .
Integrating G-CSF antibody techniques with single-cell RNA sequencing (scRNA-seq) offers a powerful methodological approach to identify and characterize novel neutrophil subpopulations based on their G-CSF response patterns. Begin by implementing G-CSF Antibody, FITC-based flow cytometry sorting to isolate neutrophils expressing different levels of G-CSF receptor or producing varying amounts of G-CSF itself . This pre-enrichment strategy creates biologically meaningful subpopulations for subsequent single-cell analysis.
For optimal experimental design:
Sort neutrophils into G-CSF-high, G-CSF-medium, and G-CSF-low populations using FITC signal
Process sorted cells immediately for single-cell RNA-seq using platforms like 10x Genomics Chromium
Index sort to retain the precise FITC fluorescence intensity of each sequenced cell
Include experimental conditions comparing healthy donors with inflammatory disease patients
Computational analysis should employ dimensionality reduction techniques (t-SNE, UMAP) to visualize neutrophil heterogeneity, followed by clustering algorithms to define subpopulations. Integration of G-CSF antibody index sorting data with transcriptomic clusters enables direct correlation between G-CSF protein expression/binding and transcriptional states.
This approach can reveal previously unrecognized neutrophil subpopulations with distinct G-CSF response patterns. For instance, research on G-CSF receptor signaling in inflammatory arthritis has demonstrated that neutrophils undergo phenotypic changes affecting trafficking molecule expression (CXCR2, CD62L) following G-CSF receptor blockade . Single-cell transcriptomics can extend these findings by identifying the complete transcriptional signatures associated with different G-CSF response states.
To validate identified subpopulations, researchers should perform functional assays on re-sorted populations based on scRNA-seq markers, creating a feedback loop between protein-level G-CSF detection and transcriptomic profiling. This integrated approach promises to reveal the full spectrum of neutrophil heterogeneity in health and disease, with potential implications for targeted therapeutic strategies .
Mouse anti-human G-CSF antibodies are monoclonal antibodies produced by immunizing mice with human G-CSF. These antibodies are highly specific and can be used in various research and diagnostic applications, including enzyme-linked immunosorbent assays (ELISA), western blotting, and flow cytometry. They are valuable tools for studying the expression, function, and regulation of G-CSF in different biological contexts .
Fluorescein isothiocyanate (FITC) is a fluorescent dye commonly used to label antibodies and other biomolecules. FITC-labeled antibodies can be detected using fluorescence microscopy or flow cytometry, allowing researchers to visualize and quantify the presence of specific antigens in cells or tissues. FITC emits green fluorescence when excited by blue light, making it a popular choice for multi-color staining experiments .
Research: FITC-labeled mouse anti-human G-CSF antibodies are widely used in research to study the role of G-CSF in various physiological and pathological processes. They can help elucidate the mechanisms underlying G-CSF-mediated signaling pathways and its effects on immune cell function and development .
Clinical Diagnostics: These antibodies can be used in diagnostic assays to measure G-CSF levels in patient samples. This is particularly useful for monitoring patients undergoing chemotherapy or bone marrow transplantation, as G-CSF levels can provide valuable information about their immune status and response to treatment .
Therapeutic Development: Understanding the interaction between G-CSF and its receptor can aid in the development of new therapeutic strategies for conditions involving neutropenia or other immune-related disorders. FITC-labeled antibodies can be used to screen for potential drug candidates that modulate G-CSF activity .