G CSF Human, His

Granulocyte-Colony Stimulating Factor Human Recombinant, His Tag
Shipped with Ice Packs
In Stock

Description

Production and Purification

Recombinant G-CSF Human, His is produced using two primary systems:

HEK Cell Expression

  • Advantages: Post-translational modifications (e.g., glycosylation) mimic native human G-CSF .

  • Purification: Proprietary chromatographic techniques yield >95% purity .

  • Formulation: Lyophilized from 1xPBS, stable at -18°C long-term with carrier proteins (e.g., 0.1% HSA) .

coli Expression

  • Advantages: Cost-effective, high yield (e.g., 18.8 kDa monomer) .

  • Purification: Affinity chromatography leveraging the His tag .

  • Reconstitution: Requires sterile water or PBS with 0.1% HSA to prevent aggregation .

Biological Activity

G-CSF Human, His binds the G-CSF receptor (GCSFR), activating JAK/STAT, MAPK, and PI3K/Akt pathways to stimulate neutrophil proliferation and survival . Key metrics:

ParameterHEK-Derived E. coli-Derived
ED₅₀ (Cell Proliferation)<0.01 ng/mL (M-NFS-60 cells)≤0.06 ng/mL (NFS-60 cells)
Stability3 weeks at RT (lyophilized)24 months at -80°C (lyophilized)
Endotoxin LevelsNot specified≤0.1 EU/µg

Oncology Support

  • Chemotherapy-Induced Neutropenia: Reduces infection risk by shortening neutropenia duration (4–7 days faster recovery) .

  • Stem Cell Mobilization: Synergizes with Stem Cell Factor (SCF) to enhance peripheral blood progenitor cell harvest .

Emerging Therapeutic Roles

  • Neuroprotection: Promotes neurogenesis and reduces apoptosis in CNS injuries .

  • Cancer Biology: Paradoxically, G-CSF/R axis may promote gastric/colorectal cancer progression via ERK/RSK signaling .

Product Specs

Introduction
Granulocyte-colony stimulating factor (G-CSF) is a cytokine secreted by macrophages, endothelial cells, and other immune cells. G-CSF stimulates the bone marrow to produce granulocytes and promotes the survival, proliferation, differentiation, and function of neutrophil granulocyte progenitors and mature neutrophils.
Description
Recombinant human granulocyte colony stimulating factor (G-CSF) with a C-terminal hexahistidine tag was expressed in E. coli. It is a single, non-glycosylated polypeptide chain containing the amino acids 31-204 of the mature G-CSF protein, resulting in a molecular mass of 23.19 kDa. The protein was purified using proprietary chromatographic techniques.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
G-CSF His is supplied in a buffer of 1x PBS and 50% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For long-term storage, the product should be stored at -20°C. Avoid repeated freeze-thaw cycles.
Purity
The purity is greater than 95.0% as determined by: (a) RP-HPLC and (b) SDS-PAGE.
Synonyms
CSF-3, MGI-1G, GM-CSF beta, Pluripoietin, Filgrastim, Lenograstim, G-CSF, MGC45931, GCSF.
Source
Escherichia Coli.

Q&A

What is the molecular structure of human G-CSF and how does the His-tag modify its properties?

Human G-CSF is a glycoprotein that exists naturally in two forms: a 174- and 177-amino-acid-long protein with a molecular weight of 19,600 grams per mole. The more-abundant and more-active 174-amino acid form is typically used in recombinant DNA technology for pharmaceutical development .

When a histidine tag is added to G-CSF, it creates a fusion protein that facilitates purification through metal affinity chromatography. The His-tag generally consists of 6-10 histidine residues added to either the N- or C-terminus of the protein. This modification allows for:

  • Enhanced purification efficiency using nickel or cobalt chelation chromatography

  • Improved protein detection capabilities in research settings

  • Minimal interference with the biological activity of the core G-CSF molecule when properly designed

The addition of a His-tag typically does not significantly alter the molecular weight or functional properties of G-CSF if positioned correctly, though validation of biological activity post-tagging is essential for research applications.

How do the biological activities of recombinant His-tagged G-CSF compare to native human G-CSF?

The biological activities of His-tagged G-CSF and native G-CSF are generally comparable when the tag is properly positioned, though specific differences should be validated experimentally:

  • Receptor binding kinetics: His-tagged G-CSF typically maintains similar binding affinity to the G-CSF receptor, though slight variations may occur depending on tag positioning. Binding involves homodimeric interactions with a 2:2 stoichiometry between G-CSF molecules and receptors .

  • Signaling cascade activation: Both forms activate similar signaling pathways including JAK/STAT, Ras/MAPK, and PI3K/Akt cascades that regulate neutrophil differentiation and function .

  • Cell proliferation induction: His-tagged G-CSF maintains the ability to stimulate proliferation of neutrophil precursors, though potency may vary slightly depending on the specific His-tag configuration.

  • In vivo neutrophil mobilization: When properly folded, His-tagged G-CSF demonstrates comparable capacity to increase absolute neutrophil counts in experimental models .

Researchers should conduct comparative assays when switching between native and His-tagged G-CSF to ensure experimental consistency, particularly for quantitative studies of receptor binding kinetics or cell proliferation rates.

What are the optimal storage and handling conditions for maintaining His-tagged G-CSF stability?

For maximal stability and activity retention of His-tagged G-CSF:

Storage Conditions:

  • Store lyophilized protein at -20°C to -80°C

  • Reconstituted protein should be stored at 4°C for short-term use (1-2 weeks)

  • For long-term storage, aliquot and freeze at -80°C to avoid freeze-thaw cycles

  • Maintain pH between 4.0-5.0 for optimal stability

Buffer Recommendations:

  • Use phosphate or acetate buffers with 0.1% human serum albumin or 0.005% polysorbate 80 as stabilizers

  • Avoid repeated freeze-thaw cycles (no more than 3)

  • Protect from direct light exposure

Working Concentrations:

  • Typical working concentrations range from 400 pg/ml to 100 ng/ml for most experimental applications

  • Higher concentrations (>1 μg/ml) may be required for receptor binding studies

Quality Control Monitoring:

  • Regularly assess protein activity using cell-based assays

  • Monitor for aggregation using size exclusion chromatography

  • Check for degradation using SDS-PAGE analysis

Careful attention to these handling conditions minimizes activity loss and ensures reproducible experimental results.

How does the G-CSF receptor complex form and initiate signaling cascades?

The G-CSF receptor (G-CSFR) belongs to the class I cytokine receptor superfamily and forms a complex signaling structure upon G-CSF binding:

Receptor Structure and Components:

  • The G-CSFR contains a large glycosylated extracellular region with:

    • An N-terminal immunoglobulin-like domain

    • A cytokine receptor homology (CRH) domain containing four conserved cysteine residues and a WSXWS motif

    • Three fibronectin type III domains

Signaling Activation Mechanism:

  • G-CSF binding to the CRH domain induces receptor dimerization with 2:2 stoichiometry (two G-CSF molecules binding to two receptors)

  • Dimerization triggers conformational changes in the receptor

  • This activates intracellular JAK kinases associated with the receptor

  • JAK phosphorylation initiates multiple downstream signaling cascades:

    • JAK/STAT pathway (particularly STAT3)

    • Ras/MAPK pathway

    • PI3K/Akt pathway

Signaling Regulation:

  • Receptor internalization and degradation following activation

  • Negative feedback through SOCS (suppressors of cytokine signaling) proteins

  • Protein tyrosine phosphatases that modulate receptor phosphorylation

Understanding this mechanism is crucial for designing experiments that involve receptor blockade, signaling pathway inhibitors, or development of G-CSF analogs with modified receptor binding properties.

What methodologies are most effective for studying G-CSF receptor binding kinetics?

Several methodologies have proven effective for studying G-CSF receptor binding kinetics, each with distinct advantages:

1. Total Internal Reflection Ellipsometry:

  • Provides real-time binding measurements

  • Allows for determination of association and dissociation rates

  • Can be evaluated using standard Langmuir kinetics models

  • Particularly useful for studying immobilized G-CSF receptors

2. Surface Plasmon Resonance (SPR):

  • Offers label-free detection of binding events

  • Enables measurement of kon and koff rates

  • Requires minimal protein amounts

  • Provides high sensitivity for weak interactions

3. Isothermal Titration Calorimetry (ITC):

  • Measures thermodynamic parameters of binding

  • Determines binding stoichiometry directly

  • Does not require protein labeling or immobilization

  • Provides complete thermodynamic profile (ΔH, ΔS, ΔG)

4. Radioligand Binding Assays:

  • Traditional approach using 125I-labeled G-CSF

  • Provides quantitative Kd and Bmax values

  • Can distinguish high and low-affinity binding sites

  • Useful for cell-based receptor studies

5. Cell-Based Functional Assays:

  • Measures biological responses in G-CSF-responsive cell lines like G-NFS-60

  • Correlates receptor binding with downstream functional outcomes

  • Can detect subtle differences in receptor activation

  • Useful for comparing different G-CSF variants

These methods can be complementary; for example, combining binding studies (SPR) with functional assays provides comprehensive characterization of receptor interactions and their biological consequences.

How can researchers differentiate between G-CSF-mediated effects on different target cell populations?

Differentiating G-CSF effects across various target cell populations requires specialized methodological approaches:

1. Cell-Type Specific Receptor Expression Analysis:

  • Flow cytometry with G-CSFR antibodies to quantify receptor levels

  • RT-qPCR to measure G-CSFR mRNA expression in isolated cell populations

  • Single-cell RNA sequencing to identify receptor distribution patterns

  • Immunohistochemistry for tissue-specific receptor localization

2. Pathway-Specific Activation Monitoring:

  • Phospho-flow cytometry to measure activation of STAT3, MAPK, or Akt in specific cell types

  • Western blotting with phospho-specific antibodies following cell sorting

  • Immunoprecipitation of cell-type specific signaling complexes

  • ChIP-seq for identifying transcriptional targets in different cell types

3. Differential Response Assays:

  • Proliferation assays (for neutrophil precursors and G-NFS-60 cells)

  • Differentiation assays (for hematopoietic progenitors)

  • Survival assays (for neurons and cardiomyocytes)

  • Migration assays (for neutrophils and stem cells)

4. In Vivo Cell Tracking:

  • Adoptive transfer of labeled cell populations before G-CSF treatment

  • In vivo imaging of reporter-expressing cells

  • Lineage tracing in transgenic animal models

  • Flow cytometric analysis of harvested tissues

5. Cell-Type Specific Receptor Knockout Models:

  • Conditional G-CSFR knockout in specific tissues

  • CRISPR-Cas9 editing of the receptor in selected cell populations

  • Chimeric animals with cell-type restricted receptor deficiency

  • Competitive transplantation assays with receptor-modified cells

These approaches enable researchers to delineate the diverse biological effects of G-CSF on various target cells including neutrophil precursors, mature neutrophils, hematopoietic stem cells, neurons, and cardiomyocytes .

What cell-based assays are most reliable for measuring G-CSF biological activity?

Several cell-based assays have been validated for reliable assessment of G-CSF biological activity:

1. NFS-60 Cell Proliferation Assay:

  • Cell Type: Murine myeloblastic cell line dependent on G-CSF

  • Readout: Cell proliferation measured by MTT/XTT or 3H-thymidine incorporation

  • Advantages: High sensitivity, established dose-response relationship

  • Concentration Range: 400 pg/ml–100 ng/ml

  • Incubation Time: 48-72 hours

  • Standardization: Results compared to reference standard curves

2. G-NFS-60 Cell Proliferation:

  • Cell Type: Modified NFS-60 cells with enhanced G-CSF response

  • Methodology: Similar to standard NFS-60 assay but with higher specificity

  • Application: Particularly useful for comparing activity of fusion proteins

  • Controls: Include monomeric G-CSF standards for comparison

3. CD34+ Cell Differentiation Assay:

  • Cell Type: Human CD34+ hematopoietic progenitor cells

  • Readout: Flow cytometric analysis of neutrophil differentiation markers

  • Culture Duration: 7-14 days

  • Analysis: Quantification of CD15+/CD11b+ mature neutrophils

  • Advantage: Directly measures physiologically relevant differentiation

4. HL-60 Differentiation Assay:

  • Cell Type: Human promyelocytic leukemia cells

  • Readout: Morphological changes and neutrophil marker expression

  • Advantage: Accessible cell line that doesn't require primary cells

  • Limitation: Less sensitive than NFS-60 proliferation assay

5. HTRF-Based Detection Systems:

  • Methodology: Homogeneous time-resolved fluorescence detection

  • Application: Quantification of G-CSF in cell supernatants

  • Sample Volume: 16 μL

  • Advantage: High-throughput compatible, suitable for screening applications

  • Analysis: Typically uses 4-parameter logistic curve fitting

When selecting an assay, researchers should consider the specific research question, required sensitivity, and available resources. For regulatory purposes or product development, multiple orthogonal assays are recommended to ensure comprehensive activity characterization.

What are the optimal strategies for purifying His-tagged G-CSF from expression systems?

Purification of His-tagged G-CSF requires strategic approaches tailored to the expression system:

Multi-Step Purification Strategy:

  • Initial Capture: Immobilized Metal Affinity Chromatography (IMAC)

    • Resin: Ni-NTA or Co-NTA agarose

    • Buffer: 20-50 mM sodium phosphate, 300-500 mM NaCl, pH 7.4-8.0

    • Imidazole gradient: 10-250 mM for elution

    • Monitoring: Track protein by UV absorption at 280 nm

  • Intermediate Purification: Ion-Exchange Chromatography

    • Typically cation exchange (G-CSF is basic with pI ~6.1)

    • Buffer: 20-50 mM sodium acetate, pH 4.5-5.5

    • Elution: NaCl gradient (0-1 M)

    • Removes host cell proteins with different charge properties

  • Polishing Step: Size Exclusion Chromatography

    • Column: Superdex 75 or similar

    • Buffer: PBS or 20 mM sodium phosphate, 150 mM NaCl, pH 7.0

    • Flow rate: 0.5-1.0 ml/min

    • Separates monomeric G-CSF from aggregates and fragments

  • Quality Assessment at Each Stage:

    • SDS-PAGE analysis

    • Reverse-phase HPLC

    • Western blot with anti-G-CSF and anti-His antibodies

    • Bradford assay for protein quantification

Expression System-Specific Considerations:

For E. coli Expression:

  • Inclusion body isolation and refolding may be necessary

  • Solubilization using 6-8 M urea or 6 M guanidine hydrochloride

  • Gradual refolding by dialysis against decreasing concentrations of denaturant

  • Addition of oxidizing/reducing agents (e.g., glutathione) to facilitate disulfide bond formation

For Mammalian Cell Expression:

  • Direct capture from cell culture supernatant

  • Filtration through 0.22 μm filter before chromatography

  • Consider adding protease inhibitors to prevent degradation

  • Less refolding required but more complex glycosylation patterns

For Insect Cell Expression:

  • Similar to mammalian cells but with different glycosylation patterns

  • May require additional lectin affinity steps for glycoform separation

Yield Optimization:

  • Typical final yields: 3-5 mg/L for E. coli, 1-3 mg/L for mammalian cells

  • Purity target: >95% as assessed by SDS-PAGE and RP-HPLC

  • Activity retention: Confirm by cell-based assays

This multi-step approach ensures high purity while preserving the biological activity of His-tagged G-CSF.

How can researchers accurately quantify G-CSF levels in experimental samples?

Accurate quantification of G-CSF in experimental samples requires selecting appropriate methods based on the sample type and concentration range:

Immunoassay-Based Methods:

  • ELISA (Enzyme-Linked Immunosorbent Assay):

    • Sensitivity: Typically 5-10 pg/ml

    • Sample Types: Cell culture supernatants, serum, plasma

    • Dynamic Range: 10-2000 pg/ml

    • Analysis: 4-parameter logistic curve fitting recommended

    • Advantages: Widely available commercial kits, well-established protocols

  • HTRF (Homogeneous Time-Resolved Fluorescence):

    • Sample Volume: 16 μL per assay point

    • Application: Rapid quantification in cell supernatants

    • Format Compatibility: Scalable to 1536-well format

    • Procedure: Single-step addition of pre-mixed antibodies

    • Advantages: High-throughput compatible, reduced handling steps

  • Multiplex Immunoassays:

    • Technology: Luminex or MSD platforms

    • Advantage: Simultaneous measurement of G-CSF alongside other cytokines

    • Sample Conservation: Reduced volume requirements for multiple analytes

    • Applications: Complex biological samples where cytokine networks are relevant

Protein Chemistry Methods:

  • Western Blotting:

    • Application: Semi-quantitative analysis with size confirmation

    • Detection: Anti-G-CSF antibodies (clone 5D7 for human G-CSF)

    • Sensitivity: Lower than ELISA (typically ng/ml range)

    • Advantages: Confirms identity by molecular weight, detects modified forms

  • Mass Spectrometry:

    • Approaches: HPLC/ESI-MS for protein identification

    • Advantage: Provides molecular weight confirmation

    • Application: Identification of post-translational modifications

    • Limitation: Requires specialized equipment and expertise

Bioactivity-Based Quantification:

  • Cell Proliferation Assays:

    • Cell Lines: NFS-60 or G-NFS-60 cells

    • Readout: Comparison to standard curves

    • Advantage: Measures functional protein rather than just immunoreactivity

    • Limitation: Influenced by presence of inhibitors or other growth factors

Method Selection Considerations:

  • For cell culture: ELISA or HTRF provide appropriate sensitivity

  • For serum/plasma: Consider enhanced sensitivity ELISA methods

  • For complex samples: Combine immunoassay with bioactivity testing

  • For structural confirmation: Include mass spectrometry analysis

To ensure accurate quantification, include appropriate calibration standards, method validation, and consideration of matrix effects particularly when analyzing complex biological samples.

How do fusion proteins containing G-CSF compare to native G-CSF in research applications?

Fusion proteins containing G-CSF offer distinct advantages and considerations compared to native G-CSF in research applications:

Structural and Functional Comparisons:

CharacteristicNative G-CSFG-CSF Fusion ProteinsResearch Implications
Molecular Weight19.6 kDaVaries (30-70 kDa) depending on fusion partnerAltered pharmacokinetics, tissue penetration
Half-life3.5-4 hoursTypically extended (6-72 hours)Reduced dosing frequency in experimental models
Receptor BindingStandard affinityMay be enhanced or reducedPotentially modified signaling dynamics
Biological ActivitySingle pathway activationCan activate multiple pathways simultaneouslyEnables study of pathway synergies
Tissue DistributionStandardMay be altered by fusion partnerTargeted delivery to specific tissues

Examples of G-CSF Fusion Proteins:

  • SCF-Lα-GCSF and GCSF-Lα-SCF:

    • Composed of human SCF and G-CSF connected by alpha-helix-forming peptide linkers

    • The SCF-Lα-GCSF fusion protein demonstrated biological activity comparable to a mixture of SCF and G-CSF in stimulating neutrophil count increase in rats

    • Provides simultaneous activation of both SCF and G-CSF receptors on target cells

  • PEGylated G-CSF (Pegfilgrastim):

    • PEG moiety extends half-life while maintaining core G-CSF activity

    • Allows for less frequent dosing in experimental models

    • Useful for long-term studies requiring sustained G-CSF activity

  • Fc-G-CSF Fusions:

    • Incorporation of immunoglobulin Fc region extends half-life

    • Enables protein A/G purification methodologies

    • May introduce antibody effector functions in certain applications

Linker Considerations:

  • Alpha-helical linkers (like SGLEA(EAAAK)4ALEA(EAAAK)4ALEGS) maintain activity of both fusion partners

  • Linker length and flexibility significantly impact bioactivity and receptor binding

  • Proper linker design prevents steric hindrance between fusion protein domains

Experimental Advantages:

  • Dual targeting of receptor systems (e.g., SCF-Lα-GCSF activating both G-CSFR and c-Kit)

  • Simplified administration of multiple cytokines

  • Potential for novel biological effects not achieved with individual proteins

  • Customizable pharmacokinetic properties

When designing experiments with G-CSF fusion proteins, researchers should include appropriate controls (individual proteins, mixture of proteins) to distinguish fusion-specific effects from those of the individual components.

What are the current methodological challenges in studying G-CSF effects across different tissue systems?

Research on G-CSF spans multiple tissue systems, presenting several methodological challenges that require specialized approaches:

Cross-System Challenges and Solutions:

  • Variability in Receptor Expression:

    • Challenge: G-CSFR expression varies widely across tissues (high in myeloid precursors, low in neurons)

    • Solution: Quantitative receptor mapping using RT-qPCR, flow cytometry, and immunohistochemistry across tissues

    • Approach: Single-cell RNA sequencing to identify cell-specific receptor expression patterns

  • Dose-Response Relationships:

    • Challenge: Effective concentrations vary by orders of magnitude between tissues

    • Solution: Establish tissue-specific dose-response curves

    • Method: Parallel testing across concentration ranges (400 pg/ml to 100 ng/ml for hematopoietic cells; potentially higher for non-hematopoietic tissues)

  • Pathway Activation Differences:

    • Challenge: G-CSF activates different signaling cascades in different tissues

    • Solution: Comprehensive pathway mapping using phosphoproteomics

    • Technique: Validation with pathway-specific inhibitors to confirm causality

Tissue-Specific Challenges:

  • Neuronal Systems:

    • Challenge: Determining direct vs. indirect effects on neurons

    • Solution: Isolation of neurons from supporting cells in culture

    • Approach: Conditional G-CSFR knockout in specific neural populations

    • Method: Electrophysiological recordings to assess functional neuronal impacts

  • Cardiac Tissue:

    • Challenge: Distinguishing direct cardiomyocyte effects from vascular effects

    • Solution: Primary cardiomyocyte cultures with controlled cell purity

    • Technique: Tissue-specific receptor ablation models

  • Skeletal Muscle:

    • Challenge: Contradictory findings regarding direct G-CSF effects

    • Solution: Time-course studies with appropriate controls

    • Method: Careful experimental design to distinguish media change effects from true G-CSF signaling

Technical Approach Solutions:

  • In Vivo-In Vitro Translation:

    • Challenge: Reconciling in vitro findings with in vivo effects

    • Solution: Organ-specific ex vivo systems (e.g., isolated perfused hearts)

    • Approach: Validation in multiple model systems and species

  • Temporal Dynamics:

    • Challenge: Different tissues respond with different kinetics

    • Solution: Comprehensive time-course studies (minutes to days)

    • Method: Real-time monitoring systems (e.g., reporter cell lines)

  • Model System Selection:

    • Challenge: Choosing appropriate models for specific tissue questions

    • Solution: Match model systems to research questions (cell lines for mechanism, primary cells for physiology, transgenic animals for systemic effects)

    • Approach: Multi-model validation of key findings

These methodological approaches help resolve the conflicting data often observed when studying G-CSF effects across diverse tissue systems, providing a framework for more consistent and translatable research findings.

How can researchers effectively design experiments to study the therapeutic potential of G-CSF in non-hematopoietic applications?

Designing robust experiments to investigate G-CSF's therapeutic potential in non-hematopoietic applications requires systematic approaches:

Experimental Design Framework:

  • Mechanistic Validation Studies:

    • Receptor Expression Verification:

      • Confirm G-CSFR expression in target tissue using RT-qPCR, Western blot, and immunohistochemistry

      • Quantify receptor density compared to hematopoietic controls

      • Verify receptor functionality through phosphorylation assays

    • Signaling Pathway Determination:

      • Map activated pathways (JAK/STAT, MAPK, PI3K/Akt) in target tissues

      • Use pathway inhibitors to confirm causality in observed effects

      • Compare with canonical hematopoietic signaling patterns

  • Dose-Finding Studies:

    • In Vitro Approach:

      • Test broad concentration ranges (400 pg/ml–100 ng/ml as starting point)

      • Establish tissue-specific dose-response curves

      • Determine EC50 values for key therapeutic endpoints

    • In Vivo Dosing:

      • Begin with established hematopoietic doses and adjust based on response

      • Consider pharmacokinetics in target tissue (blood-brain barrier penetration for neurological applications)

      • Use multiple dose groups with appropriate controls

  • Therapeutic Outcome Assessment:

    For Neurological Applications:

    • Measure neuroprotection using cell death assays

    • Assess functional recovery with behavioral testing

    • Quantify neurogenesis and axonal regeneration markers

    For Cardiovascular Applications:

    • Evaluate cardiac function (echocardiography, pressure-volume loops)

    • Measure cardiomyocyte survival after ischemic challenge

    • Assess cardiac remodeling and fibrosis long-term

    For Musculoskeletal Applications:

    • Quantify muscle strength and function

    • Analyze muscle regeneration after injury

    • Measure protein synthesis and degradation rates

  • Control Systems for Disambiguation:

    • Cellular Controls:

      • Compare with G-CSFR knockout/knockdown cells

      • Use receptor blocking antibodies as negative controls

      • Include positive controls (established growth factors for each tissue)

    • Animal Model Controls:

      • Use tissue-specific G-CSFR conditional knockout models

      • Include sham treatment and vehicle controls

      • Compare with established therapeutic interventions

    • Disambiguation Controls:

      • Always include media change controls for in vitro work

      • Distinguish neutrophil-mediated from direct tissue effects using neutropenic models

      • Control for systemic effects using local administration approaches

  • Translational Considerations:

    • Biomarker Development:

      • Identify tissue-specific response biomarkers

      • Develop minimally invasive monitoring approaches

      • Correlate molecular markers with functional outcomes

    • Delivery Optimization:

      • Test targeted delivery approaches (fusion proteins, nanoparticles)

      • Optimize administration route for target tissue access

      • Determine minimum effective dose to reduce neutrophilia side effects

    • Species Translation:

      • Validate findings across multiple species before clinical translation

      • Consider human tissue explants or organoids for final validation

      • Address species differences in receptor distribution and signaling

This comprehensive experimental design framework ensures rigorous evaluation of G-CSF's therapeutic potential beyond its established hematopoietic applications, addressing both mechanistic understanding and therapeutic development.

What are the common technical challenges in G-CSF research and how can they be addressed?

Researchers working with G-CSF frequently encounter several technical challenges that can affect experimental outcomes. Here are the most common issues and their solutions:

Protein Stability and Activity Issues:

  • Challenge: Loss of G-CSF activity during storage and handling

    • Solution: Store lyophilized protein at -80°C; maintain reconstituted protein at 4°C for short-term use

    • Method: Add carrier proteins (0.1% BSA or HSA) to prevent adsorption to containers

    • Verification: Include positive controls from fresh stocks in each experiment

  • Challenge: Batch-to-batch variation in commercial G-CSF preparations

    • Solution: Standardize using bioactivity assays rather than protein concentration alone

    • Method: Establish internal reference standards and perform parallel testing of new batches

    • Verification: Determine EC50 values in standard cell lines (NFS-60) for normalization

Experimental Design Issues:

  • Challenge: Distinguishing direct G-CSF effects from indirect effects via neutrophil activation

    • Solution: Use neutrophil-depleted systems or neutropenic animal models

    • Method: In vitro studies with purified cell populations

    • Controls: Include G-CSFR blocking antibodies or receptor knockdown controls

  • Challenge: Media change artifacts in cell culture experiments

    • Solution: Include proper time-matched controls with media changes but without G-CSF

    • Method: Staggered experimental design with synchronized media changes

    • Verification: Time course studies to distinguish immediate vs. delayed responses

Analytical Challenges:

  • Challenge: Detecting low-level G-CSFR expression in non-hematopoietic tissues

    • Solution: Use high-sensitivity detection methods (nested PCR, RNAscope, highly validated antibodies)

    • Method: Tissue enrichment techniques prior to analysis

    • Controls: Include positive (neutrophil precursors) and negative control tissues

  • Challenge: Differentiating G-CSF isoforms and modified variants

    • Solution: Combine immunological detection with mass spectrometry

    • Method: Use specific antibodies that distinguish native from His-tagged G-CSF

    • Technique: Western blotting with antibodies against both G-CSF and His-tag epitopes

Purification-Specific Issues:

  • Challenge: Co-purification of bacterial endotoxin with recombinant G-CSF

    • Solution: Multiple purification steps including ion exchange and polymyxin B affinity

    • Method: Validate endotoxin removal using LAL testing

    • Threshold: Maintain endotoxin levels below 0.1 EU/μg protein

  • Challenge: Protein aggregation during purification

    • Solution: Include low concentrations of non-ionic detergents (0.005% Tween-20)

    • Method: Size exclusion chromatography as final polishing step

    • Analysis: Monitor aggregation by dynamic light scattering or SE-HPLC

In Vivo Research Challenges:

  • Challenge: Neutrophilia confounding non-hematopoietic effects in animal models

    • Solution: Use neutrophil depletion strategies or G-CSFR tissue-specific knockout models

    • Method: Compare early vs. late effects (before significant neutrophilia occurs)

    • Analysis: Multivariate statistical approaches to control for neutrophil numbers

  • Challenge: Variable tissue penetration of G-CSF in different organ systems

    • Solution: Measure local G-CSF concentrations in target tissues

    • Method: Tissue microdialysis or tissue extraction followed by ELISA

    • Alternative: Use labeled G-CSF to track distribution

These solutions provide practical approaches to common technical challenges, enhancing the reliability and reproducibility of G-CSF research.

How can researchers overcome the challenges in studying G-CSF interactions with multiple receptor systems?

Studying G-CSF interactions across multiple receptor systems presents unique challenges requiring specialized methodological approaches:

Receptor Cross-Talk Analysis:

  • Challenge: Dissecting G-CSFR interactions with other cytokine receptors

    • Solution: Sequential receptor blockade experiments

    • Method: Use receptor-specific blocking antibodies or small molecule inhibitors

    • Analysis: Measure signaling pathway activation with and without blockade

    • Verification: Confirm with receptor knockout/knockdown approaches

  • Challenge: Identifying shared vs. unique signaling components

    • Solution: Phosphoproteomic analysis following receptor stimulation

    • Method: Stimulate cells with G-CSF alone or in combination with other cytokines

    • Analysis: Compare phosphorylation patterns to identify convergent and divergent pathways

    • Validation: Confirm key nodes with pathway-specific inhibitors

Multi-Receptor Expression Systems:

  • Challenge: Creating controlled systems for studying receptor interactions

    • Solution: Generate cell lines with defined receptor expression profiles

    • Method: CRISPR-based editing to create receptor knockout/knockin cells

    • System: Inducible expression systems for titratable receptor levels

    • Validation: Flow cytometry to confirm receptor expression patterns

  • Challenge: Addressing tissue-specific receptor distribution patterns

    • Solution: Single-cell analysis of receptor co-expression

    • Method: Single-cell RNA-seq combined with protein-level validation

    • Analysis: Computational modeling of receptor interaction networks

    • Verification: Spatial transcriptomics to maintain tissue context information

Signaling Pathway Deconvolution:

  • Challenge: Separating overlapping downstream pathways

    • Solution: Time-resolved signaling analysis

    • Method: Collect samples across multiple timepoints after stimulation

    • Analysis: Mathematical modeling to deconvolve temporal signatures

    • Validation: Targeted inhibition of specific pathway components

  • Challenge: Determining functional outcomes of pathway interactions

    • Solution: Genetically encoded biosensors for live-cell imaging

    • Method: FRET-based reporters for key signaling nodes

    • Analysis: Single-cell correlation of signaling intensity with functional outcomes

    • Technique: Combine with transcriptional reporters for pathway-specific gene activation

Experimental Design Strategies:

  • Challenge: Controlling for receptor density effects on signaling

    • Solution: Titration of receptor expression levels

    • Method: Inducible expression systems with dose-responsive control

    • Analysis: Correlate receptor density with signaling intensity

    • Verification: Surface receptor quantification using calibrated flow cytometry

  • Challenge: Differentiating physiological vs. pharmacological effects

    • Solution: Dose-response studies spanning physiological to therapeutic concentrations

    • Method: Parallel in vitro and in vivo concentration measurements

    • Analysis: Identify threshold effects and saturation points

    • Validation: Compare with endogenous G-CSF levels during various physiological states

  • Challenge: Studying receptor dynamics in complex tissues

    • Solution: Intravital imaging of fluorescently labeled receptors

    • Method: Receptor-tagged reporter mice or local virus-mediated expression

    • Analysis: Track receptor clustering, internalization, and recycling

    • Alternative: Ex vivo tissue slice cultures for improved optical access

These approaches provide a comprehensive framework for investigating the complex interactions between G-CSF and multiple receptor systems, enabling more nuanced understanding of its diverse biological effects.

What data analysis approaches are most appropriate for complex G-CSF experimental designs?

G-CSF research often generates complex datasets requiring sophisticated analytical approaches. Here are optimal methods for different experimental scenarios:

Dose-Response and Kinetic Analysis:

  • Nonlinear Regression Models:

    • 4-Parameter Logistic (4PL) regression for dose-response curves

    • Weight by 1/Y² for heteroscedastic data

    • Extract and compare EC50 values with confidence intervals

    • Apply F-test to compare curve parameters between experimental conditions

  • Time-Course Analysis:

    • Area Under the Curve (AUC) calculations for cumulative effects

    • Repeated measures ANOVA with appropriate post-hoc tests

    • Mixed-effects models for handling missing timepoints

    • Time-to-event analysis for threshold crossing

Multi-Parameter Experiments:

  • Multivariate Analysis Techniques:

    • Principal Component Analysis (PCA) to identify major sources of variation

    • Partial Least Squares Discriminant Analysis (PLS-DA) for treatment group separation

    • ANOVA-Simultaneous Component Analysis (ASCA) for multi-factorial designs

    • Implementation in R or Python with packages like mixOmics or scikit-learn

  • Network Analysis Approaches:

    • Correlation networks for receptor-signaling relationships

    • Bayesian network inference for causal relationship modeling

    • Pathway enrichment analysis for transcriptomic/proteomic data

    • Visualization tools like Cytoscape with statistical plugins

Receptor Binding Analysis:

  • Binding Kinetics Models:

    • Standard Langmuir kinetics for simple binding scenarios

    • Two-site binding models for heterogeneous receptor populations

    • Global fitting approaches for simultaneous analysis of multiple datasets

    • Analysis software: GraphPad Prism, Origin, or custom scripts in R/Python

  • Advanced Binding Analysis:

    • Surface Plasmon Resonance (SPR) data analysis with BIAevaluation software

    • Total Internal Reflection Ellipsometry data processing

    • Scatchard plot transformations for detecting binding cooperativity

    • Isothermal Titration Calorimetry (ITC) thermodynamic parameter extraction

Cell-Based Assay Analysis:

  • Proliferation Assay Analysis:

    • Normalization to positive and negative controls

    • Log-transformation of concentration values

    • Relative potency calculations compared to reference standards

    • Statistical comparisons using ANOVA with Dunnett's post-hoc test

  • High-Content Imaging Analysis:

    • Machine learning-based cell classification

    • Single-cell feature extraction and population statistics

    • Hierarchical clustering of phenotypic profiles

    • Tools: CellProfiler, ImageJ/Fiji with appropriate plugins

In Vivo Experimental Analysis:

  • Pharmacokinetic/Pharmacodynamic Modeling:

    • Non-compartmental analysis for basic PK parameters

    • Population PK modeling for inter-individual variability

    • PK/PD linking models for connecting drug levels to neutrophil counts

    • Software: Phoenix WinNonlin, NONMEM, or R packages (nlme)

  • Multi-Endpoint In Vivo Analysis:

    • Mixed-effects models for longitudinal data with nested random effects

    • Survival analysis for time-to-event data

    • Mediation analysis to assess direct vs. indirect effects

    • Meta-analytic approaches for combining multiple studies

Implementation Recommendations:

These analytical approaches enable researchers to extract maximum information from complex G-CSF experiments while maintaining statistical rigor and facilitating reproducibility.

What are the most promising future directions for G-CSF research in multidisciplinary applications?

G-CSF research is expanding beyond traditional hematopoietic applications into several promising multidisciplinary areas:

  • Neurological Applications:

    • Neuroprotection: Building on evidence that G-CSF acts as a neurotrophic factor through receptors expressed in the brain and spinal cord

    • Neuroregeneration: Exploring G-CSF's ability to promote neurogenesis and inhibit apoptosis in neurodegenerative conditions

    • Stroke Recovery: Expanding understanding of G-CSF's mechanisms in improving outcomes after ischemic stroke

    • Future Direction: Development of blood-brain barrier-penetrant G-CSF variants or fusion proteins for enhanced neurological targeting

  • Cardiovascular Applications:

    • Myocardial Protection: Further investigating G-CSF's role in improving cardiac function and cardiomyocyte survival after myocardial infarction

    • Immune Modulation: Exploring how G-CSF promotes T cell immune tolerance and tissue recovery in cardiac ischemia

    • Future Direction: Cardiac-specific delivery systems for G-CSF to maximize local effects while minimizing systemic neutrophilia

  • Tissue Engineering and Regenerative Medicine:

    • Stem Cell Mobilization: Optimizing G-CSF's ability to mobilize various stem cell populations for tissue regeneration

    • Biomaterial Integration: Incorporating G-CSF into scaffolds for enhanced tissue repair

    • Future Direction: Development of fusion proteins that combine G-CSF with tissue-specific growth factors for targeted regeneration

  • Novel Therapeutic Protein Designs:

    • Fusion Protein Development: Building on SCF-Lα-GCSF designs to create multi-functional cytokines with synergistic activities

    • Half-Life Extension: Developing new approaches beyond PEGylation for sustained G-CSF activity

    • Future Direction: Computationally designed protein variants with enhanced receptor specificity or altered signaling bias

  • Precision Medicine Applications:

    • Biomarker Development: Identifying patient-specific markers that predict G-CSF responsiveness

    • Pharmacogenomic Profiling: Understanding genetic variations that impact G-CSF efficacy

    • Future Direction: Development of companion diagnostics for G-CSF therapy selection

  • Advanced Delivery Systems:

    • Controlled Release Formulations: Creating depot formulations for sustained G-CSF delivery

    • Nanoparticle Delivery: Developing targeted nanocarriers for tissue-specific G-CSF deployment

    • Future Direction: Stimuli-responsive systems that release G-CSF in response to specific pathological triggers

  • Systems Biology Approaches:

    • Network Medicine: Mapping G-CSF's position within broader cytokine networks

    • Multi-Omics Integration: Combining transcriptomic, proteomic, and metabolomic data to understand G-CSF effects

    • Future Direction: Computational models predicting optimal G-CSF dosing regimens for individual patients

These emerging research directions represent the most promising frontiers for G-CSF applications, extending well beyond its established role in neutrophil regulation and hematopoietic stem cell mobilization.

How should researchers approach contradictory findings in the G-CSF literature?

When confronting contradictory findings in G-CSF research, a systematic approach helps resolve discrepancies and advance the field:

Methodological Reconciliation Framework:

  • Experimental Design Analysis:

    • Compare Protocol Details: Examine differences in G-CSF source, concentration, duration of exposure, and administration route

    • Cell/Tissue System Evaluation: Consider variations in cell types, culture conditions, or animal models

    • Control Systems Assessment: Evaluate the adequacy of positive and negative controls across studies

    • Resolution Strategy: Conduct side-by-side comparisons using standardized protocols

  • Technical Variable Identification:

    • G-CSF Preparation Differences: Compare recombinant sources, glycosylation status, His-tagged vs. untagged proteins

    • Assay Sensitivity Variations: Assess detection limits and dynamic ranges of analytical methods

    • Sample Processing Disparities: Examine differences in sample handling, storage conditions, or timing

    • Resolution Strategy: Perform inter-laboratory validation studies with harmonized methods

  • Biological Context Considerations:

    • Developmental Stage Differences: Compare results across different developmental stages or cell differentiation states

    • Species-Specific Variations: Assess differences between human, mouse, rat, and other model systems

    • Pathological State Influence: Consider how disease states may alter G-CSF responses

    • Resolution Strategy: Systematic review and meta-analysis stratified by biological variables

Case Example: Muscle Effects of G-CSF

Contradictory findings exist regarding direct effects of G-CSF on skeletal muscle, as noted in the search results :

  • Contradiction: Some studies report direct effects on muscle cells while others fail to replicate these findings

  • Analysis:

    • Dosing differences (400 pg/ml–100 ng/ml vs. higher concentrations)

    • Potential media change artifacts not controlled for in some studies

    • Differences in G-CSFR expression levels across muscle cell models

  • Resolution Approach:

    • Time-matched media change controls

    • G-CSFR expression quantification

    • Pathway-specific inhibitors to confirm direct signaling

Integrative Analytical Approaches:

  • Bayesian Meta-Analysis:

    • Incorporate prior probability distributions based on mechanistic understanding

    • Update with new experimental evidence

    • Quantify uncertainty in effect estimates

    • Identify moderating variables through meta-regression

  • Contradictory Results Workshops:

    • Organize focused meetings where contradictory findings are directly addressed

    • Include representatives from labs with opposing results

    • Develop consensus protocols for standardized testing

    • Establish collaborative validation studies

  • Mechanistic Hypothesis Refinement:

    • Develop testable hypotheses that could explain apparently contradictory results

    • Design critical experiments to distinguish between competing models

    • Consider threshold effects, contextual factors, or non-linear responses

    • Implement systems biology approaches to model complex interactions

Product Science Overview

Introduction

Granulocyte-Colony Stimulating Factor (G-CSF) is a glycoprotein that plays a crucial role in hematopoiesis, the process of forming blood cellular components. It specifically stimulates the bone marrow to produce granulocytes and stem cells and release them into the bloodstream . The recombinant form of G-CSF, tagged with a histidine (His) tag, is widely used in research and clinical applications.

Structure and Production

The human recombinant G-CSF with a His tag is produced in Escherichia coli (E. coli). It is a single, non-glycosylated polypeptide chain containing 174 amino acids, with a molecular mass of approximately 23.19 kDa . The His tag, typically consisting of six histidine residues, is added to the N-terminus of the protein to facilitate purification through affinity chromatography.

Biological Function

G-CSF functions as a cytokine and hormone, promoting the proliferation and differentiation of neutrophils, a type of white blood cell essential for combating infections . It binds to the G-CSF receptor on the surface of hematopoietic cells, triggering a cascade of intracellular signaling pathways that lead to the production and release of neutrophils from the bone marrow .

Clinical Applications

Recombinant human G-CSF is used to manage neutropenia, a condition characterized by an abnormally low count of neutrophils. Neutropenia can result from chemotherapy, radiation therapy, or certain diseases . By increasing neutrophil counts, G-CSF reduces the risk of infections in patients undergoing these treatments .

Research Applications

The His-tagged version of recombinant human G-CSF is particularly valuable in research settings. The His tag allows for easy purification and detection of the protein, facilitating studies on its structure, function, and interactions with other molecules . Researchers use this recombinant protein to investigate the mechanisms of hematopoiesis and to develop new therapeutic strategies for treating neutropenia and other related conditions .

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.